Skip to main content
CNS Neuroscience & Therapeutics logoLink to CNS Neuroscience & Therapeutics
. 2024 Sep 12;30(9):e70039. doi: 10.1111/cns.70039

VSports - Cuproptosis: Mechanisms, biological significance, and advances in disease treatment—A systematic review

Chengliang Pan 1, V体育2025版 - Zhilin Ji 1, Qingxuan Wang 1, Zhao Zhang 1, "VSports app下载" Zhenchuan Wang 1, Chen Li 1, Shan Lu 1, Pengfei Ge (V体育官网) 1,
PMCID: PMC11392831  PMID: 39267265

Abstract

V体育ios版 - Background

Copper is an essential trace element for biological systems, as it plays a critical role in the activity of various enzymes and metabolic processes. However, the dysregulation of copper homeostasis is closely associated with the onset and progression of numerous diseases. In recent years, copper‐induced cell death, a novel form of cellular demise, has garnered significant attention. This process is characterized by the abnormal accumulation of intracellular copper ions, leading to cellular dysfunction and eventual cell death. Copper toxicity occurs through the interaction of copper with acylated enzymes in the tricarboxylic acid (TCA) cycle. This interaction results in subsequent protein aggregation, causing proteotoxic stress and ultimately resulting in cell death. Despite the promise of these findings, the detailed mechanisms and broader implications of cuproptosis remain underexplored. Therefore, our study aimed to investigate the role of copper in cell death and autophagy, focusing on the molecular mechanisms of cuproptosis. We also aimed to discuss recent advancements in copper‐related research across various diseases and tumors, providing insights for future studies and potential therapeutic applications VSports注册入口.

Main Body

This review delves into the biological significance of copper metabolism and the molecular mechanisms underlying copper‐induced cell death. Furthermore, we discuss the role of copper toxicity in the pathogenesis of various diseases, emphasizing recent advancements in the field of oncology. Additionally, we explore the therapeutic potential of targeting copper toxicity V体育官网入口.

Conclusion

The study highlights the need for further research to explore alternative pathways of copper‐induced cell death, detailed mechanisms of cuproptosis, and biomarkers for copper poisoning. Future research should focus on exploring the molecular mechanisms of cuproptosis, developing new therapeutic strategies, and verifying their safety and efficacy in clinical trials VSports在线直播.

Keywords: apoptosis, autophagy, cancer, cell death, copper, cuproptosis, tricarboxylic acid cycle


Copper is an essential trace element in the human body, participating in a variety of biological functions and metabolic processes VSports. The homeostatic copper in the human body is regulated by the absorption, transportation, storage, and metabolism of copper. Dysregulation of copper metabolism is closely related to the occurrence and progression of various diseases. The cell death induced by copper overload is initiated by interaction between copper and acylated enzymes in the tricarboxylic acid (TCA) cycle, which then results in protein aggregation, loss of iron‐sulfur cluster proteins, and cell death. Furthermore, we discuss the role of copper in regulating autophagy, the relationship between copper‐dependent death and disease, and the potential role of copper in cancer treatment. We emphasize the necessity for further research into alternative pathways of copper‐induced cell death, the detailed mechanisms of Cuproptosis, and biomarkers for copper poisoning. It proposes future research directions, including exploring the molecular mechanisms of Cuproptosis, developing new therapeutic strategies, and verifying their safety and efficacy in clinical trials.

graphic file with name CNS-30-e70039-g003.jpg

1. BACKGROUND (VSports)

Copper, an essential trace metal, plays a crucial role in cellular physiology and metabolism. It functions as a cofactor for various enzymes and regulates cellular signal transduction and gene expression. However, the homeostasis of copper ions is vital for cellular function, as abnormal accumulation or deficiency can lead to cellular dysfunction or death. The recent discovery of cuproptosis, a novel form of copper‐dependent cell death, has attracted widespread attention. 1 Cuproptosis involves the binding of copper ions to acylated tricarboxylic acid (TCA) cycle proteins in the mitochondria. This binding triggers protein aggregation and the loss of iron–sulfur cluster proteins, ultimately leading to cell death through a nontraditional apoptotic pathway. The discovery of cuproptosis provides a new perspective on the significance of copper in cellular metabolism and offers new strategies for treating diseases associated with copper metabolism disorders VSports app下载.

The regulation of copper ions has emerged as a novel therapeutic approach in the treatment of disease. 2 Copper chelating agents have shown the potential to inhibit the proliferation of tumor cells by reducing the bioavailability of copper that is available within the cell V体育官网. Conversely, copper ionophores exert an anti‐tumor effect by increasing intracellular copper levels, inducing copper overload and cytotoxicity. The anti‐tumor activity of these copper‐based compounds may be attributed to their impact on copper metabolism within the tumor microenvironment, stimulation of immune responses, and synergistic effects with other chemotherapy drugs.

Additionally, the role of copper in regulating the process of autophagy has attracted the attention of researchers. 3 Autophagy is an intracellular process that involves the degradation and recycling of cellular components, and it plays a crucial role in maintaining intracellular homeostasis. Both an excess and deficiency of copper can affect the process of autophagy, thereby influencing the cell's response to stress and susceptibility to death. Copper regulates autophagy by affecting the AMPK‐MTOR signaling pathway, directly interacting with autophagy‐related proteins or modulating oxidative stress. Copper‐induced autophagy may help protect cells from damage, while in certain cases, copper may promote autophagy‐dependent cell death.

In summary, cuproptosis, an emerging mechanism of cell death, holds significant biological importance and demonstrates great potential in the treatment of diseases. Therefore, by reviewing the homeostasis of copper within biological systems, we aimed to gain an in‐depth understanding of the role of copper in cell death and autophagy, discuss the recent advancements in copper‐related research in various diseases and tumors, and provide insight for future studies and applications of cuproptosis.

2. MAIN TEXT (VSports注册入口)

2.1. Copper homeostasis in the body

2.1.1. Copper physiology in the body

Copper is an essential micronutrient in the human body, crucial for a variety of biological functions. 4 , 5 The absorption, transportation, storage, and metabolism of copper in the human body is a complex and meticulously regulated process. Copper is primarily absorbed in the small intestine, especially in the duodenum and jejunum. 6 The presence of copper in the diet is primarily in the form of inorganic compounds or as part of organic substances. Copper absorption in the small intestine is predominantly carried out through an active transport mechanism that involves specific copper transport proteins such as copper transport protein 1 (CTR1) and ATP7A. 7 These transport proteins use the energy generated from the hydrolysis of ATP within the cell to transport copper ions (Cu2+) from the intestinal lumen into the enterocytes. In addition, some copper can also permeate the cells through passive diffusion. 8

After entering the enterocytes, copper binds to ceruloplasmin, a plasma protein with a high copper‐binding capacity, responsible for transporting copper throughout the body. 9 Ceruloplasmin not only transports copper but also possesses antioxidant properties, capable of eliminating free radicals within the body. 10 In the bloodstream, copper is predominantly circulated in the form of ceruloplasmin, although a portion of it also exists in a free state. Copper is transported to various tissues and organs, particularly the liver, brain, heart, and skeletal muscle.

The liver serves as the primary storage site for copper in the body. Copper is stored in hepatocytes within the liver through the action of copper transport proteins such as ATP7B. Additionally, copper is stored in various tissues, such as the brain and heart. 11 Copper plays a role in the functioning of various enzymes, including oxidases and superoxide dismutases, that are crucial for maintaining normal physiological functions in these tissues.

Copper metabolism involves a complex network of enzymes and proteins in the body. Serving as an essential cofactor for numerous enzymes, copper plays a pivotal role in numerous biological processes, including iron metabolism, neurotransmitter synthesis, antioxidant defense, DNA repair, and energy production. 12 , 13 Copper metabolism also involves the regulation of its redox state, which is highly important for the control of intracellular signal transduction and gene expression. 14 Copper is primarily excretion through bile. In the liver, excess copper is excreted into the intestine via bile, where some of it is removed from the body through feces, while another portion can be reabsorbed again in the intestine, forming a cycle 15 (Figure 1). Moreover, copper can be excreted in small quantities through urine and sweat, although this results in only a minor loss of copper in the human body.

FIGURE 1.

FIGURE 1

The metabolic pathway of copper within cells is a finely regulated process. Cu2+ from outside the cell can enter the cell through passive diffusion. Inside the cell, the reductase FDX1 can reduce Cu2+ to Cu+. Additionally, Cu2+ outside the cell can be reduced to Cu+ by the reductase STEAP and then transported into the cell with the assistance of CTR1. Within the cell, copper chaperones such as ATOX1, CCS, and SOD1 bind to copper ions and transport them to specific organelles to perform their biological functions. In the mitochondria, the protein COX17 transports copper ions from the cytoplasm to the mitochondrial inner membrane, where they are subsequently passed on to mitochondrial membrane proteins (SCO1 and SCO2) as well as COX11, ultimately combining with cytochrome c oxidase (CCO) to participate in the electron transport chain. In the cell's trans‐Golgi network (TGN), ATP7A and ATP7B are two important copper‐transporting ATPases that regulate the excretion of copper to maintain the homeostasis of intracellular copper ions. The coordinated action of these chaperones and transport proteins ensures the rational distribution and functional expression of copper ions within the cell while also preventing cytotoxicity that excess copper ions may cause.

2.1.2. Cellular regulation of copper

Copper, an essential trace element, is necessary for a wide range of physiological processes in nearly all cell types. Since the accumulation of copper within cells can cause oxidative stress and disrupt cellular function, the intake, distribution, and elimination of copper are strictly regulated. Copper homeostasis at the cellular level is coordinated by a series of copper‐dependent proteins, including copper enzymes, copper chaperones, and membrane transport proteins. These proteins work together to coordinate the import, export, and intracellular utilization of copper, thereby maintaining cellular copper levels within a specific range. This regulation helps to prevent the consequences of both copper excess and copper deficiency.

CTR1 is a transporter protein located on the cell membrane, primarily responsible for transporting copper ions (Cu+) from outside the cell to the inside. 16 CTR1 consists of multiple regions containing methionine and histidine, which may endow it with high affinity and selectivity for copper ions and cisplatin, facilitating efficient Cu+ transport from the extracellular environment into the cell. In particular, a hydrophobic region at the N‐terminus, consisting of 60 amino acids, contains several such clusters, including the M2 region. 17 Studies suggest that CTR1 may work in conjunction with some metal reductases (such as STEAP and DCYTB) to transport copper ions across the membrane. 18 , 19 The expression and activity of CTR1 are regulated by various factors, including intracellular copper demand, oxidative stress, and the activation of signaling pathways. For instance, when intracellular copper levels are low, the expression of CTR1 increases to enhance the uptake of copper ions; under oxidative stress conditions, the function of CTR1 may be affected, thereby impacting copper ion homeostasis. 20 Kuo et al. found that mice experiencing copper deficiency exhibit enhanced CTR1 staining in the kidneys, duodenum, and choroid plexus when compared to mice with sufficient copper levels. This finding underscores the specialized role of CTR1 in the transport of copper ions. 21 , 22 Further recent studies indicate that cells respond to increased extracellular copper levels by upregulating the expression of Refrex1 expression, which regulates copper acquisition through interaction with the primary copper transporter CTR1. 23 In yeast, CTR2 serves as a vacuolar copper export protein, capable of mobilizing copper from the vacuolar lumen to the cytoplasm when extracellular copper is scarce. In mammalian cells, CTR2 is believed to play a role in copper homeostasis, but the specific mechanism remains clear. 24 Additionally, studies have shown that CTR2 physically interacts with CTR1 in vivo, potentially forming heterotrimeric or homotrimeric complexes of CTR1 and CTR2. 25 Mice with complete knockout of both alleles of the CTR1 gene exhibit severe growth and developmental defects at mid‐gestation and die in utero, indicating that CTR1 is also crucial for normal embryonic development. 26

In the cytoplasm, the transportation of copper is precisely regulated by a network of copper chaperones with high affinity and synergistic action. Antioxidant‐1 (ATOX1) plays a key role in the transfer of copper ions to ATP7A and ATP7B in the trans‐Golgi network (TGN), facilitating the biosynthesis of copper‐dependent enzymes such as lysyl oxidase, tyrosinase, and plasma copper blue protein 32. 27 ATOX1 is a metallo‐chaperone protein that directly interacts with copper‐transporting ATPases and is crucial for perinatal copper homeostasis. Hamza et al. observed a significant increase in intracellular copper content in ATOX1‐deficient cells due to impaired copper excretion. 28 These findings also reveal that ATOX1 is essential for establishing the threshold of copper‐dependent transport ATPases in the secretory pathway, and in the absence of ATOX1, this transport alone is not sufficient to restore normal copper excretion. 29

SOD1 is an enzyme widely present in cells, responsible for eliminating superoxide and protecting cells from oxidative damage. The activity of SOD1 depends on its post‐translational modification, including the acquisition of copper and zinc, the formation of intramolecular disulfide bonds, and dimerization. 30 CCS is responsible for the copper insertion and disulfide bond formation of SOD1 and regulates the localization of SOD1 between the cytoplasm and mitochondria. The transportation of SOD1 and CCS into the mitochondrial is regulated by the Mia40‐Erv1 disulfide relay system, which is associated with the function of the mitochondrial respiratory chain. 31 Experiments have shown that the concentration of protein level of CCS increases during copper deficiency, and the degradation of CCS accelerates when cells transition from a state of copper deficiency to a state of copper sufficiency. Additionally, CCS may prioritize the utilization of copper by regulating its degradation when there is copper scarcity, which is crucial for maintaining the balance and functioning of intracellular copper. 32

COX17 functions as a copper chaperone protein responsible for transporting copper ions from the cytoplasm to mitochondrial membrane proteins, specifically SCO1 and SCO2. These membrane proteins are responsible for incorporating copper into the mitochondrial‐encoded cytochrome c oxidase II (MT‐CO2/COX2). Additionally, COX17 can facilitate the transportation of copper from the cytoplasm to MT‐CO1/COX1 by delivering it to COX11. 3 , 33 , 34 The active core of the cytochrome oxidase (COX) complex in humans consists of two key subunits, namely COX1 and COX2. These two subunits have a structural correspondence to the conserved CuB and CuA binding sites. These binding sites are specific locations where the subunits bind to copper ions, ensuring their normal function and efficiency in the mitochondrial electron transport chain. 35 Studies have shown that the overexpression of COX17 in cells from patients with SCO2 mutations can partially restore COX activity. In addition, increasing copper concentration can also partially restore COX activity, further supporting the importance of COX17 in copper transfer and COX function. 36 In summary, mutations in the COX17, SCO1, and SCO2 genes can disrupt the function of COX, affecting cellular respiration and potentially leading to the production of excessive reactive oxygen species (ROS), ultimately leading to cell death. 37

Copper‐dependent ATPases, ATP7A and ATP7B, play complementary roles in regulating copper metabolism in the body. They utilize adenosine 5′‐triphosphate (ATP) to transport copper ions, thereby maintaining cellular and systemic copper balance. ATP7A plays a multifaceted function across various tissues, primarily transporting copper ions absorbed from the small intestine to cells throughout the body, except for the liver. In contrast, the primary function of ATP7B is the excretion of copper; it is active in the liver, where it regulates copper homeostasis by eliminating excess copper from the body through bile secretion to maintain a balance in copper metabolism. 38 ATP7A and ATP7B exhibit dynamic movement within the cell, primarily shuttling between the endoplasmic reticulum (ER), the trans‐golgi network (TGN), and the cell membrane. This shuttling is facilitated by the endocytic recycling mechanism involving the retromer complex and other associated factors. 39 The transport activity of ATP7A and ATP7B is dependent on the intracellular copper concentration. Under basal copper levels, these ATPases are located in the TGN; however, when copper concentrations rise, they are translocated to the cell periphery to remove excess copper ions. 40 , 41 The phosphorylation levels of ATP7A and ATP7B correspond to copper concentrations, which affects their intracellular distribution. It is widely accepted that hyperphosphorylation of ATP7A/B triggers their translocation from the TGN to the cell membrane, while dephosphorylation facilitates their retrieval. 42 Recent studies suggest that the expression levels of Cu‐ATPases, particularly ATP7A and ATP7B, may be related to the occurrence and progression of cancer. For example, high expression of ATP7A in invasive breast cancer cells suggests that Cu‐ATPases may be involved in the metastasis process of cancer. 43 Additionally, upregulation of ATP7A expression has been observed in pancreatic cancer cells 44 ; however, whether this can directly impact the occurrence and progression of tumors remains unclear.

2.2. Molecular mechanisms of cuproptosis

2.2.1. The role of copper ions in the TCA cycle

Copper‐dependent death occurs through the direct binding of copper to acylated protein components in the TCA cycle 1 (Figure 2). The TCA cycle is an important metabolic pathway within the cell, where some key enzymes require acylation modification to function. Acylation is a post‐translational modification that involves the covalent attachment of fatty acids or other lipid molecules to proteins, which can be either permanent or reversible. 45 These acylated proteins include succinyl‐CoA transferase, glycine cleavage system protein H, succinyl‐CoA transferase, and succinyl‐CoA acetyltransferase, which regulate the carbon entry points of the TCA cycle. 46 The interaction between copper and the TCA cycle involves the direct influence of copper on acylated proteins. The interaction between copper ions and acylated proteins, especially DLAT, is facilitated by the involvement of FDX1 (ferredoxin 1). FDX1 typically converts Cu2+ to Cu+, resulting in Cu+ binding more tightly and increased toxicity to acylated proteins. 47 The binding of copper to acylated proteins can cause these proteins to aggregate, which may occur through copper bridging the thiol groups in the acylated proteins. The aggregation of acylated proteins may lead to the loss of their function and further induce protein toxicity stress within the cell. 48 In addition, the binding and aggregation of copper with acylated proteins can lead to the loss of iron–sulfur cluster proteins, which can impair mitochondrial function, prevent the cell from maintaining normal metabolic activity, and thus affect cell survival. 49 Copper‐induced aggregation of acylated proteins and the loss of iron–sulfur cluster proteins activate the cellular protein quality control mechanisms, such as the increased expression of heat shock protein 70 (HSP70), ultimately leading to cell death. 50 In summary, cells that depend on mitochondrial respiration or have higher levels of acylated TCA enzymes are more sensitive to copper ionophore‐induced cell death. 51

FIGURE 2.

FIGURE 2

Cuproptosis is a complex process involving various molecular mechanisms. Initially, copper ions are absorbed into the cell through the mediation of the SLC31A1 protein. Subsequently, the ATP7A/B proteins transport copper ions from inside the cell to the outside, maintaining the homeostasis of copper ions. In this process, the FDX1 protein plays a crucial role. FDX1 reduces Cu2+ to Cu+, thereby promoting the process of palmitoylation of the DLAT protein. The binding of copper ions to palmitoylated proteins leads to their aggregation and loss of normal function, ultimately triggering cell death. Additionally, FDX1 is involved in the loss of iron–sulfur cluster proteins, which damages mitochondrial function and further induces cell death. Meanwhile, the binding of copper ions to the CCS protein triggers the succinylation of the SOD1 protein, which weakens the ability of SOD1 to clear reactive oxygen species (ROS). Excessive accumulation of ROS within the cell negatively affects the cell's tricarboxylic acid cycle (TCA cycle), ultimately leading to cell death.

Moreover, related research has found that the binding of copper ionophore protein (CCS1) leads to the succinylation of cysteine residue (Cys146) on SOD1. 52 As a crucial antioxidant enzyme, the succinylation of SOD1 may alter its catalytic function, thereby affecting its ability to eliminate superoxides. Consequently, this could increase superoxide levels, potentially impacting the activity of enzymes involved in redox reactions within the TCA cycle and induce cell death. 53 , 54 However, current research on the specific mechanisms and extent of this impact is relatively limited. Future studies may provide more detailed information on how succinylation of SOD1 regulates the TCA cycle and the implications of this regulation on cellular metabolism and the progression of diseases.

2.2.2. Redox activity of copper ions

The interaction between copper ions and hydrogen peroxide can result in the production of ROS, a process also known as the Fenton reaction. However, the catalytic efficiency of copper ions is generally lower than that of iron ions. 55 , 56 At low concentrations of iron ions, the presence of copper ions can significantly promote the generation of ROS. In the context of copper‐induced cell damage, the production of ROS and its impact is multifaceted; ROS can directly damage cellular components and also indirectly lead to cell damage by affecting cellular signaling and gene expression. 57 , 58 Furthermore, recent studies have found that copper ions can enhance oxidative stress and endoplasmic reticulum stress responses. 59 This can exacerbate zinc ion‐induced neuronal cell death by activating the SAPK/JNK signaling pathway, 60 which may be one of the key mechanisms underlying cuproptosis.

2.2.3. Copper ions and DNA damage

The programmed cell death induced by copper may also be associated with its interaction with DNA. Research by Anchordoquy et al. reported that culturing bovine cumulus cells in an environment with high concentrations of copper significantly increased their genetic damage index (GDI), and an increase in DNA damage was observed across all tested concentrations of copper. 61 These findings suggest that copper may induce the death of bovine cumulus cells by producing genotoxic effects. Additionally, other studies have found that nanoparticles of CuO exhibit stronger genotoxic activity than microparticles of CuO when administered at the same dosage. 62 , 63 This is attributed to nanoparticles' higher cellular uptake, leading to an increased level of copper in the cells. Although the induction of cell death by copper through the initiation of DNA damage has been demonstrated in numerous experiments, 64 , 65 , 66 it remains unclear whether copper can directly interact with DNA. Current research indicates that redox‐active metal ions interact with the bases found in DNA molecules, which may lead to DNA strand breaks and alterations in the chemical structure of the bases. 67 Therefore, the presence of copper ions may promote abnormal base pairing through oxidative modification of the bases, resulting in base substitution mutations. Understanding the mechanism by which copper ions induce base substitution mutations is of significant importance and may represent a unique direction for research into cuproptosis.

2.2.4. Cuproptosis and apoptosis

Apoptosis is a programmed cell death process mediated by both intrinsic and extrinsic signals. 68 The intrinsic pathway, also known as the mitochondrial pathway, primarily responds to internal cellular stress signals, such as DNA damage or the absence of growth factors, which can activate a series of protein kinases, including the p53 protein. 69 The activation of the p53 protein can further promote the expression of proteins, such as Bax and Bak, which play a key role in regulating mitochondrial membrane permeability. 70 The activation of Bax and Bak leads to increased mitochondrial outer membrane permeability, facilitating the release of cytochrome c. 71 Cytochrome c, once released, binds with apoptotic protease activating factor 1 (APAF‐1) and the precursor of caspase‐9 (procaspase‐9) to form the apoptosome. 72 The apoptosome further activates caspase‐9, which subsequently activates downstream effector caspases, such as caspase‐3, ‐6, and ‐7. 73 These effector caspases are responsible for executing the programmed cell death.

Cuproptosis is a newly discovered form of cell death that involves the interaction of copper ions with proteins associated with the mitochondrial respiratory chain. However, it has been noted that simply inhibiting apoptosis is insufficient to prevent elesclomol‐induced cell death. Conversely, the use of copper chelators to treat cell death caused by an excess of Cu2+ can demonstrate a significant protective effect. This indicates that the mechanism of cell death caused by copper toxicity is inherently different from that of apoptosis. 1 In addition, copper‐induced neurodegeneration and oxidative damage can promote the expression of apoptosis‐related proteins Bax, copper‐induced cell death‐related proteins FDX1 and DLAT, and the protein toxicity stress marker HSP70, while reducing the expression of Fe‐S cluster proteins. 74 Further research has shown that an excess Cu can lead to the accumulation of ROS within cells and a significant increase in levels of SOD and LPO. Concurrently, this state of copper excess can upregulate the mRNA and protein expression levels of Bak1, Bax, CytC, and Caspase3, while suppressing the mRNA and protein expression of Bcl2, exhibiting a clear dose‐dependent effect, thereby inducing apoptosis. 75 These findings reveal a potential intrinsic connection between copper‐induced cell death and apoptosis. Although the two processes differ in molecular mechanisms, they may influence each other in the regulation of programmed cell death.

2.2.5. Copper and autophagy

Autophagy is an intracellular process characterized by the formation of double‐membrane structures that facilitate self‐digestion. 76 It plays a crucial role in clearing damaged or misfolded proteins that could potentially lead to disease, thereby maintaining cellular homeostasis and health. 77 The initiation of autophagy is regulated by multiple signaling pathways, including the AMPK‐mTOR pathway. The mTOR kinase serves as a pivotal regulator of autophagy; its activation suppresses autophagy, while its inhibition promotes it. AMPK enhances autophagy by phosphorylating the mTOR complex 1 (mTORC1), 78 whereas active mTORC1 inhibits autophagy by binding to and phosphorylating the ULK1/2 (unc‐51 like autophagy activating kinase 1/2) and ATG13 (autophagy‐related 13) protein complex. 79 The ULK1 complex is an essential positive regulator for autophagosome formation, which recruits the PI3K complex to function collaboratively in this process. 80 The fusion of autophagosomes with lysosomes requires the participation of LAMP‐1 and Rab7 proteins, followed by the degradation of the contents within the autophagosomes. 81 Copper, through its redox activity, can induce autophagy by generating ROS via the Fenton reaction. Excessive ROS can hyperactivate the autophagic mechanism, providing positive feedback that accelerates cell death by selectively degrading damage‐resistant or antioxidant proteins. 82 It has been reported that copper can modulate the AMPK‐MTOR pathway by increasing ATG expression, inducing oxidative stress through the mitochondrial pathway, thereby triggering autophagy. 83 The knockout of the cellular ATP7B leads to a significant increase in the expression of autophagy genes. This occurs as reduced mTOR activity results in the translocation of the mTOR substrate transcription factor EB to the nucleus, which then triggers the activation of autophagy‐related genes. 84 Copper can also induce autophagic flux by directly binding and activating the ULK1/2 in tumor cells. In addition, the absence of Ctr1 or mutations in ULK1 results in reduced Cu binding and impaired formation of the autophagosome complex. 85 Recent reports indicate that copper promotes the autophagic degradation of GPX4 protein, mediated by TAX1BP1, which can facilitate ferroptosis in pancreatic cancer cells. 86 However, the role of copper in cellular autophagy is dualistic; the use of copper chelators or the knockout of SLC31A1 increases autophagy in pancreatic cancer cells. 87 Further in‐depth research into the mechanisms by which copper affects cellular autophagy and the evaluation of its clinical application prospects are necessary. Such research will be instrumental in providing insights for future cancer therapies.

2.3. Copper‐dependent death and disease

Imbalances in copper metabolism can lead to various adverse effects on human health. These harmful effects can result from disturbances in copper absorption, reduced copper excretion, and abnormal accumulation of copper in different vital organs. Additionally, studies have shown that dysregulation of copper homeostasis is closely related to the occurrence and progression of certain cancers.

2.3.1. Diseases caused by copper deficiency

Menkes disease (MD), a representative of copper metabolism disorders, is an X‐linked lethal neurodegenerative condition caused by mutations in the ATP7A gene, which encodes a copper‐transporting P1B‐type ATPase. 88 Mutations in the ATP7A gene lead to impaired function of the ATP7A protein, preventing the transport of copper from intestinal cells to other vital organs, and resulting in systemic copper deficiency. 89 Consequently, copper cannot be effectively delivered to copper‐dependent enzymes, such as cytochrome c oxidase and superoxide dismutase, leading to a decrease in their activity and adverse effects on key physiological processes such as cellular respiration, neurotransmitter synthesis, free radical scavenging, and melanin production. 90 , 91 MD is a highly lethal disease, with typical clinical manifestations in the neonatal period, including hypothermia, hypoglycemia, malnutrition, and occasionally hemorrhagic damage, such as cephalohematoma. 92 , 93 Older infants often exhibit seizures and neurodegenerative changes, likely due to reduced activity of dopamine‐β‐hydroxylase (DβH), 90 impaired mitochondrial function from reduced COX activity, and cerebral lactic acidosis caused by mitochondrial dysfunction. In addition to the clinical manifestations caused by copper deficiency in the brain tissue, connective tissue dysfunction is also a classic feature of MD, most notably characterized by colorless and brittle hair. 93 This is likely due to the weakened function of ATP7A, inhibiting the activity of lipoxygenase (LOX) in the skin, muscles, and connective tissues, thereby hindering the normal oxidation of lysine and hydroxylysine within elastin and collagen. 94 Reduced activity of LOX, decreases the formation of covalent cross‐links, which is essential for maintaining the tensile strength and elasticity of connective tissues in the skeleton, muscles, and cardiovascular system. 95 , 96

Genetic screening is usually the most effective diagnostic method for MD, 97 and the analysis of plasma catecholamine levels can also accurately diagnose neonatal MD. 98 Because the essence of the disease is the inability to transport copper from intestinal cells to other tissues and organs, direct copper supplementation may be an effective treatment method for MD. Currently, the use of copper salts (such as copper‐histidine complex) that can be directly transported through the bloodstream is the primary clinical treatment for MD. 99 This treatment is effective in patients with ATP7A mutations that still allow copper to cross the blood–brain barrier. 100 In addition, patients diagnosed prenatally or shortly after birth and receiving early treatment have longer survival time and better prognoses, 101 which suggests that copper is crucial for the development of the human nervous and motor systems. Interestingly, patients with asymptomatic MD respond better to copper‐histidine complex treatment compared to those with typical symptoms, 102 which may be related to the duration and extent of their copper deficiency.

In addition to the classic MD, studies have found that copper deficiency may also be associated with anemia, specifically copper‐deficiency anemia. Various causes can lead to copper deficiency in the human body, such as zinc‐induced copper deficiency, copper loss due to surgery, copper deficiency in parenteral or enteral nutrition owing to celiac disease, etc. 103 , 104 , 105 Copper deficiency can impair the activity of iron oxidases and disrupt the transportation of iron from cells to plasma, thereby affecting hemoglobin synthesis. 106 , 107 However, whether copper deficiency directly impacts the hematopoietic function of the bone marrow is currently unknown. Measuring serum copper, serum ceruloplasmin, or 24‐hour urinary copper levels in conjunction with bone marrow biopsy is important for diagnosing copper‐deficiency anemia. 108 Additionally, there are case reports suggesting that copper deficiency may cause spinal cord lesions. 109 Although the importance of copper to the nervous system has been confirmed, the pathogenesis of copper‐deficiency myelopathy remains to be explored.

2.3.2. Diseases caused by copper overload

Just as abnormal copper accumulation within cells can induce cell death, copper overload in the human body can also lead to the onset of related diseases, the most notable being Wilson's disease (WD). WD is an autosomal recessive genetic disorder caused by mutations in the ATP7B gene, which encodes a transmembrane copper‐transporting ATPase primarily responsible for transporting copper out of cells. 110 The loss of ATP7B function leads to abnormal accumulation of copper in vital organs and tissues in the human body, such as the liver and brain, thereby causing a range of clinical symptoms. 111

Copper is primarily excreted from the body through bile, making the liver the main regulatory organ for copper homeostasis. Consequently, liver pathology is often the first and most common manifestation of WD. The hallmark of this condition is an abnormally elevated copper concentration in hepatocytes of patients with WD, which initially bind to metallothionein in the cell cytoplasm. As copper accumulation intensifies, copper ions are released from their bound state with metallothionein and may transfer to other parts of the cell, especially lysosomes. 112 Within lysosomes, excessive copper can disrupt their normal degradation function and damage cellular structures, including proteins, lipids, DNA, and RNA, through the generation of oxidative stress. 113

In addition, the accumulation of copper can trigger apoptosis pathways, leading to hepatocyte death, through the activation of acid sphingomyelinase and other pathways. 114 Long‐term copper accumulation and cell death can lead to a series of pathological changes in the liver, such as fatty liver, hepatitis, and hepatic fibrosis, potentially progressing to cirrhosis. Mutations in the ATP7B gene disrupt the transport function of ATP7B and impair the synthesis of ceruloplasmin, an important copper‐binding protein. 115 As hepatocytes perish and plasma ceruloplasmin levels decline, the concentration of free copper ions in the bloodstream increases, facilitating the transportation of copper to various tissues and organs via the blood. Once copper accumulation reaches a critical threshold, it induces clinical symptoms, with brain dysfunction being the most significant, followed by effects on the eyes, heart, bones, and other organs. 111 , 116

Moreover, relevant studies have confirmed a positive correlation between copper concentration in brain tissue cells and neurological damage. 117 Astrocytes, a type of glial cell in the central nervous system, play multiple important roles in the brain. These roles include supporting and protecting neurons, maintaining the stability of the extracellular environment, forming the blood–brain barrier, and participating in the repair process of the nervous system. 118 Astrocytes can take up excess copper, upregulate metallothionein levels, and mitigate copper's neurotoxicity by increasing cell number and volume. 117 However, long‐term exposure to a high copper concentration may damage and eventually kill astrocytes, further impairing the blood–brain barrier and causing damage to neurons and other brain tissues. 119 The clinical manifestations of brain damage in patients with WD vary according to the affected cerebral region, with the most common and severe manifestation being damage to the putamen, clinically presenting as dystonia and Parkinson's disease. 120

Another important diagnostic feature observed in patients with WD is the Kayser–Fleischer ring, resulting from abnormal copper accumulation in the eyes, which is especially common among patients with neurological damage. 121 While measuring serum ceruloplasmin levels and nonceruloplasmin‐bound copper levels can serve as an effective method for diagnosing WD, they often lack specificity. 122 , 123 , 124 In addition, clinical manifestations and imaging examinations serve as supplementary diagnostic tools for WD, while DNA analysis of ATP7B gene mutations provides genetic confirmation. WD is a systemic disease caused by mutations in the ATP7B gene, which lead to abnormal copper accumulation. Treatment primarily involves regulating copper intake and enhancing copper excretion, which can be maintained for life. 121 , 125 Moreover, liver transplantation is considered an effective treatment for WD due to its primary manifestation as liver damage; however, there are still several limitations in its therapeutic effects. 126 Furthermore, owing to the role of astrocytes in copper metabolism and neuroprotection, they could be potential targets for WD treatment strategies. These strategies may involve enhancing their copper buffering capacity or antioxidant defenses to reduce the toxicity of copper.

2.3.3. Copper and cancer

Copper, an essential trace element for the human body, exhibits a dual effect on cells due to its redox properties. Copper influences cellular signal transduction by activating or inhibiting specific copper‐dependent enzymes. For instance, in the RAF–MEK–ERK signaling pathway, copper can enhance the kinase activity of MEK1 and MEK2 through metal isoform regulation, leading to the phosphorylation of ERK1 and ERK2, thereby regulating the expression of genes associated with cell proliferation. 127 Several reports indicate that the plasma and cellular copper concentrations in patients with cancer are higher than normal levels. 128 , 129 , 130 , 131 , 132 , 133 , 134 Simultaneously, copper can directly activate various pro‐angiogenic factors (such as VEGF, FGF2, etc.) to promote new blood vessel formation, a key step in tumor growth and metastasis. 135 , 136 The role of copper in cell biology extends beyond maintaining basic metabolic processes; it also participates in more complex cellular behaviors. For example, it acts on the ATOX‐ATP7A‐LOX signaling pathway, where ATOX1, as a cytosolic copper chaperone, transfer copper to ATP7A, thereby regulating the intracellular distribution and export. The LOX family of enzymes uses copper as a cofactor to catalyze the cross‐linking of collagen and elastin, affecting the structure and function of the extracellular matrix, which can promote the invasive expansion of tumors. 137 This demonstrates the relationship between copper and the onset and development of cancer, with tumor tissues requiring more copper compared to healthy tissues. 138 Considering copper's role in cancer development, it emerges as a potential target for therapeutic intervention. Current potential cancer treatment strategies primarily involve two approaches: using chelating agents to counteract excess copper and increasing copper concentration to mediate programmed cell death in tumor cells. 139 Copper chelating agents such as triethylenethiophosphoramide (TTM) or D‐penicillamine can form stable complexes with copper ions in the body, thereby reducing the concentration of available copper ions within cells and inhibiting the activity of angiogenic factors. 140 , 141 Protein aggregation induced by copper ions and the loss of Fe‐S cluster proteins trigger a protein toxicity stress response, activating the cell's unfolded protein response (UPR) and autophagy process. However, when these mechanisms fail to clear misfolded proteins, they lead to cell death. 142

Copper ionophores (such as elesclomol, DSF, etc.) primarily enhance intracellular copper levels, resulting in mitochondrial dysfunction and the production of ROS. These combined effects ultimately promote copper‐induced death, contributing to their anti‐tumor effects. 143 , 144 , 145 Several studies have found that the concurrent administration of DSF and copper treatment can downregulate the expression of tumor suppressor gene PTEN and activate the AKT signaling pathway. 145 This lays a theoretical basis for future research into the synergistic use of copper ionophores with PI3K‐AKT pathway inhibitors. In addition, due to DSF's effective penetration of blood–brain barrier, relevant studies have confirmed that its combination treatment with temozolomide can significantly extend the survival period of patients with specific types of cancer. 146

"V体育2025版" 3. CONCLUSION

Copper, as one of the essential trace elements for the human body, plays an irreplaceable role in both the macro and micro regulatory processes. It serves as an indispensable cofactor for certain key enzymes, and a deficiency in copper can lead to a series of adverse effects on the organism. However, an excess of copper can induce oxidative stress and trigger cell death through its action on the mitochondrial respiratory chain, a process known as cuproptosis. As a novel form of cell death, cuproptosis provides a new perspective on the role of copper ions in cellular respiration metabolism and diseases. While the acylation of proteins in the mitochondrial respiratory chain and the lipoic acid pathway have been identified as crucial in cuproptosis, exploring alternative pathways through which copper may induce cell death is a significant research avenue. In addition, there are no detailed reports on how the acylated mitochondrial respiratory chain proteins trigger cell death through specific signaling pathways. Therefore, further research is needed to clarify the exact mechanism of cuproptosis and to address some intractable diseases in clinical practice through this process. Incidentally, although the biomarkers for copper poisoning are not yet clear, identifying their specific markers is of great significance for promoting the clinical diagnosis and treatment of copper poisoning under human pathological conditions, which will be a key direction for future research.

Programmed cell death induced by metal ions has gradually gained significant attention in recent years. One example is ferroptosis, a form of cell death induced by iron ions causing lipid peroxidation of the cell membrane, leading to an excessive accumulation of ROS and ultimately triggering programmed necrosis. Interestingly, mitochondria have emerged as a key factor in both processes, as they are responsible for cell deaths induced by metal ions. This suggests the potential for interaction between the two. Recent studies have confirmed that mitochondrial glutathione (GSH) can effectively downregulate Cu‐induced cell death by inhibiting enzyme acylation and promoting DLAT oligomerization. Additionally, in the management of ferroptosis, the use of MitoQ as a ROS scavenger can increase GSH levels and maintain the integrity of mitochondrial. During ferroptosis, mitochondria undergo a series of morphological changes; however, it remains unclear whether mitochondria undergo similar morphological changes in mitochondria during cuproptosis. Exploring the relationship between iron and copper, the two metal ions in cell death may emerge as a promising research direction.

By analyzing the various diseases resulting from the disturbance of copper homeostasis in the body, a significant correlation between these diseases and cuproptosis can be identified. Therefore, while investigating cuproptosis, additional therapeutic targets for diseases characterized by copper ion disorders can be uncovered. To better understand the dynamic process through which copper toxicity triggers and accelerates the advancement of diseases, further evidence is required. In the future, prospective studies will be particularly crucial in conditions such as WD, and neurodegenerative disorders, notably in oncology, where the induction and regulation of cuproptosis could yield great therapeutic potential. This will involve exploring different copper ion carriers and the precise regulation of copper ion levels across various tissues.

In summary, the field of cuproptosis research is rapidly expanding. Therefore, future research should explore the molecular mechanisms of cuproptosis, develop new therapeutic strategies, and verify their safety and efficacy in clinical trials.

AUTHOR CONTRIBUTIONS

Chengliang Pan was a major contributor in writing the manuscript. Pengfei Ge guided the direction of the writing for this article. Other authors assisted in completing the literature search and organization‐related tasks. All authors read and approved the final manuscript.

FUNDING INFORMATION

This work is funded by the National Natural Science Foundation of China (82372690, 82173027).

CONFLICT OF INTEREST STATEMENT

The authors declare that they have no competing interests.

Pan C, Ji Z, Wang Q, et al. Cuproptosis: Mechanisms, biological significance, and advances in disease treatment—A systematic review. CNS Neurosci Ther. 2024;30:e70039. doi: 10.1111/cns.70039

DATA AVAILABILITY STATEMENT

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

REFERENCES

  • 1. Tsvetkov P, Coy S, Petrova B, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science (New York, NY). 2022;375(6586):1254‐1261. doi: 10.1126/science.abf0529 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Chen L, Min J, Wang F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct Target Ther. 2022;7(1):378. doi: 10.1038/s41392-022-01229-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Xue Q, Kang R, Klionsky DJ, Tang D, Liu J, Chen X. Copper metabolism in cell death and autophagy. Autophagy. 2023;19(8):2175‐2195. doi: 10.1080/15548627.2023.2200554 [VSports手机版 - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Maung MT, Carlson A, Olea‐Flores M, et al. The molecular and cellular basis of copper dysregulation and its relationship with human pathologies. FASEB J. 2021;35(9):e21810. doi: 10.1096/fj.202100273RR [V体育官网入口 - DOI] [PubMed] [Google Scholar]
  • 5. Festa RA, Thiele DJ. Copper: an essential metal in biology. Curr Biol. 2011;21(21):R877‐R883. doi: 10.1016/j.cub.2011.09.040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Doguer C, Ha JH, Collins JF. Intersection of iron and copper metabolism in the mammalian intestine and liver. Compr Physiol. 2018;8(4):1433‐1461. doi: 10.1002/cphy.c170045 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Chen J, Jiang Y, Shi H, Peng Y, Fan X, Li C. The molecular mechanisms of copper metabolism and its roles in human diseases. Pflugers Arch Eur J Physiol. 2020;472(10):1415‐1429. doi: 10.1007/s00424-020-02412-2 [DOI] [PubMed] [Google Scholar]
  • 8. Schoeberl A, Gutmann M, Theiner S, et al. The copper transporter CTR1 and cisplatin accumulation at the single‐cell level by LA‐ICP‐TOFMS. Front Mol Biosci. 2022;9:1055356. doi: 10.3389/fmolb.2022.1055356 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Pierson H, Yang H, Lutsenko S. Copper transport and disease: what can we learn from organoids? Annu Rev Nutr. 2019;39:75‐94. doi: 10.1146/annurev-nutr-082018-124242 [V体育安卓版 - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Siddiqi UR, Begum S, Shahjadi S, et al. Plasma zinc, copper and serum ceruloplasmin levels of autism spectrum disorder children in Bangladesh. Heliyon. 2023;9(8):e18624. doi: 10.1016/j.heliyon.2023.e18624 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Schushan M, Bhattacharjee A, Ben‐Tal N, Lutsenko S. A structural model of the copper ATPase ATP7B to facilitate analysis of Wilson disease‐causing mutations and studies of the transport mechanism. Metallomics. 2012;4(7):669‐678. doi: 10.1039/c2mt20025b ["VSports最新版本" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Lutsenko S. Copper trafficking to the secretory pathway. Metallomics. 2016;8(9):840‐852. doi: 10.1039/c6mt00176a [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Lutsenko S, Bhattacharjee A, Hubbard AL. Copper handling machinery of the brain. Metallomics. 2010;2(9):596‐608. doi: 10.1039/c0mt00006j [DOI] [PubMed] [Google Scholar]
  • 14. Lutsenko S, LeShane ES, Shinde U. Biochemical basis of regulation of human copper‐transporting ATPases. Arch Biochem Biophys. 2007;463(2):134‐148. doi: 10.1016/j.abb.2007.04.013 ["VSports" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Aggett PJ. An overview of the metabolism of copper. Eur J Med Res. 1999;4(6):214‐216. [VSports app下载 - PubMed] [Google Scholar]
  • 16. Larson CA, Adams PL, Jandial DD, Blair BG, Safaei R, Howell SB. The role of the N‐terminus of mammalian copper transporter 1 in the cellular accumulation of cisplatin. Biochem Pharmacol. 2010;80(4):448‐454. doi: 10.1016/j.bcp.2010.04.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Das A, Ash D, Fouda AY, et al. Cysteine oxidation of copper transporter CTR1 drives VEGFR2 signalling and angiogenesis. Nat Cell Biol. 2022;24(1):35‐50. doi: 10.1038/s41556-021-00822-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Ohgami RS, Campagna DR, McDonald A, Fleming MD. The Steap proteins are metalloreductases. Blood. 2006;108(4):1388‐1394. doi: 10.1182/blood-2006-02-003681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. McKie AT, Barrow D, Latunde‐Dada GO, et al. An Iron‐Regulated Ferric Reductase Associated with the Absorption of Dietary Iron. Science (New York, NY). 2001;291(5509):1755‐1759. doi: 10.1126/science.1057206 [DOI] [PubMed] [Google Scholar]
  • 20. Liang ZD, Tsai WB, Lee MY, Savaraj N, Kuo MT. Specificity protein 1 (sp1) oscillation is involved in copper homeostasis maintenance by regulating human high‐affinity copper transporter 1 expression. Mol Pharmacol. 2012;81(3):455‐464. doi: 10.1124/mol.111.076422 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Kuo YM, Gybina AA, Pyatskowit JW, Gitschier J, Prohaska JR. Copper transport protein (Ctr1) levels in mice are tissue specific and dependent on copper status. J Nutr. 2006;136(1):21‐26. doi: 10.1093/jn/136.1.21 ["V体育ios版" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Nose Y, Kim BE, Thiele DJ. Ctr1 drives intestinal copper absorption and is essential for growth, iron metabolism, and neonatal cardiac function. Cell Metab. 2006;4(3):235‐244. doi: 10.1016/j.cmet.2006.08.009 ["VSports在线直播" DOI] [PubMed] [Google Scholar]
  • 23. Tury S, Chauveau L, Lecante A, Courgnaud V, Battini JL. A co‐opted endogenous retroviral envelope promotes cell survival by controlling CTR1‐mediated copper transport and homeostasis. Cell Rep. 2023;42(9):113065. doi: 10.1016/j.celrep.2023.113065 [DOI] [PubMed] [Google Scholar]
  • 24. Öhrvik H, Thiele DJ. The role of Ctr1 and Ctr2 in mammalian copper homeostasis and platinum‐based chemotherapy. J Trace Elem Med Biol. 2015;31:178‐182. doi: 10.1016/j.jtemb.2014.03.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Öhrvik H, Nose Y, Wood LK, et al. Ctr2 regulates biogenesis of a cleaved form of mammalian Ctr1 metal transporter lacking the copper‐ and cisplatin‐binding ecto‐domain. Proc Natl Acad Sci USA. 2013;110(46):E4279‐E4288. doi: 10.1073/pnas.1311749110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Lee J, Petris MJ, Thiele DJ. Characterization of mouse embryonic cells deficient in the ctr1 high affinity copper transporter. Identification of a Ctr1‐independent copper transport system. J Biol Chem. 2002;277(43):40253‐40259. doi: 10.1074/jbc.M208002200 [DOI] [PubMed] [Google Scholar]
  • 27. Lutsenko S, Barnes NL, Bartee MY, Dmitriev OY. Function and regulation of human copper‐transporting ATPases. Physiol Rev. 2007;87(3):1011‐1046. doi: 10.1152/physrev.00004.2006 [DOI] [PubMed] [Google Scholar]
  • 28. Hamza I, Prohaska J, Gitlin JD. Essential role for Atox1 in the copper‐mediated intracellular trafficking of the Menkes ATPase. Proc Natl Acad Sci USA. 2003;100(3):1215‐1220. doi: 10.1073/pnas.0336230100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Itoh S, Kim HW, Nakagawa O, et al. Novel role of antioxidant‐1 (Atox1) as a copper‐dependent transcription factor involved in cell proliferation. J Biol Chem. 2008;283(14):9157‐9167. doi: 10.1074/jbc.M709463200 [DOI (VSports在线直播)] [PMC free article] [PubMed] [Google Scholar]
  • 30. Kawamata H, Manfredi G. Import, maturation, and function of SOD1 and its copper chaperone CCS in the mitochondrial intermembrane space. Antioxid Redox Signal. 2010;13(9):1375‐1384. doi: 10.1089/ars.2010.3212 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Tafuri F, Ronchi D, Magri F, Comi GP, Corti S. SOD1 misplacing and mitochondrial dysfunction in amyotrophic lateral sclerosis pathogenesis. Front Cell Neurosci. 2015;9:336. doi: 10.3389/fncel.2015.00336 ["VSports" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Bertinato J, L'Abbé MR. Copper modulates the degradation of copper chaperone for Cu,Zn superoxide dismutase by the 26 S proteosome. J Biol Chem. 2003;278(37):35071‐35078. doi: 10.1074/jbc.M302242200 [DOI] [PubMed] [Google Scholar]
  • 33. Prohaska JR. Role of copper transporters in copper homeostasis. Am J Clin Nutr. 2008;88(3):826S‐829S. doi: 10.1093/ajcn/88.3.826S [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Horng YC, Cobine PA, Maxfield AB, Carr HS, Winge DR. Specific copper transfer from the Cox17 metallochaperone to both Sco1 and Cox11 in the assembly of yeast cytochrome C oxidase. J Biol Chem. 2004;279(34):35334‐35340. doi: 10.1074/jbc.M404747200 [DOI] [PubMed] [Google Scholar]
  • 35. Nývltová E, Dietz JV, Seravalli J, Khalimonchuk O, Barrientos A. Coordination of metal center biogenesis in human cytochrome c oxidase. Nat Commun. 2022;13(1):3615. doi: 10.1038/s41467-022-31413-1 ["VSports app下载" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Leary SC, Kaufman BA, Pellecchia G, et al. Human SCO1 and SCO2 have independent, cooperative functions in copper delivery to cytochrome c oxidase. Hum Mol Genet. 2004;13(17):1839‐1848. doi: 10.1093/hmg/ddh197 [DOI] [PubMed] [Google Scholar]
  • 37. Takahashi Y, Kako K, Kashiwabara S, et al. Mammalian copper chaperone Cox17p has an essential role in activation of cytochrome C oxidase and embryonic development. Mol Cell Biol. 2002;22(21):7614‐7621. doi: 10.1128/MCB.22.21.7614-7621.2002 ["V体育官网入口" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Zhou Y, Zhang L. The interplay between copper metabolism and microbes: in perspective of host copper‐dependent ATPases ATP7A/B. Front Cell Infect Microbiol. 2023;13:1267931. doi: 10.3389/fcimb.2023.1267931 ["VSports最新版本" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Markossian KA, Kurganov BI. Copper chaperones, intracellular copper trafficking proteins. Function, structure, and mechanism of action. Biochemistry Biokhimiia. 2003;68(8):827‐837. doi: 10.1023/a:1025740228888 [DOI] [PubMed] [Google Scholar]
  • 40. La Fontaine S, Mercer JF. Trafficking of the copper‐ATPases, ATP7A and ATP7B: role in copper homeostasis. Arch Biochem Biophys. 2007;463(2):149‐167. doi: 10.1016/j.abb.2007.04.021 [DOI] [PubMed] [Google Scholar]
  • 41. Guo Y, Nyasae L, Braiterman LT, Hubbard AL. NH2‐terminal signals in ATP7B Cu‐ATPase mediate its Cu‐dependent anterograde traffic in polarized hepatic cells. Am J Physiol Gastrointest Liver Physiol. 2005;289(5):G904‐G916. doi: 10.1152/ajpgi.00262.2005 [V体育官网入口 - DOI] [PubMed] [Google Scholar]
  • 42. Tadini‐Buoninsegni F, Smeazzetto S. Mechanisms of charge transfer in human copper ATPases ATP7A and ATP7B. IUBMB Life. 2017;69(4):218‐225. doi: 10.1002/iub.1603 [DOI (V体育ios版)] [PubMed] [Google Scholar]
  • 43. Nagaraja GM, Othman M, Fox BP, et al. Gene expression signatures and biomarkers of noninvasive and invasive breast cancer cells: comprehensive profiles by representational difference analysis, microarrays and proteomics. Oncogene. 2006;25(16):2328‐2338. doi: 10.1038/sj.onc.1209265 [DOI] [PubMed] [Google Scholar]
  • 44. Crnogorac‐Jurcevic T, Gangeswaran R, Bhakta V, et al. Proteomic analysis of chronic pancreatitis and pancreatic adenocarcinoma. Gastroenterology. 2005;129(5):1454‐1463. doi: 10.1053/j.gastro.2005.08.012 [DOI] [PubMed] [Google Scholar]
  • 45. Resh MD. Fatty acylation of proteins: the long and the short of it. Prog Lipid Res. 2016;63:120‐131. doi: 10.1016/j.plipres.2016.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Martínez‐Reyes I, Chandel NS. Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun. 2020;11(1):102. doi: 10.1038/s41467-019-13668-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Zulkifli M, Spelbring AN, Zhang Y, et al. FDX1‐dependent and independent mechanisms of elesclomol‐mediated intracellular copper delivery. Proc Natl Acad Sci USA. 2023;120(10):e2216722120. doi: 10.1073/pnas.2216722120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Vázquez G, Caballero AB, Kokinda J, Hijano A, Sabaté R, Gamez P. Copper, dityrosine cross‐links and amyloid‐β aggregation. J Biol Inorgan Chem. 2019;24(8):1217‐1229. doi: 10.1007/s00775-019-01734-6 [DOI] [PubMed] [Google Scholar]
  • 49. Galy B, Conrad M, Muckenthaler M. Mechanisms controlling cellular and systemic iron homeostasis. Nat Rev Mol Cell Biol. 2024;25(2):133‐155. doi: 10.1038/s41580-023-00648-1 [V体育安卓版 - DOI] [PubMed] [Google Scholar]
  • 50. Shan Q, Ma F, Wei J, Li H, Ma H, Sun P. Physiological functions of heat shock proteins. Curr Protein Pept Sci. 2020;21(8):751‐760. doi: 10.2174/1389203720666191111113726 [DOI] [PubMed] [Google Scholar]
  • 51. Wang D, Tian Z, Zhang P, et al. The molecular mechanisms of cuproptosis and its relevance to cardiovascular disease. Biomed Pharmacother. 2023;163:114830. doi: 10.1016/j.biopha.2023.114830 [DOI (VSports在线直播)] [PubMed] [Google Scholar]
  • 52. Fetherolf MM, Boyd SD, Taylor AB, et al. Copper‐zinc superoxide dismutase is activated through a sulfenic acid intermediate at a copper ion entry site. J Biol Chem. 2017;292(29):12025‐12040. doi: 10.1074/jbc.M117.775981 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Rajaratnam S, Soman AP, Phalguna KS, et al. Integrated omic analysis delineates pathways modulating toxic TDP‐43 protein aggregates in amyotrophic lateral sclerosis. Cells. 2023;12(9):1228. doi: 10.3390/cells12091228 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Sataranatarajan K, Pharaoh G, Brown JL, et al. Molecular changes in transcription and metabolic pathways underlying muscle atrophy in the CuZnSOD null mouse model of sarcopenia. GeroScience. 2020;42(4):1101‐1118. doi: 10.1007/s11357-020-00189-x ["VSports" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Teschke R. Aluminum, arsenic, beryllium, cadmium, chromium, cobalt, copper, iron, lead, mercury, molybdenum, nickel, platinum, thallium, titanium, vanadium, and zinc: molecular aspects in experimental liver injury. Int J Mol Sci. 2022;23(20):12213. doi: 10.3390/ijms232012213 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Lomozová Z, Hrubša M, Conte PF, et al. The effect of flavonoids on the reduction of cupric ions, the copper‐driven Fenton reaction and copper‐triggered haemolysis. Food Chem. 2022;394:133461. doi: 10.1016/j.foodchem.2022.133461 [DOI] [PubMed] [Google Scholar]
  • 57. Luo Z, Xu X, Sho T, et al. ROS‐induced autophagy regulates porcine trophectoderm cell apoptosis, proliferation, and differentiation. Am J Physiol Cell Physiol. 2019;316(2):C198‐C209. doi: 10.1152/ajpcell.00256.2018 [DOI (V体育2025版)] [PubMed] [Google Scholar]
  • 58. Tsymbal SA, Moiseeva AA, Agadzhanian NA, et al. Copper‐containing nanoparticles and organic complexes: metal reduction triggers rapid cell death via oxidative burst. Int J Mol Sci. 2021;22(20):11065. doi: 10.3390/ijms222011065 ["VSports在线直播" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Mahesh VB, Zamorano P, De Sevilla L, Lewis D, Brann DW. Characterization of ionotropic glutamate receptors in rat hypothalamus, pituitary and immortalized gonadotropin‐releasing hormone (GnRH) neurons (GT1‐7 cells). Neuroendocrinology. 1999;69(6):397‐407. doi: 10.1159/000054442 [DOI] [PubMed] [Google Scholar]
  • 60. Tanaka KI, Shimoda M, Kasai M, Ikeda M, Ishima Y, Kawahara M. Involvement of SAPK/JNK signaling pathway in copper enhanced zinc‐induced neuronal cell death. Toxicol Sci. 2019;169(1):293‐302. doi: 10.1093/toxsci/kfz043 [DOI] [PubMed] [Google Scholar]
  • 61. Anchordoquy JM, Anchordoquy JP, Nikoloff N, Pascua AM, Furnus CC. High copper concentrations produce genotoxicity and cytotoxicity in bovine cumulus cells. Environ Sci Pollut Res Int. 2017;24(24):20041‐20049. doi: 10.1007/s11356-017-9683-0 [DOI] [PubMed] [Google Scholar]
  • 62. Siivola KM, Suhonen S, Hartikainen M, Catalán J, Norppa H. Genotoxicity and cellular uptake of nanosized and fine copper oxide particles in human bronchial epithelial cells in vitro. Mutation Res. 2020;856–857:503217. doi: 10.1016/j.mrgentox.2020.503217 [DOI] [PubMed] [Google Scholar]
  • 63. Alarifi S, Ali D, Verma A, Alakhtani S, Ali BA. Cytotoxicity and genotoxicity of copper oxide nanoparticles in human skin keratinocytes cells. Int J Toxicol. 2013;32(4):296‐307. doi: 10.1177/1091581813487563 [DOI] [PubMed] [Google Scholar]
  • 64. Xu K, Tang Z, Liu S, et al. Effects of low concentrations copper on antioxidant responses, DNA damage and genotoxicity in thick shell mussel Mytilus coruscus . Fish Shellfish Immunol. 2018;82:77‐83. doi: 10.1016/j.fsi.2018.08.016 ["VSports最新版本" DOI] [PubMed] [Google Scholar]
  • 65. Cadavid‐Vargas JF, Leon IE, Etcheverry SB, Santi E, Torre MH, Di Virgilio AL. Copper(II) complexes with saccharinate and glutamine as antitumor agents: cytoand genotoxicity in human osteosarcoma cells. Anti Cancer Agents Med Chem. 2017;17(3):424‐433. doi: 10.2174/1871520616666160513130204 [DOI] [PubMed] [Google Scholar]
  • 66. Alimba CG, Dhillon V, Bakare AA, Fenech M. Genotoxicity and cytotoxicity of chromium, copper, manganese and lead, and their mixture in WIL2‐NS human B lymphoblastoid cells is enhanced by folate depletion. Mutation Res. 2016;798‐799:35‐47. doi: 10.1016/j.mrgentox.2016.02.002 [DOI] [PubMed] [Google Scholar]
  • 67. Morris DL Jr. DNA‐bound metal ions: recent developments. Biomol Concepts. 2014;5(5):397‐407. doi: 10.1515/bmc-2014-0021 [DOI] [PubMed] [Google Scholar]
  • 68. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35(4):495‐516. doi: 10.1080/01926230701320337 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Chen J. The cell‐cycle arrest and apoptotic functions of p53 in tumor initiation and progression. Cold Spring Harb Perspect Med. 2016;6(3):a026104. doi: 10.1101/cshperspect.a026104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Saelens X, Festjens N, Vande Walle L, van Gurp M, van Loo G, Vandenabeele P. Toxic proteins released from mitochondria in cell death. Oncogene. 2004;23(16):2861‐2874. doi: 10.1038/sj.onc.1207523 [DOI] [PubMed] [Google Scholar]
  • 71. Cai J, Yang J, Jones DP. Mitochondrial control of apoptosis: the role of cytochrome c. Biochim Biophys Acta. 1998;1366(1–2):139‐149. doi: 10.1016/s0005-2728(98)00109-1 [DOI] [PubMed] [Google Scholar]
  • 72. Chinnaiyan AM. The Apoptosome: Heart and Soul of the Cell Death Machine. Neoplasia (New York, NY). 1999;1(1):5‐15. doi: 10.1038/sj.neo.7900003 [DOI (VSports注册入口)] [PMC free article] [PubMed] [Google Scholar]
  • 73. Koenig U, Eckhart L, Tschachler E. Evidence that caspase‐13 is not a human but a bovine gene. Biochem Biophys Res Commun. 2001;285(5):1150‐1154. doi: 10.1006/bbrc.2001.5315 [DOI] [PubMed] [Google Scholar]
  • 74. Zhang Y, Zhou Q, Lu L, et al. Copper induces cognitive impairment in mice via modulation of cuproptosis and CREB signaling. Nutrients. 2023;15(4):972. doi: 10.3390/nu15040972 [V体育平台登录 - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Yang F, Pei R, Zhang Z, et al. Copper induces oxidative stress and apoptosis through mitochondria‐mediated pathway in chicken hepatocytes. Toxicol In Vitro. 2019;54:310‐316. doi: 10.1016/j.tiv.2018.10.017 [DOI] [PubMed] [Google Scholar]
  • 76. Xie Y, Kang R, Sun X, et al. Posttranslational modification of autophagy‐related proteins in macroautophagy. Autophagy. 2015;11(1):28‐45. doi: 10.4161/15548627.2014.984267 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Levine B, Kroemer G. Biological functions of autophagy genes: a disease perspective. Cell. 2019;176(1–2):11‐42. doi: 10.1016/j.cell.2018.09.048 [DOI (VSports注册入口)] [PMC free article] [PubMed] [Google Scholar]
  • 78. Kim J, Kundu M, Viollet B, Guan KL. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 2011;13(2):132‐141. doi: 10.1038/ncb2152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Wirawan E, Van den Berghe T, Lippens S, Agostinis P, Vandenabeele P. Autophagy: for better or for worse. Cell Res. 2012;22(1):43‐61. doi: 10.1038/cr.2011.152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Russell RC, Tian Y, Yuan H, et al. ULK1 induces autophagy by phosphorylating Beclin‐1 and activating VPS34 lipid kinase. Nat Cell Biol. 2013;15(7):741‐750. doi: 10.1038/ncb2757 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Ao X, Zou L, Wu Y. Regulation of autophagy by the Rab GTPase network. Cell Death Differ. 2014;21(3):348‐358. doi: 10.1038/cdd.2013.187 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Denton D, Kumar S. Autophagy‐dependent cell death. Cell Death Differ. 2019;26(4):605‐616. doi: 10.1038/s41418-018-0252-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Wan F, Zhong G, Ning Z, et al. Long‐term exposure to copper induces autophagy and apoptosis through oxidative stress in rat kidneys. Ecotoxicol Environ Saf. 2020;190:110158. doi: 10.1016/j.ecoenv.2019.110158 [DOI] [PubMed] [Google Scholar]
  • 84. Polishchuk EV, Merolla A, Lichtmannegger J, et al. Activation of autophagy, observed in liver tissues from patients with Wilson disease and from ATP7B‐deficient animals, protects hepatocytes from copper‐induced apoptosis. Gastroenterology. 2019;156(4):1173‐1189.e5. doi: 10.1053/j.gastro.2018.11.032 [DOI] [PubMed] [Google Scholar]
  • 85. Tsang T, Posimo JM, Gudiel AA, Cicchini M, Feldser DM, Brady DC. Copper is an essential regulator of the autophagic kinases ULK1/2 to drive lung adenocarcinoma. Nat Cell Biol. 2020;22(4):412‐424. doi: 10.1038/s41556-020-0481-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Xue Q, Yan D, Chen X, et al. Copper‐dependent autophagic degradation of GPX4 drives ferroptosis. Autophagy. 2023;19(7):1982‐1996. doi: 10.1080/15548627.2023.2165323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Yu Z, Zhou R, Zhao Y, et al. Blockage of SLC31A1‐dependent copper absorption increases pancreatic cancer cell autophagy to resist cell death. Cell Prolif. 2019;52(2):e12568. doi: 10.1111/cpr.12568 [V体育平台登录 - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Møller LB, Mogensen M, Horn N. Molecular diagnosis of Menkes disease: genotype‐phenotype correlation. Biochimie. 2009;91(10):1273‐1277. doi: 10.1016/j.biochi.2009.05.011 [DOI] [PubMed] [Google Scholar]
  • 89. Tümer Z, Møller LB. Menkes disease. Eur J Human Genet. 2010;18(5):511‐518. doi: 10.1038/ejhg.2009.187 ["V体育平台登录" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Kaler SG, Goldstein DS, Holmes C, Salerno JA, Gahl WA. Plasma and cerebrospinal fluid neurochemical pattern in Menkes disease. Ann Neurol. 1993;33(2):171‐175. doi: 10.1002/ana.410330206 [DOI] [PubMed] [Google Scholar]
  • 91. Ojha R, Prasad AN. Menkes disease: what a multidisciplinary approach can do. J Multidiscip Healthc. 2016;9:371‐385. doi: 10.2147/JMDH.S93454 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Shim H, Harris ZL. Genetic defects in copper metabolism. J Nutr. 2003;133(5 suppl 1):1527S‐1531S. doi: 10.1093/jn/133.5.1527S [DOI] [PubMed] [Google Scholar]
  • 93. Ashrafi MR, Ghasemi D, Safavi M. Menkes disease. Arch Iran Med. 2021;24(12):919‐920. doi: 10.34172/aim.2021.138 [DOI] [PubMed] [Google Scholar]
  • 94. Byers PH, Siegel RC, Holbrook KA, Narayanan AS, Bornstein P, Hall JG. X‐linked cutis laxa: defective cross‐link formation in collagen due to decreased lysyl oxidase activity. N Engl J Med. 1980;303(2):61‐65. doi: 10.1056/NEJM198007103030201 [DOI] [PubMed] [Google Scholar]
  • 95. Szauter KM, Cao T, Boyd CD, Csiszar K. Lysyl oxidase in development, aging and pathologies of the skin. Pathol Biol. 2005;53(7):448‐456. doi: 10.1016/j.patbio.2004.12.033 [DOI] [PubMed] [Google Scholar]
  • 96. Royce PM, Camakaris J, Danks DM. Reduced lysyl oxidase activity in skin fibroblasts from patients with Menkes' syndrome. Biochem J. 1980;192(2):579‐586. doi: 10.1042/bj1920579 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Kim JH, Lee BH, Kim YM, et al. Novel mutations and clinical outcomes of copper‐histidine therapy in Menkes disease patients. Metab Brain Dis. 2015;30(1):75‐81. doi: 10.1007/s11011-014-9569-5 [DOI] [PubMed] [Google Scholar]
  • 98. Vairo FPE, Chwal BC, Perini S, Ferreira MAP, de Freitas Lopes AC, Saute JAM. A systematic review and evidence‐based guideline for diagnosis and treatment of Menkes disease. Mol Genet Metab. 2019;126(1):6‐13. doi: 10.1016/j.ymgme.2018.12.005 [DOI] [PubMed] [Google Scholar]
  • 99. George DH, Casey RE. Menkes disease after copper histidine replacement therapy: case report. Pediatr Develop Pathol. 2001;4(3):281‐288. doi: 10.1007/s100240010142 [DOI] [PubMed] [Google Scholar]
  • 100. Kaler SG, Holmes CS, Goldstein DS, et al. Neonatal diagnosis and treatment of Menkes disease. N Engl J Med. 2008;358(6):605‐614. doi: 10.1056/NEJMoa070613 [DOI (VSports在线直播)] [PMC free article] [PubMed] [Google Scholar]
  • 101. Fujisawa C, Kodama H, Sato Y, et al. Early clinical signs and treatment of Menkes disease. Mol Genet Metab Rep. 2022;31:100849. doi: 10.1016/j.ymgmr.2022.100849 ["VSports注册入口" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Kaler SG. Neurodevelopment and brain growth in classic Menkes disease is influenced by age and symptomatology at initiation of copper treatment. J Trace Elem Med Biol. 2014;28(4):427‐430. doi: 10.1016/j.jtemb.2014.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Fosmire GJ. Zinc toxicity. Am J Clin Nutr. 1990;51(2):225‐227. doi: 10.1093/ajcn/51.2.225 [VSports - DOI] [PubMed] [Google Scholar]
  • 104. Ernst B, Thurnheer M, Schultes B. Copper deficiency after gastric bypass surgery. Obesity (Silver Spring, Md). 2009;17(11):1980‐1981. doi: 10.1038/oby.2009.237 [VSports最新版本 - DOI] [PubMed] [Google Scholar]
  • 105. Sternlieb I, Janowitz HD. Absorption of copper in malabsorption syndromes. J Clin Invest. 1964;43(6):1049‐1055. doi: 10.1172/JCI104988 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Linder MC, Hazegh‐Azam M. Copper biochemistry and molecular biology. Am J Clin Nutr. 1996;63(5):797S‐811S. doi: 10.1093/ajcn/63.5.797 [DOI] [PubMed] [Google Scholar]
  • 107. Bleackley MR, Wong AY, Hudson DM, Wu CH, Macgillivray RT. Blood iron homeostasis: newly discovered proteins and iron imbalance. Transfus Med Rev. 2009;23(2):103‐123. doi: 10.1016/j.tmrv.2008.12.001 [DOI] [PubMed] [Google Scholar]
  • 108. Myint ZW, Oo TH, Thein KZ, Tun AM, Saeed H. Copper deficiency anemia: review article. Ann Hematol. 2018;97(9):1527‐1534. doi: 10.1007/s00277-018-3407-5 [DOI (VSports)] [PubMed] [Google Scholar]
  • 109. Schleper B, Stuerenburg HJ. Copper deficiency‐associated myelopathy in a 46‐year‐old woman. J Neurol. 2001;248(8):705‐706. doi: 10.1007/s004150170118 [DOI] [PubMed] [Google Scholar]
  • 110. Bandmann O, Weiss KH, Kaler SG. Wilson's disease and other neurological copper disorders. Lancet Neurol. 2015;14(1):103‐113. doi: 10.1016/S1474-4422(14)70190-5 [VSports最新版本 - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Członkowska A, Litwin T, Dusek P, et al. Wilson disease. Nat Rev Dis Primers. 2018;4(1):21. doi: 10.1038/s41572-018-0018-3 [DOI (VSports在线直播)] [PMC free article] [PubMed] [Google Scholar]
  • 112. Mounajjed T, Oxentenko AS, Qureshi H, Smyrk TC. Revisiting the topic of histochemically detectable copper in various liver diseases with special focus on venous outflow impairment. Am J Clin Pathol. 2013;139(1):79‐86. doi: 10.1309/AJCPDZR4OHDQNG3L [DOI] [PubMed] [Google Scholar]
  • 113. Polishchuk EV, Polishchuk RS. The emerging role of lysosomes in copper homeostasis. Metallomics. 2016;8(9):853‐862. doi: 10.1039/c6mt00058d [DOI] [PubMed] [Google Scholar]
  • 114. Lang PA, Schenck M, Nicolay JP, et al. Liver cell death and anemia in Wilson disease involve acid sphingomyelinase and ceramide. Nat Med. 2007;13(2):164‐170. doi: 10.1038/nm1539 [V体育安卓版 - DOI] [PubMed] [Google Scholar]
  • 115. Linder MC. Ceruloplasmin and other copper binding components of blood plasma and their functions: an update. Metallomics. 2016;8(9):887‐905. doi: 10.1039/c6mt00103c [DOI] [PubMed] [Google Scholar]
  • 116. Horoupian DS, Sternlieb I, Scheinberg IH. Neuropathological findings in penicillamine‐treated patients with Wilson's disease. Clin Neuropathol. 1988;7(2):62‐67. ["V体育官网入口" PubMed] [Google Scholar]
  • 117. Scheiber IF, Dringen R. Copper‐treatment increases the cellular GSH content and accelerates GSH export from cultured rat astrocytes. Neurosci Lett. 2011;498(1):42‐46. doi: 10.1016/j.neulet.2011.04.058 ["V体育2025版" DOI] [PubMed] [Google Scholar]
  • 118. Freeman MR. Specification and Morphogenesis of Astrocytes. Science (New York, NY). 2010;330(6005):774‐778. doi: 10.1126/science.1190928 [V体育平台登录 - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Bertrand E, Lewandowska E, Szpak GM, et al. Neuropathological analysis of pathological forms of astroglia in Wilson's disease. Folia Neuropathol. 2001;39(2):73‐79. ["V体育安卓版" PubMed] [Google Scholar]
  • 120. Yang Y, Wei T, Yang W, et al. Dysfunction of the lenticular nucleus is associated with dystonia in Wilson's disease. Brain Sci. 2022;13(1):7. doi: 10.3390/brainsci13010007 [DOI (VSports在线直播)] [PMC free article] [PubMed] [Google Scholar]
  • 121. Członkowska A, Litwin T. Wilson disease—currently used anticopper therapy. Handb Clin Neurol. 2017;142:181‐191. doi: 10.1016/B978-0-444-63625-6.00015-X [DOI] [PubMed] [Google Scholar]
  • 122. Cauza E, Maier‐Dobersberger T, Polli C, Kaserer K, Kramer L, Ferenci P. Screening for Wilson's disease in patients with liver diseases by serum ceruloplasmin. J Hepatol. 1997;27(2):358‐362. doi: 10.1016/s0168-8278(97)80182-1 [DOI] [PubMed] [Google Scholar]
  • 123. Korman JD, Volenberg I, Balko J, et al. Screening for Wilson disease in acute liver failure: a comparison of currently available diagnostic tests. Hepatology (Baltimore, Md). 2008;48(4):1167‐1174. doi: 10.1002/hep.22446 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Walshe JM. Serum ‘free’ copper in Wilson disease. QJM. 2012;105(5):419‐423. doi: 10.1093/qjmed/hcr229 [DOI] [PubMed] [Google Scholar]
  • 125. Ferenci P, Caca K, Loudianos G, et al. Diagnosis and phenotypic classification of Wilson disease. Liver Int. 2003;23(3):139‐142. doi: 10.1034/j.1600-0676.2003.00824.x [DOI] [PubMed] [Google Scholar]
  • 126. Ahmad A, Torrazza‐Perez E, Schilsky ML. Liver transplantation for Wilson disease. Handb Clin Neurol. 2017;142:193‐204. doi: 10.1016/B978-0-444-63625-6.00016-1 [DOI] [PubMed] [Google Scholar]
  • 127. Grasso M, Bond GJ, Kim YJ, et al. The copper chaperone CCS facilitates copper binding to MEK1/2 to promote kinase activation. J Biol Chem. 2021;297(6):101314. doi: 10.1016/j.jbc.2021.101314 [DOI (V体育平台登录)] [PMC free article] [PubMed] [Google Scholar]
  • 128. Ding X, Jiang M, Jing H, et al. Analysis of serum levels of 15 trace elements in breast cancer patients in Shandong, China. Environ Sci Pollut Res Int. 2015;22(10):7930‐7935. doi: 10.1007/s11356-014-3970-9 [DOI] [PubMed] [Google Scholar]
  • 129. Pavithra V, Sathisha TG, Kasturi K, Mallika DS, Amos SJ, Ragunatha S. Serum levels of metal ions in female patients with breast cancer. J Clin Diagn Res. 2015;9(1):BC25–c27. doi: 10.7860/JCDR/2015/11627.5476 [V体育2025版 - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Zowczak M, Iskra M, Torliński L, Cofta S. Analysis of serum copper and zinc concentrations in cancer patients. Biol Trace Elem Res. 2001;82(1–3):1‐8. doi: 10.1385/BTER:82:1-3:001 [DOI] [PubMed] [Google Scholar]
  • 131. Stepien M, Jenab M, Freisling H, et al. Pre‐diagnostic copper and zinc biomarkers and colorectal cancer risk in the European Prospective Investigation into Cancer and Nutrition cohort. Carcinogenesis. 2017;38(7):699‐707. doi: 10.1093/carcin/bgx051 ["VSports app下载" DOI] [PubMed] [Google Scholar]
  • 132. Sohrabi M, Gholami A, Azar MH, et al. Trace element and heavy metal levels in colorectal cancer: comparison between cancerous and non‐cancerous tissues. Biol Trace Elem Res. 2018;183(1):1‐8. doi: 10.1007/s12011-017-1099-7 [DOI] [PubMed] [Google Scholar]
  • 133. Saleh SAK, Adly HM, Abdelkhaliq AA, Nassir AM. Serum levels of selenium, zinc, copper, manganese, and iron in prostate cancer patients. Curr Urol. 2020;14(1):44‐49. doi: 10.1159/000499261 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Yaman M, Kaya G, Yekeler H. Distribution of trace metal concentrations in paired cancerous and non‐cancerous human stomach tissues. World J Gastroenterol. 2007;13(4):612‐618. doi: 10.3748/wjg.v13.i4.612 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Mortada WI, Awadalla A, Khater S, et al. Copper and zinc levels in plasma and cancerous tissues and their relation with expression of VEGF and HIF‐1 in the pathogenesis of muscle invasive urothelial bladder cancer: a case‐controlled clinical study. Environ Sci Pollut Res Int. 2020;27(13):15835‐15841. doi: 10.1007/s11356-020-08113-8 ["V体育安卓版" DOI] [PubMed] [Google Scholar]
  • 136. Conforti RA, Delsouc MB, Zabala AS, Vallcaneras SS, Casais M. The copper chelator ammonium tetrathiomolybdate inhibits the progression of experimental endometriosis in TNFR1‐deficient mice. Sci Rep. 2023;13(1):10354. doi: 10.1038/s41598-023-37031-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Shanbhag V, Jasmer‐McDonald K, Zhu S, et al. ATP7A delivers copper to the lysyl oxidase family of enzymes and promotes tumorigenesis and metastasis. Proc Natl Acad Sci USA. 2019;116(14):6836‐6841. doi: 10.1073/pnas.1817473116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Blockhuys S, Celauro E, Hildesjö C, et al. Defining the human copper proteome and analysis of its expression variation in cancers. Metallomics. 2017;9(2):112‐123. doi: 10.1039/c6mt00202a [DOI] [PubMed] [Google Scholar]
  • 139. da Silva DA, De Luca A, Squitti R, et al. Copper in tumors and the use of copper‐based compounds in cancer treatment. J Inorg Biochem. 2022;226:111634. doi: 10.1016/j.jinorgbio.2021.111634 [DOI] [PubMed] [Google Scholar]
  • 140. Crowe A, Jackaman C, Beddoes KM, Ricciardo B, Nelson DJ. Rapid copper acquisition by developing murine mesothelioma: decreasing bioavailable copper slows tumor growth, normalizes vessels and promotes T cell infiltration. PLoS ONE. 2013;8(8):e73684. doi: 10.1371/journal.pone.0073684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Pan Q, Bao LW, Merajver SD. Tetrathiomolybdate inhibits angiogenesis and metastasis through suppression of the NFkappaB signaling cascade. Mol Cancer Res. 2003;1(10):701‐706. [PubMed] [Google Scholar]
  • 142. Xie J, Yang Y, Gao Y, He J. Cuproptosis: mechanisms and links with cancers. Mol Cancer. 2023;22(1):46. doi: 10.1186/s12943-023-01732-y [VSports - DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Denoyer D, Pearson HB, Clatworthy SA, et al. Copper as a target for prostate cancer therapeutics: copper‐ionophore pharmacology and altering systemic copper distribution. Oncotarget. 2016;7(24):37064‐37080. doi: 10.18632/oncotarget.9245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Blackman RK, Cheung‐Ong K, Gebbia M, et al. Mitochondrial electron transport is the cellular target of the oncology drug elesclomol. PLoS ONE. 2012;7(1):e29798. doi: 10.1371/journal.pone.0029798 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Zhang H, Chen D, Ringler J, et al. Disulfiram treatment facilitates phosphoinositide 3‐kinase inhibition in human breast cancer cells in vitro and in vivo. Cancer Res. 2010;70(10):3996‐4004. doi: 10.1158/0008-5472.CAN-09-3752 ["V体育安卓版" DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Lun X, Wells JC, Grinshtein N, et al. Disulfiram when combined with copper enhances the therapeutic effects of temozolomide for the treatment of glioblastoma. Clin Cancer Res. 2016;22(15):3860‐3875. doi: 10.1158/1078-0432.CCR-15-1798 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement (V体育官网入口)

Data sharing is not applicable to this article as no new data were created or analyzed in this study.


Articles from CNS Neuroscience & Therapeutics are provided here courtesy of Wiley

RESOURCES (V体育2025版)