- Research
- Open access
- Published:
Curcumin suppresses copper accumulation in non-small cell lung cancer by binding ATOX1
BMC Pharmacology and Toxicology volume 25, Article number: 54 (2024)
Abstract
Background
Non-small cell lung cancer (NSCLC) is associated with intracellular copper accumulation. Antioxidant 1 (ATOX1) is a copper chaperone. This study aimed to analyze the anti-cancer effects of curcumin on the ATOX1-mediated copper pathway in NSCLC.
Methods
A binding activity between curcumin and ATOX1 was measured using molecular docking. NSCLC cells, A549 and H1299, were treated with different doses of curcumin (10, 20, 40 µM) or DC-AC50 (5, 10, 20 µM) for 24 h. The cell viability and levels of ATOX1, ATP7A and COX17 proteins were observed in cells. Overexpressing ATOX1 in cells was established by pcDNA3.1-ATOX1 transfection for 24 h. The ATOX1 overexpressing cells were treated with 40 µM curcumin or 20 µM DC-AC50 for 24 h to analyze the mechanism of curcumin in NSCLC treatment. Cell viability was measured by CCK-8, and levels of proteins were measured by western blotting. The copper level in cells was labeled by copper sensor-1. Moreover, nude mice models were induced by injection of A549 cells and treated with 20 mg/kg/d DC-AC50 or 40 mg/kg/d curcumin. Tumor growth was observed by measuring tumor volume and tumor weight. The levels of ATOX1, ATP7A and COX17 in tumors were measured by immunohistochemistry and western blotting.
Results
Curcumin bound to ATOX1 (score = −6.1 kcal/mol) and decreased the levels of ATOX1, ATP7A and COX17 proteins in NSCLC cells. The curcumin or DC-AC50 treatment suppressed cell viability by inhibiting the ATOX1-mediated copper signaling in NSCLC cells. The ATOX1 overexpression in cells significantly weakened the effects of curcumin on suppressing copper accumulation and the ATOX1-mediated copper pathway (p < 0.05). In mice models, curcumin or DC-AC50 treatment also suppressed tumor growth by suppressing the ATOX1-mediated copper pathway in tumors.
Conclusion
This study demonstrated that curcumin bound ATOX1 to suppress copper accumulation in NSCLC cells, providing a new mechanism of curcumin for NSCLC treatment.
Introduction
Copper is an essential micronutrient required as a catalytic cofactor for several metalloenzymes [1]. Copper-dependent enzymes are crucial for maintaining cellular biological processes, such as oxidative stress, biosynthesis of chemical messengers, and pigment construction [2]. Many studies have shown that high levels of copper are closely related to tumor proliferation and invasion [3, 4]. Clinical studies have found that the prognosis for patients with non-small cell lung cancer (NSCLC) is associated with intracellular copper levels [5]. In preclinical models of lung cancer, copper chelation therapy has demonstrated a favorable response [6]. Therefore, finding an effective compound with little side effect to control the copper accumulation in NSCLC is necessary.
Traditional Chinese medicine (TCM) shows significant benefits on prolonging survival time and improving life quality among patients with NSCLC [7]. Curcumin is a well-known polyphenol derived from the plant Curcuma longa, which shows anticancer activities through many mechanisms, such as oxidative stress, ferroptosis, and autophagy [8]. Recently, researchers have found that curcumin can induce ferroptosis and cuproptosis in hepatocellular carcinoma (HCC) [9]. Cuproptosis is characterized by elevated toxic intracellular copper levels, which triggers oxidative stress and culminates in cell demise [3]. Moreover, curcumin can prevent cisplatin-induced ototoxicity by decreasing the cuproptosis-related proteins in the inner ear organ of corti-1 cells and animal models (zebrafish and guinea pigs) [10]. However, the anti-cancer effects of curcumin on controlling the copper accumulation in NSCLC are unknown.
Copper transport within cells is facilitated by tiny molecules called copper chaperones, including antioxidant 1 (ATOX1), cytochrome c oxidase 17 (COX17), and copper chaperone for superoxide dismutase (CCS) [6]. ATOX1 collects copper that has entered the cell and delivers the metal to two multidomain P1B-type adenosine triphosphatases (ATPases), ATP7A and ATP7B [11]. The ATP7A/7B is essential for properly functioning cellular processes, such as redox homeostasis, energy production, and cell signaling [12]. Previous studies have shown that ATP7A/7B is considered to be a potential target for the NSCLC [13, 14]. Overexpressing ATP7B leads to a decreased accumulation and more rapid efflux of copper in NSCLC cells, leading to diminished effects of disulfiram on cancer cell clonogenic cell killing [15]. However, there is little research on the ATP7A-mediated copper signaling in NSCLC. ATOX1 is a prognostic marker and therapeutic target in various cancers, including colorectal cancer [16], breast cancer [17], and NSCLC [6]. In NSCLC cells, ATOX1 knockdown inhibited the copper-stimulated cell proliferation [18]. Therefore, it is necessary to investigate the association between curcumin and ATOX1-mediated copper signaling in treating NSCLC.
This study first analyzed a binding activity between curcumin and ATOX1 protein, and observed the effects of different doses of curcumin treatment on levels of copper-dependent proteins (ATOX1, ATP7A, and COX17) in NSCLC cells (A549 and H1299). Next, this research studied the association between curcumin and ATOX1-mediated copper signaling in NSCLC cells and animal models.
Materials and methods
Binding activity between curcumin and ATOX1 protein
According to a reported method [19], three-dimensional (3D) structures of ATOX1 and curcumin (CAS: 458-37-7) were separately downloaded from the PDB (https://www.rcsb.org/) and PubChem (https://pubchem.ncbi.nlm.nih.gov/). The molecular docking between ATOX1 and curcumin was performed by the AutoDock Vina tool using the UCSF chimera software. The stability of the curcumin-ATOX1 binding is represented using intermolecular energy (kcal/mol).
Cell culture
Human NSCLC cell lines, A549 (#CL-0016) and H1299 (#CL-0165), were purchased from the Wuhan Procella Biotechnology Co., Ltd. (Procell.com.cn, Wuhan, China) and identified by the short tandem repeats (STR). The cells were cultured in Roswell Park Memorial Institute (RPMI) 1640 medium containing 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin at 37 °C with 5% CO2. At about 80% confluency, the cells were used for experiments.
Curcumin treatment
Curcumin (#805204, Macklin. cn, Shanghai, China) was dissolved in 0.1% dimethyl sulfoxide (DMSO). The cells (A549 and H1299) were treated with different doses of curcumin (10, 20, 40 µM), and the 0.1% DMSO treatment was used as vehicle treatment. After 24 h, the cell viability was measured by cell counting kit-8 (CCK-8) kit. The levels of ATOX1, ATP7A and COX17 proteins in cells were measured by western blotting.
ATOX1 overexpression
The pcDNA3.1-ATOX1 plasmid and pcDNA3.1 plasmid control were purchased from GenePharma (Shanghai, China). The primers used for the amplification of ATOX1 were as follows: sense 5′-ccggatccatgccgaagcacgagttct-3′ and antisense 5′-ttgaattctcactactcaaggccaaggtagg-3′. The cells (A549 and H1299) were transfected with 2 µg of pcDNA3.1-ATOX1 plasmid or pcDNA3.1 plasmid control using x-tremegene HP DNA transfection reagent (Roche Diagnostics, Mannheim, Germany) for 24 h at 37 °C with 5% CO2. After transfection, the cell viability and expression of ATOX1 protein were observed.
DC-AC50 treatment
DC-AC50 is a dual inhibitor of ATOX1 and CCS [20]. The DC-AC50 was purchased from MedChemExpress (#HY-107636, Shanghai, China) and dissolved in 0.1% DMSO. The cells (A549 and H1299) were treated with different doses of DC-AC50 (5, 10, 20 µM), and the 0.1% DMSO treatment was used as vehicle treatment. After 24 h, the cell viability was measured by a CCK-8 kit.
Cell group
Based on the results of curcumin or DC-AC50 treatment on cell viability, 40 µM curcumin and 20 µM DC-AC50 were used for cell grouping research. The cells were divided into four groups: control group, curcumin group, DC-AC50 group, and pcDNA-ATOX1 + curcumin group. In the control group, cells were treated with 0.1% DMSO. In the curcumin group, the cells were treated with 40 µM curcumin. In the DC-AC50 group, the cells were treated with 20 µM DC-AC50. In the pcDNA-ATOX1 + curcumin group, the pcDNA3.1-ATOX1 plasmid-transfected cells were treated with 40 µM curcumin. After 24 h culture, the cells were collected for experiments.
Cell viability
Cells (2 × 104 per well) were cultured in a 96-well plate and cultured with 10 µL of CCK-8 solution (#C0037, Beyotime, China) for 4 h at 37 °C with 5% CO2. The absorbance of each well was measured at 450 nm by a microplate reader (#ELx808, BioTek, USA). Cell viability (%) was calculated as [(As-Ab)/(Ac-Ab)] × 100, where As, Ab, and Ac are absorbance in test wells, blank wells, and control wells, respectively.
Protein expression
According to the methods of western blotting [21], the cells or tissues were lysed in a RIPA buffer and collected at 4 °C at 20,000 g for 20 min. The concentration of proteins was measured using a bicinchoninic acid reagent (#B9643-1L, Sigma, China). The proteins (1 µg/µL, 20 µL) were diluted with loading buffer and separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE, #P0690, Beyotime, China). The separated proteins were transferred to polyvinylidene fluoride membranes (PVDF, #FFP20, Beyotime, China) and blocked with 5% milk PBST (PBS, 0.1% Tween 20, pH 7.2) for 60 min at 25 °C. After washing with PBST, the membranes were soaked in methanol for 5 min, and washed with PBST three times (5 min each time). The primary antibodies were diluted in 5% milk PBST and incubated with the membranes at 4 °C overnight. The primary antibodies included rabbit anti-ATOX1 (1:1500, #A6874, ABclonal, China), rabbit anti-ATP7A (1:1500, #DF8506, Affinity, China), rabbit anti-COX17 (1:1500, #DF0626, Affinity, China), and rabbit anti-beta actin (1:8000, #DF7018, Affinity, China). After washing with PBST three times, the membranes were incubated with the goat anti-rabbit IgG (H + L) HRP (1:8000, #S0001, Affinity, China) for 120 min at 25 °C. After washing, the membranes were visualized by chemiluminescence. The semi-quantitative analysis of the blots was performed by densitometry using ImageJ software [21].
Copper sensor-1 labeling
Cells (1 × 104/mL) were cultured in a 35-mm Petri dish containing 3 mL PBS buffer and labeled using 15 µL of a 1mM copper sensor-1 solution (#HY-141511, MedChemExpress, Shanghai, China). The copper sensor-1 was dissolved in DMSO. After incubation for 15 min in the dark at 25 °C, the results were observed at 543 nm excitation under a confocal microscope (#LSM800, Zeiss, Germany). The semi-quantitative analysis of the results was conducted by mean gray value using ImageJ software [21].
Animals
Eighteen male BALB/c nude mice (5 weeks, 90–120 g) were obtained from Ji’nan Pengyue Laboratory Animal Breeding Co., Ltd (Ji’nan, Shandong, China). All mice were housed in a sterile environment (20–23 °C, 50–60% humidity) with a 12 h/12 h light-dark cycle, providing unlimited access to water and food.
Animal treatment
To reduce the numbers of animals, the A549 cells were used for the animal experiments. The mice were intraperitoneally anesthetized using pentobarbital sodium (45 mg/kg) and injected with 50 µL of A549 cells (1 × 106) in the subcutaneous tissue of the right hind limb. After 6 days, the mice were randomly divided into three groups: control group, DC-AC50 group, and curcumin group. In the control group, mice were treated with 1% DMSO by intraperitoneal injection for 21 days. In the DC-AC50 or curcumin group, mice were treated with 20 mg/kg/d DC-AC50 or 40 mg/kg/d curcumin by intraperitoneal injection for 21 days. Tumor growth was measured weekly by the same person. Tumor volume = 1/2 (length × width2).
Sample collection
After 21 days from the injection, the mice were sacrificed using pentobarbital sodium (120 mg/kg) intraperitoneal injection. Overdose of anesthesia can cause animals to stop breathing, followed by cardiac arrest. The tumors were collected and weighed. The levels of ATOX1, ATP7A and COX17 in tumors were measured by western blotting and immunohistochemistry.
Immunohistochemistry
According to the immunohistochemistry staining procedures [22], the paraffin-embedded tissues were cut into 3 μm thick sections. The sections were soaked in xylene twice (12 min each time) and 100%, 95%, 80%, and 70% ethanol (2 min each time). After that, the sections were washed with PBS and soaked in sodium citrate antigen repair solution for 15 min at 100 °C. The washed sections were cultured with 3% H2O2 for 15 min and incubated with primary antibodies overnight at 4 °C. The primary antibodies included rabbit anti-ATOX1 (1:150, #DF13934, Affinity, China), rabbit anti-ATP7A (1:150, #DF8506, Affinity, China) and rabbit anti-COX17 (1:150, #DF0626, Affinity, China). After washing, the sections were incubated with goat anti-rabbit IgG antibody (1:1000, #S0001, Affinity, China) for 120 min at 25 °C. Finally, the sections were soaked in ethanol (90%, 95%, 100%) for 3 min each time and xylene twice (5 min each time). After the coverslip, a light microscope (DM1000 LED, Leica, Germany) was used to observe the results. The quantitative analysis of the results was conducted by stained area percentage using ImageJ software [21].
Statistics
Statistics were performed by SPSS 20.0 statistical analysis software (IBM, Chicago, IL, USA) and the results were expressed as the mean ± standard deviation. The ANOVA with post hoc Tukey’s test was used to compare significance among multiple groups. P < 0.05 is considered as significant.
Results
Curcumin suppressed ATOX1 in A549 and H1299 cells
In this study, it was found that curcumin and ATOX1 have a good binding activity (score = −6.1 kcal/mol, Fig. 1A). Observing the cell viability of A549 and H1299 cells (Fig. 1B), no significance was found between the control cells and the vehicle-treated cells, but the cell viability was significantly decreased with the increasing dose of curcumin (p < 0.05). Observing the levels of copper chaperones, ATOX1, ATP7A, and COX17 (Fig. 1C), no significance was found between the control cells and the vehicle-treated cells, but their levels were significantly decreased with the increasing dose of curcumin in the A549 and H1299 cells (p < 0.05, Fig. 1D). Moreover, the cell viability of A549 and H1299 cells was observed after treatment with different doses of DC-AC50 (Fig. 1E). It showed that no significance was found between the control cells and the vehicle-treated cells, but the cell viability decreased with the increasing dose of DC-AC50 (p < 0.05).
Curcumin promoted copper depletion by targeting ATOX1 in A549 and H1299 cells
Overexpression of ATOX1 in cells was obtained by pcDNA3.1-ATOX1 plasmid transfection. The effects of plasmid transfection on cell viability were observed (Fig. 2A), and there was no significance between the plasmid-transfected cells and the control cells. Observing the levels of ATOX1 protein in cells (Fig. 2B), no significance was found between the control cells and the plasmid control-transfected cells, but the pcDNA3.1-ATOX1 plasmid transfection significantly increased the levels of ATOX1 protein compared with the plasmid control transfection (Fig. 2C, p < 0.05). The levels of copper in cells were labeled by copper sensor-1 (Fig. 2D). It showed no significance between the control cells and the plasmid control-transfected cells, but the pcDNA3.1-ATOX1 plasmid transfection increased the levels of copper in cells compared with the plasmid control transfection (p < 0.05).
Based on the results of different doses of curcumin or DC-AC50 treatment on cell viability, 40 µM of curcumin or 20 µM of DC-AC50 were used for the following experiments. To confirm that curcumin targets ATOX1 to suppress copper accumulation in A549 and H1299 cells, the cells overexpressing ATOX1 were treated with 40 µM of curcumin or 20 µM of DC-AC50. The levels of copper in A549 (Fig. 3A) and H1299 (Fig. 3B) cells were suppressed after the curcumin or DC-AC50 treatment by comparison with the control cells (p < 0.01). However, the ATOX1 overexpression suppressed the roles of curcumin or DC-AC-50, which increased the levels of copper in cells compared with the curcumin or DC-AC50 treatment (p < 0.05).
Curcumin suppressed copper-associated proteins by targeting ATOX1 in A549 and H1299 cells
To confirm that curcumin suppresses cell viability by decreasing the ATOX1-associated proteins, the cell viability (Fig. 4A) and levels of ATP7A and COX17 proteins (Fig. 4B and C) were observed in the cell groups. The curcumin or DC-AC50 treatment significantly suppressed the cell viability compared with the control group (p < 0.01). However, the ATOX1 overexpression blocked the roles of curcumin or DC-AC50, which enhanced the cell viability compared with the curcumin or DC-AC50 treatment (p < 0.05). Meanwhile, the levels of ATP7A and COX17 proteins were downregulated after the curcumin or DC-AC50 treatment when compared to the control group (p < 0.01, Fig. 4C). However, the ATOX1 overexpression upregulated the levels of the above proteins compared with the curcumin or DC-AC50 treatment (p < 0.01).
Curcumin inhibited tumor growth by suppressing ATOX1
To reduce the number of mice, the A549 cells were used to establish animal models, and the animal models were treated with 20 mg/kg/d DC-AC50 or 40 mg/kg/d curcumin to observe the tumor growth. Through observing the tumor volume (Fig. 5A) and tumor weight (Fig. 5B), the DC-AC50 or curcumin treatment suppressed the tumor growth compared with the vehicle-treated control mice (p < 0.01). However, the tumor growth in the DC-AC50-treated mice was higher than that in the curcumin-treated mice (p < 0.05). Additionally, the levels of ATOX1 in tumors (Fig. 5C and D) were suppressed after the DC-AC50 or curcumin treatment when contrasted to the vehicle-treated control mice (p < 0.01).
Curcumin suppressed the levels of ATP7A and COX17 in tumors
The expressions of ATP7A and COX17 in tumors were also measured by immunohistochemistry (Fig. 6A and B) and western blotting (Fig. 6C). It showed that the DC-AC50 or curcumin treatment suppressed the expressions of ATP7A and COX17 when contrasted to the vehicle-treated control mice (p < 0.05). However, the expressions of COX17 in the tumors of DC-AC50-treated mice were lower than those in the tumors of curcumin-treated mice (p < 0.05).
Discussion
As one of the most lethal malignancies, NSCLC accounts for around 2 million new cases and 1.8 million deaths every year in the world [6]. Numerous studies have shown that curcumin delays the initiation and progression of NSCLC by affecting a wide range of molecular targets and cell signaling pathways, including inflammation, ROS, ferroptosis, autophagy, and so on [23, 24]. In this study, we further found that curcumin could bind ATOX1 to suppress the copper accumulation in the NSCLC cells, suggesting that inhibition of ATOX1 may be a molecular mechanism of curcumin for NSCLC treatment.
Numerous observations point to a requirement for higher levels of copper for tumors compared with healthy tissues [25]. Cancer cells have a higher demand for copper compared with non-dividing cells, due to the requirement for copper as a cofactor of mitochondrial cytochrome C oxidase, which is necessary to meet the energy demands of rapidly dividing cells [25]. Copper can promote tumor initiation, growth and metastasis by regulating mitochondrial function, copper pathways, autophagy, and so on [25, 26]. The ATOX1 is a copper chaperone protein that delivers copper to the export pumps ATP7A or ATP7B, permitting copper transport into the secretory pathway and copper export from the cell. Alternatively, ATOX1 can function as a copper-dependent transcription factor [2]. In NSCLC cells, ATOX1 plays an important role in copper-stimulated proliferation, and the knockdown of ATOX1 can inhibit the proliferation of NSCLC cells [18]. The copper pathway, ATOX1-ATP7A-lysyl oxidase (LOX) pathway, can promote tumor metastatic expansion in breast cancer cells [11]. In this study, we found that the levels of ATOX1 and ATP7A proteins in the NSCLC cells (A549 and H1299) were decreased with the increasing curcumin, indicating that the anti-cancer effect of curcumin is associated with the ATOX1-mediated copper pathway.
To confirm that curcumin targets ATOX1 to control the copper accumulation in NSCLC cells, ATOX1 overexpression in cells was established by the pcDNA3.1-ATOX1 plasmid transfection. Moreover, the ATOX1 inhibitor, DC-AC50 [20], was used as a positive control treatment to confirm the effects of curcumin treatment on the ATOX1-mediated copper pathway in NSCLC cells. The results showed that the curcumin suppressed the copper accumulation by inhibiting the ATOX1-AT7PA pathway in NSCLC cells. In lung carcinoma mice model, curcumin or DC-AC50 treatment also suppressed the tumor growth by suppressing the levels of ATOX1 and ATP7A in tumors, confirming that curcumin targets ATOX1 to suppress copper accumulation in NSCLC.
Moreover, the levels of COX17 in NSCLC cells were decreased with the increasing curcumin. The COX17 is a copper chaperone, that is matured in the mitochondrial intermembrane space and its activity is subject to the copper supply in mitochondria [27]. It has been found that COX17 is essential for cancer cell metabolism, including uncontrolled cancer cell proliferation, dysregulated metabolism, and invasion [28]. Inhibition of COX17 also suppressed the proliferation of NSCLC cells [29]. In this study, curcumin or DC-AC50 treatment also suppressed the COX17 expression in NSCLC cells and animal tumors. However, no significance was found in the COX17 expression between the curcumin treatment and DC-AC50 treatment in NSCLC cells, but significance was found in animal tumors. The significance between the curcumin treatment and DC-AC50 treatment in vivo might be induced by the regulation of curcumin on the tumor microenvironment (TME). Curcumin can regulate TME by inhibiting the growth of its cellular components, such as cancer-associated adipocytes, cancer-associated fibroblasts, and tumor endothelial cells [30]. Furthermore, curcumin can inhibit the interplay of tumor cells to TME by suppressing non-cellular components, such as extracellular matrix, and associated tumor-promoting signaling pathways [30].
However, more studies are necessary for curcumin before the commencement of clinical trials. The blood-brain barrier (BBB) permeability, synergistic combination treatment, and bioavailability are necessary on the pathway toward clinical implementation.
Conclusion
This study demonstrated that suppression of the ATOX1-associated copper pathway is a molecular mechanism of curcumin for NSCLC treatment. This study provides a new mechanism of curcumin for NSCLC treatment, enhancing the possibility of its clinical application.
Data availability
The datasets generated during the current study are available from the corresponding author.
References
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 U S A. 2019;116(14):6836–41.
Muller PA, Klomp LW. ATOX1: a novel copper-responsive transcription factor in mammals? Int J Biochem Cell Biol. 2009;41(6):1233–6.
Chen L, Min J, Wang F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct Target Ther. 2022;7(1):378.
Liu WQ, Lin WR, Yan L, et al. Copper homeostasis and cuproptosis in cancer immunity and therapy. Immunol Rev. 2024;321(1):211–27.
Hu Q, Wang R, Ma H, et al. Cuproptosis predicts the risk and clinical outcomes of lung adenocarcinoma. Front Oncol. 2022;12:922332.
Deng R, Zhu L, Jiang J et al. Cuproptosis-related gene LIPT1 as a prognostic indicator in non-small cell lung cancer: functional involvement and regulation of ATOX1 expression. Biomol Biomed. 2023.
Li Z, Feiyue Z, Gaofeng L. Traditional Chinese medicine and lung cancer–from theory to practice. Biomed Pharmacother. 2021;137:111381.
Tomeh MA, Hadianamrei R, Zhao X. A review of Curcumin and its derivatives as anticancer agents. Int J Mol Sci. 2019;20(5).
Liu Z, Ma H, Lai Z. The role of ferroptosis and cuproptosis in curcumin against hepatocellular carcinoma. Molecules. 2023;28(4).
Hu A, Zhang JW, Yang LY, et al. Curcumin-loaded graphene oxide quantum dots enhance otoprotective effects via blocking cuproptosis. Front Bioeng Biotechnol. 2023;11:1183197.
Blockhuys S, Zhang X, Wittung-Stafshede P. Single-cell tracking demonstrates copper chaperone Atox1 to be required for breast cancer cell migration. Proc Natl Acad Sci U S A. 2020;117(4):2014–9.
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.
Tian H, Li Y, Mei J, et al. Effects of Salvia miltiorrhiza extract on lung adenocarcinoma. Exp Ther Med. 2021;22(2):794.
Li P, Sun Q, Bai S et al. Combination of the cuproptosis inducer disulfiram and anti–PD–L1 abolishes NSCLC resistance by ATP7B to regulate the HIF–1 signaling pathway. Int J Mol Med. 2024;53(2).
Falls-Hubert KC, Butler AL, Gui K, et al. Disulfiram causes selective hypoxic cancer cell toxicity and radio-chemo-sensitization via redox cycling of copper. Free Radic Biol Med. 2020;150:1–11.
Jana A, Das A, Krett NL, et al. Nuclear translocation of Atox1 potentiates activin A-induced cell migration and colony formation in colon cancer. PLoS ONE. 2020;15(1):e0227916.
Blockhuys S, Hildesjö C, Olsson H et al. Evaluation of ATOX1 as a potential predictive biomarker for Tetrathiomolybdate treatment of breast Cancer patients with high risk of recurrence. Biomedicines. 2021;9(12).
Cai H, Peng F. Knockdown of copper chaperone antioxidant-1 by RNA interference inhibits copper-stimulated proliferation of non-small cell lung carcinoma cells. Oncol Rep. 2013;30(1):269–75.
Xu J, Yang L, Lin T. β-sitosterol targets glucocorticoid receptor to reduce airway inflammation and remodeling in allergic asthma. Pulm Pharmacol Ther. 2023;78:102183.
Wang J, Luo C, Shan C, et al. Inhibition of human copper trafficking by a small molecule significantly attenuates cancer cell proliferation. Nat Chem. 2015;7(12):968–79.
Kurien BT, Scofield RH. Western blotting. Methods. 2006;38(4):283–93.
Hussaini HM, Seo B, Rich AM. Immunohistochemistry and immunofluorescence. Methods Mol Biol. 2023;2588:439–50.
Tang C, Liu J, Yang C et al. Curcumin and its analogs in non-small cell lung cancer treatment: challenges and expectations. Biomolecules. 2022;12(11).
Salehi M, Movahedpour A, Tayarani A, et al. Therapeutic potentials of curcumin in the treatment of non-small-cell lung carcinoma. Phytother Res. 2020;34(10):2557–76.
Ge EJ, Bush AI, Casini A, et al. Connecting copper and cancer: from transition metal signalling to metalloplasia. Nat Rev Cancer. 2022;22(2):102–13.
Zhao Q, Qi T. The implications and prospect of cuproptosis-related genes and copper transporters in cancer progression. Front Oncol. 2023;13:1117164.
Cui L, Gouw AM, LaGory EL, et al. Mitochondrial copper depletion suppresses triple-negative breast cancer in mice. Nat Biotechnol. 2021;39(3):357–67.
Xue Q, Kang R, Klionsky DJ, et al. Copper metabolism in cell death and autophagy. Autophagy. 2023;19(8):2175–95.
Suzuki C, Daigo Y, Kikuchi T, et al. Identification of COX17 as a therapeutic target for non-small cell lung cancer. Cancer Res. 2003;63(21):7038–41.
Prasad S, Saha P, Chatterjee B, et al. Complexity of tumor microenvironment: therapeutic role of curcumin and its metabolites. Nutr Cancer. 2023;75(1):1–13.
Funding
This study was founded by the Qilu Traditional Chinese Medicine Advantageous Specialty Cluster Construction Project (Luwei Letter No. 202246) and the Project of Yantai Science and Technology Plan (No.2023YD016).
Author information
Authors and Affiliations
Contributions
X.Q. and W.T.S. participated in the study design. P.W. and H.Y.L. participated in the experimental work and data collection. X.Q. and H.Y.L. participated in the data analysis. P.W. partook in the manuscript preparation. X.Q. wrote the manuscript. All authors read and agreed to the final manuscript.
Corresponding author
Ethics declarations
Ethical approval
This study was approved by the Animal Care and Ethics Committee of Yantai Traditional Chinese Medicine Hospital (No. 2023-KY-033). All experimental procedures were carried out in accordance with the National Institutes of Health guide for the care and use of laboratory animals (NIH Publications No. 8023, revised 1978), and comply with the ARRIVE guidelines.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Electronic supplementary material
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Qin, X., Wang, P., Liang, H. et al. Curcumin suppresses copper accumulation in non-small cell lung cancer by binding ATOX1. BMC Pharmacol Toxicol 25, 54 (2024). https://doi.org/10.1186/s40360-024-00784-0
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s40360-024-00784-0