Copper homeostasis and cuproptosis in health and disease

Yunuo Yang , Jiaxuan Wu , Lisheng Wang , Guang Ji , Yanqi Dang

MedComm ›› 2024, Vol. 5 ›› Issue (10) : e724

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (10) : e724 DOI: 10.1002/mco2.724
REVIEW

Copper homeostasis and cuproptosis in health and disease

Author information +
History +
PDF

Abstract

Copper is a vital trace element in human physiology, essential for the synthesis of numerous crucial metabolic enzymes and facilitation of various biological processes. Regulation of copper levels within a narrow range is imperative for maintaining metabolic homeostasis. Numerous studies have demonstrated the significant roles of copper homeostasis and cuproptosis in health and disease pathogenesis. However, a comprehensive and up-to-date systematic review in this domain remains absent. This review aims to consolidate recent advancements in understanding the roles of cuproptosis and copper homeostasis in health and disease, focusing on the underlying mechanisms and potential therapeutic interventions. Dysregulation of copper homeostasis, manifesting as either copper excess or deficiency, is implicated in the etiology of various diseases. Cuproptosis, a recently identified form of cell death, is characterized by intracellular copper overload. This phenomenon mediates a diverse array of evolutionary processes in organisms, spanning from health to disease, and is implicated in genetic disorders, liver diseases, neurodegenerative disorders, and various cancers. This review provides a comprehensive summary of the pathogenic mechanisms underlying cuproptosis and copper homeostasis, along with associated targeted therapeutic agents. Furthermore, it explores future research directions with the potential to yield significant advancements in disease treatment, health management, and disease prevention.

Keywords

cancer / cardiovascular disease / copper homeostasis / cuproptosis / hereditary disease / liver disease / neurodegenerative disease

Cite this article

Download citation ▾
Yunuo Yang, Jiaxuan Wu, Lisheng Wang, Guang Ji, Yanqi Dang. Copper homeostasis and cuproptosis in health and disease. MedComm, 2024, 5(10): e724 DOI:10.1002/mco2.724

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Gao L, Zhang A. Copper-instigated modulatory cell mortality mechanisms and progress in oncological treatment investigations. Front Immunol. 2023; 14: 1236063.

[2]

Blades B, Ayton S, Hung YH, et al. Copper and lipid metabolism: a reciprocal relationship. Biochim Biophys Acta Gen Subj. 2021; 1865(11): 129979.

[3]

Guan D, Zhao L, Shi X, et al. Copper in cancer: From pathogenesis to therapy. Biomed Pharmacother. 2023; 163: 114791.

[4]

Liu T, Liu Y, Zhang F, Gao Y. Copper homeostasis dysregulation promoting cell damage and the association with liver diseases. Chin Med J (Engl). 2023; 136(14): 1653-1662.

[5]

Yang S, Li Y, Zhou L, et al. Copper homeostasis and cuproptosis in atherosclerosis: metabolism, mechanisms and potential therapeutic strategies. Cell Death Discov. 2024; 10(1): 25.

[6]

Kim BE, Nevitt T, Thiele DJ. Mechanisms for copper acquisition, distribution and regulation. Nat Chem Biol. 2008; 4(3): 176-185.

[7]

Ruiz LM, Libedinsky A, Elorza AA. Role of copper on mitochondrial function and metabolism. Front Mol Biosci. 2021; 8: 711227.

[8]

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.

[9]

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.

[10]

Aigner E, Weiss G, Datz C. Dysregulation of iron and copper homeostasis in nonalcoholic fatty liver. World J Hepatol. 2015; 7(2): 177-188.

[11]

Arredondo M, Núñez MT. Iron and copper metabolism. Mol Aspects Med. 2005; 26(4-5): 313-327.

[12]

Wazir SM, Ghobrial I. Copper deficiency, a new triad: anemia, leucopenia, and myeloneuropathy. J Community Hosp Intern Med Perspect. 2017; 7(4): 265-268.

[13]

Hegazy AA, Zaher MM, Abd El-Hafez MA, et al. Relation between anemia and blood levels of lead, copper, zinc and iron among children. BMC Res Notes. 2010; 3: 133.

[14]

Collins JF, Prohaska JR, Knutson MD. Metabolic crossroads of iron and copper. Nutr Rev. 2010; 68(3): 133-147.

[15]

Arredondo M, Martínez R, Núñez MT, et al. Inhibition of iron and copper uptake by iron, copper and zinc. Biol Res. 2006; 39(1): 95-102.

[16]

Crowe A, Morgan EH. Iron and copper interact during their uptake and deposition in the brain and other organs of developing rats exposed to dietary excess of the two metals. J Nutr. 1996; 126(1): 183-194.

[17]

Reeves PG, DeMars LC. Copper deficiency reduces iron absorption and biological half-life in male rats. J Nutr. 2004; 134(8): 1953-1957.

[18]

Arredondo M, Cambiazo V, Tapia L, et al. Copper overload affects copper and iron metabolism in Hep-G2 cells. Am J Physiol Gastrointest Liver Physiol. 2004; 287(1): G27-G32.

[19]

Shawki A, Anthony SR, Nose Y, et al. Intestinal DMT1 is critical for iron absorption in the mouse but is not required for the absorption of copper or manganese. Am J Physiol Gastrointest Liver Physiol. 2015; 309(8): G635-G647.

[20]

Chen L, Min J, Wang F. Copper homeostasis and cuproptosis in health and disease. Signal Transduct Target Ther. 2022; 7(1): 378.

[21]

Linder MC. Copper homeostasis in mammals, with emphasis on secretion and excretion. A review. Int J Mol Sci. 2020; 21(14): 4932.

[22]

Taylor AA, Tsuji JS, Garry MR, et al. Critical review of exposure and effects: implications for setting regulatory health criteria for ingested copper. Environ Manage. 2020; 65(1): 131-159.

[23]

Tsvetkov PA-O, Coy SA-O, Petrova BA-O, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022; 375(6586): 1254-1261.

[24]

Gusti AMT, Qusti SY, Alshammari EM, et al. Antioxidants-related superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPX), glutathione-S-transferase (GST), and nitric oxide synthase (NOS) gene variants analysis in an obese population: a preliminary case-control study. Antioxidants (Basel). 2021; 10(4): 595.

[25]

Zischka H, Einer C. Mitochondrial copper homeostasis and its derailment in Wilson disease. Int J Biochem Cell Biol. 2018; 102: 71-75.

[26]

Trasnea PI, Utz M, Khalfaoui-Hassani B, et al. Cooperation between two periplasmic copper chaperones is required for full activity of the cbb3 - type cytochrome c oxidase and copper homeostasis in Rhodobacter capsulatus. Mol Microbiol. 2016; 100(2): 345-361.

[27]

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.

[28]

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.

[29]

Baker ZN, Cobine PA, Leary SC. The mitochondrion: a central architect of copper homeostasis. Metallomics. 2017; 9(11): 1501-1512.

[30]

Cox TR, Gartland A, Erler JT. Lysyl oxidase, a targetable secreted molecule involved in cancer metastasis. Cancer Res. 2016; 76(2): 188-192.

[31]

Chen W, Yang A, Jia J, et al. Lysyl oxidase (LOX) family members: rationale and their potential as therapeutic targets for liver fibrosis. Hepatology. 2020; 72(2): 729-741.

[32]

Johnston KA, Lopez KM. Lysyl oxidase in cancer inhibition and metastasis. Cancer Lett. 2018; 417: 174-181.

[33]

Al-U’datt D, Allen BG, Nattel S. Role of the lysyl oxidase enzyme family in cardiac function and disease. Cardiovasc Res. 2019; 115(13): 1820-1837.

[34]

Timón-Gómez A, Nývltová E, Abriata LA, et al. Mitochondrial cytochrome c oxidase biogenesis: recent developments. Semin Cell Dev Biol. 2018; 76: 163-178.

[35]

Ramzan R, Napiwotzki J, Weber P, et al. Cholate disrupts regulatory functions of cytochrome c oxidase. Cells. 2021; 10(7): 1579.

[36]

Garza NM, Griffin AT, Zulkifli M, et al. A genome-wide copper-sensitized screen identifies novel regulators of mitochondrial cytochrome c oxidase activity. J Biol Chem. 2021; 296: 100485.

[37]

Banks CJ, Andersen JL. Mechanisms of SOD1 regulation by post-translational modifications. Redox Biol. 2019; 26: 101270.

[38]

Tsang CK, Liu Y, Thomas J, et al. Superoxide dismutase 1 acts as a nuclear transcription factor to regulate oxidative stress resistance. Nat Commun. 2014; 5: 3446.

[39]

Skórska KB, Płaczkowska S, Prescha A, et al. Serum total SOD activity and SOD1/2 concentrations in predicting all-cause mortality in lung cancer patients. Pharmaceuticals (Basel). 2021; 14(11): 1067.

[40]

Zhao G, Sun H, Zhang T, Liu JX. Copper induce zebrafish retinal developmental defects via triggering stresses and apoptosis. Cell Commun Signal. 2020; 18(1): 45.

[41]

Martínez-Reyes I, Chandel NS. Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun. 2020; 11(1): 102.

[42]

Tsvetkov P, Coy S, Petrova B, et al. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science. 2022; 375(6586): 1254-1261.

[43]

Tian Z, Jiang S, Zhou J, Zhang W. Copper homeostasis and cuproptosis in mitochondria. Life Sci. 2023; 334: 122223.

[44]

Li SR, Bu LL, Cai L. Cuproptosis: lipoylated TCA cycle proteins-mediated novel cell death pathway. Signal Transduct Target Ther. 2022; 7(1): 158.

[45]

Qin Y, Liu Y, Xiang X, et al. Cuproptosis correlates with immunosuppressive tumor microenvironment based on pan-cancer multiomics and single-cell sequencing analysis. Mol Cancer. 2023; 22(1): 59.

[46]

Dekker E, Tanis PJ, Vleugels JLA, et al. Colorectal cancer. Lancet. 2019; 394(10207): 1467-1480.

[47]

Patel SG, Karlitz JJ, Yen T, et al. The rising tide of early-onset colorectal cancer: a comprehensive review of epidemiology, clinical features, biology, risk factors, prevention, and early detection. Lancet Gastroenterol Hepatol. 2022; 7(3): 262-274.

[48]

Baszuk P, Marciniak W, Derkacz R, et al. Blood copper levels and the occurrence of colorectal cancer in Poland. Biomedicines. 2021; 9(11): 1628.

[49]

Popovic D, Vucic D, Dikic I. Ubiquitination in disease pathogenesis and treatment. Nat Med. 2014; 20(11): 1242-1253.

[50]

Squitti R, Pal A, Dhar A, et al. Serum copper status of patients with colorectal cancer: a systematic review and meta-analysis. J Trace Elem Med Biol. 2024; 82: 127370.

[51]

Ruiz MC, Perelmulter K, Levín P, et al. Antiproliferative activity of two copper (II) complexes on colorectal cancer cell models: Impact on ROS production, apoptosis induction and NF-κB inhibition. Eur J Pharm Sci. 2022; 169: 106092.

[52]

Shao Y, Fan X, Yang X, et al. Impact of Cuproptosis-related markers on clinical status, tumor immune microenvironment and immunotherapy in colorectal cancer: a multi-omic analysis. Comput Struct Biotechnol J. 2023; 21: 3383-3403.

[53]

Xu L, Wu P, Rong A, et al. Systematic pan-cancer analysis identifies cuproptosis-related gene DLAT as an immunological and prognostic biomarker. Aging (Albany NY). 2023; 15(10): 4269-4287.

[54]

Li L, Sun F, Kong F, et al. Characterization of a cuproptosis-related signature to evaluate immune features and predict prognosis in colorectal cancer. Front Oncol. 2023; 13: 1083956.

[55]

Gong X, Wu Q, Tan Z, et al. Identification and validation of cuproptosis and disulfidptosis related genes in colorectal cancer. Cell Signal. 2024; 119: 111185.

[56]

Zhu Z, Zhao Q, Song W, et al. A novel cuproptosis-related molecular pattern and its tumor microenvironment characterization in colorectal cancer. Front Immunol. 2022; 13: 940774.

[57]

Wang L, Cao Y, Guo W, Xu J. High expression of cuproptosis-related gene FDX1 in relation to good prognosis and immune cells infiltration in colon adenocarcinoma (COAD). J Cancer Res Clin Oncol. 2023; 149(1): 15-24.

[58]

Wang C, Guo J, Zhang Y, et al. Cuproptosis-related gene FDX1 suppresses the growth and progression of colorectal cancer by retarding EMT progress. Biochem Genet. 2024.

[59]

Wu Z, Lin C, Zhang F, et al. TIGD1 function as a potential cuproptosis regulator following a novel cuproptosis-related gene risk signature in colorectal cancer. Cancers (Basel). 2023; 15(8): 2286.

[60]

Yang W, Wang Y, Huang Y, et al. 4-Octyl itaconate inhibits aerobic glycolysis by targeting GAPDH to promote cuproptosis in colorectal cancer. Biomed Pharmacother. 2023; 159: 114301.

[61]

Zhu Z, Guo T, Weng J, et al. Cuproptosis-related miRNAs signature and immune infiltration characteristics in colorectal cancer. Cancer Med. 2023; 12(15): 16661-16678.

[62]

Shi X, Li Y, Jia M, et al. A novel copper chelator for the suppression of colorectal cancer. Drug Dev Res. 2023; 84(2): 312-325.

[63]

Barzaman K, Karami J, Zarei Z, et al. Breast cancer: biology, biomarkers, and treatments. Int Immunopharmacol. 2020; 84: 106535.

[64]

Britt KL, Cuzick J, Phillips KA. Key steps for effective breast cancer prevention. Nat Rev Cancer. 2020; 20(8): 417-436.

[65]

Sha S, Si L, Wu X, et al. Prognostic analysis of cuproptosis-related gene in triple-negative breast cancer. Front Immunol. 2022; 13: 922780.

[66]

Wu X, Zhang Y, Liang G, Ye H. Cuproptosis-related lncRNAs potentially predict prognosis and therapy sensitivity of breast cancer. Front Pharmacol. 2023; 14: 1199883.

[67]

Wang R, Xu K, Chen Q, et al. Cuproptosis engages in c-Myc-mediated breast cancer stemness. J Transl Med. 2023; 21(1): 409.

[68]

Qi Y, Yao Q, Li X, et al. Cuproptosis-related gene SLC31A1: prognosis values and potential biological functions in cancer. Sci Rep. 2023; 13(1): 17790.

[69]

Ning S, Lyu M, Zhu D, et al. Type-I AIE photosensitizer loaded biomimetic system boosting cuproptosis to inhibit breast cancer metastasis and rechallenge. ACS Nano. 2023; 17(11): 10206-10217.

[70]

Xiong C, Ling H, Hao Q, Zhou X. Cuproptosis: p53-regulated metabolic cell death? Cell Death Differ. 2023; 30(4): 876-884.

[71]

Wu JH, Cheng TC, Zhu B, et al. Identification of cuproptosis-related gene SLC31A1 and upstream LncRNA-miRNA regulatory axis in breast cancer. Sci Rep. 2023; 13(1): 18390.

[72]

Liu Y, Wang J, Jiang M. Copper-related genes predict prognosis and characteristics of breast cancer. Front Immunol. 2023; 14: 1145080.

[73]

Sha R, Dong X, Yan S, et al. Cuproptosis-related genes predict prognosis and trastuzumab therapeutic response in HER2-positive breast cancer. Sci Rep. 2024; 14(1): 2908.

[74]

Yang X, Deng L, Diao X, et al. Targeting cuproptosis by zinc pyrithione in triple-negative breast cancer. iScience. 2023; 26(11): 108218.

[75]

Wang C, Tan S, Li J, et al. CircRNAs in lung cancer - Biogenesis, function and clinical implication. Cancer Lett. 2020; 492: 106-115.

[76]

The L. Lung cancer: some progress, but still a lot more to do. Lancet. 2019; 394(10212): 1880.

[77]

Thai AA, Solomon BJ, Sequist LV, et al. Lung cancer. Lancet. 2021; 398(10299): 535-554.

[78]

Zhang W, Qu H, Ma X, et al. Identification of cuproptosis and immune-related gene prognostic signature in lung adenocarcinoma. Front Immunol. 2023; 14: 1179742.

[79]

Chen Y, Tang L, Huang W, et al. Identification and validation of a novel cuproptosis-related signature as a prognostic model for lung adenocarcinoma. Front Endocrinol (Lausanne). 2022; 13: 963220.

[80]

Xiaona X, Liu Q, Zhou X, et al. Comprehensive analysis of cuproptosis-related genes in immune infiltration and prognosis in lung adenocarcinoma. Comput Biol Med. 2023; 158: 106831.

[81]

Yang J, Liu K, Yang L, et al. Identification and validation of a novel cuproptosis-related stemness signature to predict prognosis and immune landscape in lung adenocarcinoma by integrating single-cell and bulk RNA-sequencing. Front Immunol. 2023; 14: 1174762.

[82]

Wang X, Jing H, Li H. A novel cuproptosis-related lncRNA signature to predict prognosis and immune landscape of lung adenocarcinoma. Transl Lung Cancer Res. 2023; 12(2): 230-246.

[83]

Ma C, Li F, He Z, et al. Prognosis and personalized treatment prediction in lung adenocarcinoma: an in silico and in vitro strategy adopting cuproptosis related lncRNA towards precision oncology. Front Pharmacol. 2023; 14: 1113808.

[84]

Deng L, Jiang A, Zeng H, et al. Comprehensive analyses of PDHA1 that serves as a predictive biomarker for immunotherapy response in cancer. Front Pharmacol. 2022; 13: 947372.

[85]

Tang Y, Wang T, Li Q, Shi J. A cuproptosis score model and prognostic score model can evaluate clinical characteristics and immune microenvironment in NSCLC. Cancer Cell Int. 2024; 24(1): 68.

[86]

Wang W, Lu K, Jiang X, et al. Ferroptosis inducers enhanced cuproptosis induced by copper ionophores in primary liver cancer. J Exp Clin Cancer Res. 2023; 42(1): 142.

[87]

Li A, Huang K, Pan W, et al. Thiosemicarbazone mixed-valence Cu(I/II) complex against lung adenocarcinoma cells through multiple pathways involving cuproptosis. J Med Chem. 2024; 67(11): 9091-9103.

[88]

Tang W, Wu J, Wang L, et al. Bioactive layered double hydroxides for synergistic sonodynamic/cuproptosis anticancer therapy with elicitation of the immune response. ACS Nano. 2024; 18(15): 10495-10508.

[89]

Stoffel EM, Brand RE, Goggins M. Pancreatic cancer: changing epidemiology and new approaches to risk assessment, early detection, and prevention. Gastroenterology. 2023; 164(5): 752-765.

[90]

Klein AP. Pancreatic cancer epidemiology: understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol. 2021; 18(7): 493-502.

[91]

Yao HF, Xu DP, Zheng JH, et al. Analysis of cuproptosis-related lncRNA signature for predicting prognosis and tumor immune microenvironment in pancreatic cancer. Apoptosis. 2023; 28(7-8): 1090-1112.

[92]

Sun Y, Yao L, Man C, et al. Development and validation of cuproptosis-related lncRNAs associated with pancreatic cancer immune microenvironment based on single-cell. Front Immunol. 2023; 14: 1220760.

[93]

Chen L, Zhang L, He H, et al. Systemic analyses of cuproptosis-related lncRNAs in pancreatic adenocarcinoma, with a focus on the molecular mechanism of LINC00853. Int J Mol Sci. 2023; 24(9): 7923.

[94]

Zhang X, Zhou Y, Hu J, et al. Comprehensive analysis identifies cuproptosis-related gene DLAT as a potential prognostic and immunological biomarker in pancreatic adenocarcinoma. BMC Cancer. 2023; 23(1): 560.

[95]

Yu Q, Zhou J, Liu Y, et al. DNAzyme-mediated cascade nanoreactor for cuproptosis-promoted pancreatic cancer synergistic therapy. Adv Healthc Mater. 2023; 12(28): e2301429.

[96]

Hu F, Huang J, Bing T, et al. Stimulus-responsive copper complex nanoparticles induce cuproptosis for augmented cancer immunotherapy. Adv Sci (Weinh). 2024; 11(13): e2309388.

[97]

Yu N, Zhou J, Ding M, et al. Sono-triggered cascade lactate depletion by semiconducting polymer nanoreactors for cuproptosis-immunotherapy of pancreatic cancer. Angew Chem Int Ed Engl. 2024; 63(30): e202405639.

[98]

Xiao C, Li J, Hua A, et al. Hyperbaric oxygen boosts antitumor efficacy of copper-diethyldithiocarbamate nanoparticles against pancreatic ductal adenocarcinoma by regulating cancer stem cell metabolism. Research (Wash D C). 2024; 7: 0335.

[99]

Bandmann O, Weiss KH, Kaler SG. Wilson’s disease and other neurological copper disorders. Lancet Neurol. 2015; 14(1): 103-113.

[100]

Wang Q, Sun J, Chen T, et al. Ferroptosis, pyroptosis, and cuproptosis in Alzheimer’s disease. ACS Chem Neurosci. 2023; 14(19): 3564-3587.

[101]

Scheltens P, De Strooper B, Kivipelto M, et al. Alzheimer’s disease. Lancet. 2021; 397(10284): 1577-1590.

[102]

Ma C, Hong F, Yang S. Amyloidosis in Alzheimer’s disease: pathogeny, etiology, and related therapeutic directions. Molecules. 2022; 27(4): 1210.

[103]

Chen LL, Fan YG, Zhao LX, et al. The metal ion hypothesis of Alzheimer’s disease and the anti-neuroinflammatory effect of metal chelators. Bioorg Chem. 2023; 131: 106301.

[104]

Tiwari S, Atluri V, Kaushik A, et al. Alzheimer’s disease: pathogenesis, diagnostics, and therapeutics. Int J Nanomedicine. 2019; 14: 5541-5554.

[105]

Li K, Li A, Mei Y, et al. Trace elements and Alzheimer dementia in population-based studies: a bibliometric and meta-analysis. Environ Pollut. 2023; 318: 120782.

[106]

Squitti R, Ventriglia M, Simonelli I, et al. Copper imbalance in Alzheimer’s disease: meta-analysis of serum, plasma, and brain specimens, and replication study evaluating ATP7B gene variants. Biomolecules. 2021; 11(7): 960.

[107]

Singh SK, Balendra V, Obaid AA, et al. Copper-mediated β-amyloid toxicity and its chelation therapy in Alzheimer’s disease. Metallomics. 2022; 14(6): mfac018.

[108]

Regland B, Lehmann W, Abedini I, et al. Treatment of Alzheimer’s disease with clioquinol. Dement Geriatr Cogn Disord. 2001; 12(6): 408-414.

[109]

Al-Chalabi A, Hardiman O. The epidemiology of ALS: a conspiracy of genes, environment and time. Nat Rev Neurol. 2013; 9(11): 617-628.

[110]

Feldman EL, Goutman SA, Petri S, et al. Amyotrophic lateral sclerosis. Lancet. 2022; 400(10360): 1363-1380.

[111]

Tafuri F, Ronchi D, Magri F, et al. SOD1 misplacing and mitochondrial dysfunction in amyotrophic lateral sclerosis pathogenesis. Front Cell Neurosci. 2015; 9: 336.

[112]

Tokuda E, Okawa E, Watanabe S, et al. Dysregulation of intracellular copper homeostasis is common to transgenic mice expressing human mutant superoxide dismutase-1s regardless of their copper-binding abilities. Neurobiol Dis. 2013; 54: 308-319.

[113]

Chen QY, Wu P, Wen T, et al. Association of cerebral spinal fluid copper imbalance in amyotrophic lateral sclerosis. Front Aging Neurosci. 2022; 14: 970711.

[114]

Furukawa Y, O’Halloran TV. Posttranslational modifications in Cu, Zn-superoxide dismutase and mutations associated with amyotrophic lateral sclerosis. Antioxid Redox Signal. 2006; 8(5-6): 847-867.

[115]

Swindell WR, Kruse CPS, List EO, et al. ALS blood expression profiling identifies new biomarkers, patient subgroups, and evidence for neutrophilia and hypoxia. J Transl Med. 2019; 17(1): 170.

[116]

Jia F, Zhang B, Yu W, et al. Exploring the cuproptosis-related molecular clusters in the peripheral blood of patients with amyotrophic lateral sclerosis. Comput Biol Med. 2024; 168: 107776.

[117]

Solovyev N, Lucio M, Mandrioli J, et al. Interplay of Metallome and Metabolome in Amyotrophic Lateral Sclerosis: A Study on Cerebrospinal Fluid of Patients Carrying Disease-Related Gene Mutations. ACS Chem Neurosci. 2023; 14(17): 3035-3046.

[118]

Andreassen OA, Dedeoglu A, Friedlich A, et al. Effects of an inhibitor of poly(ADP-ribose) polymerase, desmethylselegiline, trientine, and lipoic acid in transgenic ALS mice. Exp Neurol. 2001; 168(2): 419-424.

[119]

Nagano S, Fujii Y, Yamamoto T, et al. The efficacy of trientine or ascorbate alone compared to that of the combined treatment with these two agents in familial amyotrophic lateral sclerosis model mice. Exp Neurol. 2003; 179(2): 176-180.

[120]

LeBlanc A, Cuperlovic-Culf M, Morin PJ, Touaibia M. Structurally Related Edaravone Analogues: Synthesis, Antiradical, Antioxidant, and Copper-Chelating Properties. CNS Neurol Disord Drug Targets. 2019; 18(10): 779-790.

[121]

Xiao G, Fan Q, Wang X, Zhou B. Huntington disease arises from a combinatory toxicity of polyglutamine and copper binding. Proc Natl Acad Sci U S A. 2013; 110(37): 14995-5000.

[122]

Tabrizi SJ, Estevez-Fraga C, van Roon-Mom WMC, et al. Potential disease-modifying therapies for Huntington’s disease: lessons learned and future opportunities. Lancet Neurol. 2022; 21(7): 645-658.

[123]

Lobato AG, Ortiz-Vega N, Zhu Y, et al. Copper enhances aggregational toxicity of mutant huntingtin in a Drosophila model of Huntington’s Disease. Biochim Biophys Acta Mol Basis Dis. 2024; 1870(1): 166928.

[124]

Cordeiro LM, Soares MV, da Silva AF, et al. Toxicity of copper and zinc alone and in combination in Caenorhabditis elegans model of Huntington’s disease and protective effects of rutin. Neurotoxicology. 2023; 97: 120-132.

[125]

Pfalzer AC, Yan Y, Kang H, et al. Alterations in metal homeostasis occur prior to canonical markers in Huntington disease. Sci Rep. 2022; 12(1): 10373.

[126]

Scholefield M, Unwin RD, Cooper GJS. Shared perturbations in the metallome and metabolome of Alzheimer’s, Parkinson’s, Huntington’s, and dementia with Lewy bodies: a systematic review. Ageing Res Rev. 2020; 63: 101152.

[127]

Martínez-Lazcano JC, Montes S, Sánchez-Mendoza MA, et al. Sub-chronic copper pretreatment reduces oxidative damage in an experimental Huntington’s disease model. Biol Trace Elem Res. 2014; 162(1-3): 211-218.

[128]

Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021; 71(3): 209-249.

[129]

EASL clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol. 2018; 69(1): 182-236.

[130]

Akinyemiju T, Abera S, Ahmed M, et al. The burden of primary liver cancer and underlying etiologies from 1990 to 2015 at the global, regional, and national level: results from the global burden of disease study 2015. JAMA Oncol. 2017; 3(12): 1683-1691.

[131]

Shah PA, Patil R, Harrison SA. NAFLD-related hepatocellular carcinoma: the growing challenge. Hepatology. 2023; 77(1): 323-338.

[132]

Tamai Y, Iwasa M, Eguchi A, et al. Serum copper, zinc and metallothionein serve as potential biomarkers for hepatocellular carcinoma. PLoS One. 2020; 15(8): e0237370.

[133]

Qin H, Sheng W, Zhang G, et al. Comprehensive analysis of cuproptosis-related prognostic gene signature and tumor immune microenvironment in HCC. Front Genet. 2023; 14: 1094793.

[134]

Wang Y, Zhang Y, Wang L, et al. Development and experimental verification of a prognosis model for cuproptosis-related subtypes in HCC. Hepatol Int. 2022; 16(6): 1435-1447.

[135]

Li Z, Zhou H, Zhai X, et al. MELK promotes HCC carcinogenesis through modulating cuproptosis-related gene DLAT-mediated mitochondrial function. Cell Death Dis. 2023; 14(11): 733.

[136]

Zhang Z, Zeng X, Wu Y, et al. Cuproptosis-related risk score predicts prognosis and characterizes the tumor microenvironment in hepatocellular carcinoma. Front Immunol. 2022; 13: 925618.

[137]

Yan C, Niu Y, Ma L, et al. System analysis based on the cuproptosis-related genes identifies LIPT1 as a novel therapy target for liver hepatocellular carcinoma. J Transl Med. 2022; 20(1): 452.

[138]

Wang XX, Wu LH, Ji H, et al. A novel cuproptosis-related prognostic signature and potential value in HCC immunotherapy. Front Mol Biosci. 2022; 9: 1001788.

[139]

Chen Y, Tang L, Huang W, et al. Identification of a prognostic cuproptosis-related signature in hepatocellular carcinoma. Biol Direct. 2023; 18(1): 4.

[140]

Wang X, Chen D, Shi Y, et al. Copper and cuproptosis-related genes in hepatocellular carcinoma: therapeutic biomarkers targeting tumor immune microenvironment and immune checkpoints. Front Immunol. 2023; 14: 1123231.

[141]

Chen S, Liu P, Zhao L, et al. A novel cuproptosis-related prognostic lncRNA signature for predicting immune and drug therapy response in hepatocellular carcinoma. Front Immunol. 2022; 13: 954653.

[142]

Forner A, Reig M, Bruix J. Hepatocellular carcinoma. Lancet. 2018; 391(10127): 1301-1314.

[143]

Guo B, Yang F, Zhang L, et al. Cuproptosis induced by ros responsive nanoparticles with elesclomol and copper combined with αPD-L1 for enhanced cancer immunotherapy. Adv Mater. 2023; 35(22): e2212267.

[144]

Moawad AW, Szklaruk J, Lall C, et al. Angiogenesis in hepatocellular carcinoma; pathophysiology, targeted therapy, and role of imaging. J Hepatocell Carcinoma. 2020; 7: 77-89.

[145]

Fodor D, Jung I, Turdean S, et al. Angiogenesis of hepatocellular carcinoma: an immunohistochemistry study. World J Hepatol. 2019; 11(3): 294-304.

[146]

Yoshii J, Yoshiji H, Kuriyama S, et al. The copper-chelating agent, trientine, suppresses tumor development and angiogenesis in the murine hepatocellular carcinoma cells. Int J Cancer. 2001; 94(6): 768-773.

[147]

Liu Z, Ma H, Lai Z. The role of ferroptosis and cuproptosis in curcumin against hepatocellular carcinoma. Molecules. 2023; 28(4): 1623.

[148]

Xu S, Dong K, Gao R, et al. Cuproptosis-related signature for clinical prognosis and immunotherapy sensitivity in hepatocellular carcinoma. J Cancer Res Clin Oncol. 2023; 149(13): 12249-12263.

[149]

Paschos P, Paletas K. Non alcoholic fatty liver disease and metabolic syndrome. Hippokratia. 2009; 13(1): 9-19.

[150]

Guo X, Yin X, Liu Z, Wang J. Non-alcoholic fatty liver disease (NAFLD) pathogenesis and natural products for prevention and treatment. Int J Mol Sci. 2022; 23(24): 15489.

[151]

Godoy-Matos AF, Silva Junior WS, Valerio CM. NAFLD as a continuum: from obesity to metabolic syndrome and diabetes. Diabetol Metab Syndr. 2020; 12: 60.

[152]

Younossi ZM. Non-alcoholic fatty liver disease - a global public health perspective. J Hepatol. 2019; 70(3): 531-544.

[153]

Cotter TG, Rinella M. Nonalcoholic fatty liver disease 2020: the state of the disease. Gastroenterology. 2020; 158(7): 1851-1864.

[154]

Porcu C, Antonucci L, Barbaro B, et al. Copper/MYC/CTR1 interplay: a dangerous relationship in hepatocellular carcinoma. Oncotarget. 2018; 9(10): 9325-9343.

[155]

Wu C, Liu X, Zhong L, et al. Identification of cuproptosis-related genes in nonalcoholic fatty liver disease. Oxid Med Cell Longev. 2023; 2023: 9245667.

[156]

Ouyang G, Wu Z, Liu Z, et al. Identification and validation of potential diagnostic signature and immune cell infiltration for NAFLD based on cuproptosis-related genes by bioinformatics analysis and machine learning. Front Immunol. 2023; 14: 1251750.

[157]

Zhao Z, Luan T, Wan J, et al. Elucidating cuproptosis-associated genes in the progression from nash to HCC using bulk and single-cell RNA sequencing analyses and experimental validation. Medicina (Kaunas). 2023; 59(9): 1639.

[158]

Zhong CC, Zhao T, Hogstrand C, et al. Copper (Cu) induced changes of lipid metabolism through oxidative stress-mediated autophagy and Nrf2/PPARgamma pathways. J Nutr Biochem. 2022; 100: 108883.

[159]

Polvani S, Tarocchi M, Galli A. PPARgamma and oxidative stress: con(beta) catenating NRF2 and FOXO. PPAR Res. 2012; 2012: 641087.

[160]

Zhong CC, Zhao T, Hogstrand C, et al. Copper (Cu) induced changes of lipid metabolism through oxidative stress-mediated autophagy and Nrf2/PPARγ pathways. J Nutr Biochem. 2022; 100: 108883.

[161]

Mota de Sa P, Richard AJ, Hang H, Stephens JM. Transcriptional regulation of adipogenesis. Compr Physiol. 2017; 7(2): 635-674.

[162]

Horn N, Møller LB, Nurchi VM, Aaseth J. Chelating principles in Menkes and Wilson diseases: choosing the right compounds in the right combinations at the right time. J Inorg Biochem. 2019; 190: 98-112.

[163]

Członkowska A, Litwin T, Dusek P, et al. Wilson disease. Nat Rev Dis Primers. 2018; 4(1): 21.

[164]

Shribman S, Marjot T, Sharif A, et al. Investigation and management of Wilson’s disease: a practical guide from the British Association for the Study of the Liver. Lancet Gastroenterol Hepatol. 2022; 7(6): 560-575.

[165]

Tümer Z, Møller LB. Menkes disease. Eur J Hum Genet. 2010; 18(5): 511-518.

[166]

Springer C, Humayun D, Skouta R. Cuproptosis: unraveling the mechanisms of copper-induced cell death and its implication in cancer therapy. Cancers (Basel). 2024; 16(3): 647.

[167]

Horn N, Wittung-Stafshede P. ATP7A-regulated enzyme metalation and trafficking in the menkes disease puzzle. Biomedicines. 2021; 9(4): 391.

[168]

Ahuja A, Dev K, Tanwar RS, et al. Copper mediated neurological disorder: visions into amyotrophic lateral sclerosis, Alzheimer and Menkes disease. J Trace Elem Med Biol. 2015; 29: 11-23.

[169]

Garza NM, Swaminathan AB, Maremanda KP, et al. Mitochondrial copper in human genetic disorders. Trends Endocrinol Metab. 2023; 34(1): 21-33.

[170]

Yuan S, Korolnek T, Kim BE. Oral elesclomol treatment alleviates copper deficiency in animal models. Front Cell Dev Biol. 2022; 10: 856300.

[171]

Guthrie LM, Soma S, Yuan S, et al. Elesclomol alleviates Menkes pathology and mortality by escorting Cu to cuproenzymes in mice. Science. 2020; 368(6491): 620-625.

[172]

Zulkifli M, Spelbring AN, Zhang Y, et al. FDX1-dependent and independent mechanisms of elesclomol-mediated intracellular copper delivery. Proc Natl Acad Sci U S A. 2023; 120(10): e2216722120.

[173]

Haberkiewicz O, Lipiński P, Starzyński RR, et al. Decreased expression of the Slc31a1 gene and cytoplasmic relocalization of membrane CTR1 protein in renal epithelial cells: a potent protective mechanism against copper nephrotoxicity in a mouse model of menkes disease. Int J Mol Sci. 2022; 23(19): 11441.

[174]

Sluysmans S, Méan I, Xiao T, et al. PLEKHA5, PLEKHA6, and PLEKHA7 bind to PDZD11 to target the Menkes ATPase ATP7A to the cell periphery and regulate copper homeostasis. Mol Biol Cell. 2021; 32(21): ar34.

[175]

Aguila-Rosas J, García-Martínez BA, Ríos C, et al. Copper release by MOF-74(Cu): a novel pharmacological alternative to diseases with deficiency of a vital oligoelement. RSC Adv. 2024; 14(2): 855-862.

[176]

EASL clinical practice guidelines: Wilson’s disease. J Hepatol. 2012; 56(3): 671-685.

[177]

Schilsky ML. Wilson disease: clinical manifestations, diagnosis, and treatment. Clin Liver Dis (Hoboken). 2014; 3(5): 104-107.

[178]

Shribman S, Poujois A, Bandmann O, et al. Wilson’s disease: update on pathogenesis, biomarkers and treatments. J Neurol Neurosurg Psychiatry. 2021; 92(10): 1053-1061.

[179]

Lorincz MT. Neurologic Wilson’s disease. Ann N Y Acad Sci. 2010; 1184: 173-187.

[180]

Kaler SG. Inborn errors of copper metabolism. Handb Clin Neurol. 2013; 113: 1745-1754.

[181]

Pierson H, Muchenditsi A, Kim BE, et al. The function of ATPase copper transporter ATP7B in intestine. Gastroenterology. 2018; 154(1): 168-180. e5.

[182]

Polishchuk EV, Concilli M, Iacobacci S, et al. Wilson disease protein ATP7B utilizes lysosomal exocytosis to maintain copper homeostasis. Dev Cell. 2014; 29(6): 686-700.

[183]

Muchenditsi A, Yang H, Hamilton JP, et al. Targeted inactivation of copper transporter Atp7b in hepatocytes causes liver steatosis and obesity in mice. Am J Physiol Gastrointest Liver Physiol. 2017; 313(1): G39-G49.

[184]

Ruturaj, Mishra M, Saha S, et al. Regulation of the apico-basolateral trafficking polarity of the homologous copper-ATPases ATP7A and ATP7B. J Cell Sci. 2024; 137(5): jcs261258.

[185]

Zischka H, Lichtmannegger J. Pathological mitochondrial copper overload in livers of Wilson’s disease patients and related animal models. Ann N Y Acad Sci. 2014; 1315: 6-15.

[186]

Schilsky ML, Czlonkowska A, Zuin M, et al. Trientine tetrahydrochloride versus penicillamine for maintenance therapy in Wilson disease (CHELATE): a randomised, open-label, non-inferiority, phase 3 trial. Lancet Gastroenterol Hepatol. 2022; 7(12): 1092-1102.

[187]

Weiss KH, Askari FK, Czlonkowska A, et al. Bis-choline tetrathiomolybdate in patients with Wilson’s disease: an open-label, multicentre, phase 2 study. Lancet Gastroenterol Hepatol. 2017; 2(12): 869-876.

[188]

Monestier M, Pujol AM, Lamboux A, et al. A liver-targeting Cu(i) chelator relocates Cu in hepatocytes and promotes Cu excretion in a murine model of Wilson’s disease. Metallomics. 2020; 12(6): 1000-1008.

[189]

Krishnan N, Felice C, Rivera K, et al. DPM-1001 decreased copper levels and ameliorated deficits in a mouse model of Wilson’s disease. Genes Dev. 2018; 32(13-14): 944-952.

[190]

Lichtmannegger J, Leitzinger C, Wimmer R, et al. Methanobactin reverses acute liver failure in a rat model of Wilson disease. J Clin Invest. 2016; 126(7): 2721-2735.

[191]

Einer C, Munk DE, Park E, et al. ARBM101 (methanobactin SB2) drains excess liver copper via biliary excretion in Wilson’s disease rats. Gastroenterology. 2023; 165(1): 187-200. e7.

[192]

Liu Y, Miao J. An emerging role of defective copper metabolism in heart disease. Nutrients. 2022; 14(3): 700.

[193]

Parsanathan R. Copper’s dual role: unravelling the link between copper homeostasis, cuproptosis, and cardiovascular diseases. Hypertens Res. 2024; 47(5): 1440-1442.

[194]

Chen X, Cai Q, Liang R, et al. Copper homeostasis and copper-induced cell death in the pathogenesis of cardiovascular disease and therapeutic strategies. Cell Death Dis. 2023; 14(2): 105.

[195]

Cui Y, Chen Y, Gan N, et al. A novel cuproptosis-related diagnostic gene signature and differential expression validation in atherosclerosis. Mol Biomed. 2023; 4(1): 21.

[196]

Wang M, Cheng L, Xiang Q, et al. Evaluation the role of cuproptosis-related genes in the pathogenesis, diagnosis and molecular subtypes identification of atherosclerosis. Heliyon. 2023; 9(10): e21158.

[197]

Chen YT, Xu XH, Lin L, et al. Identification of three cuproptosis-specific expressed genes as diagnostic biomarkers and therapeutic targets for atherosclerosis. Int J Med Sci. 2023; 20(7): 836-848.

[198]

Chen Z, Li YY, Liu X. Copper homeostasis and copper-induced cell death: Novel targeting for intervention in the pathogenesis of vascular aging. Biomed Pharmacother. 2023; 169: 115839.

[199]

Ding C, Min J, Tan Y, et al. Combating atherosclerosis with chirality/phase dual-engineered nanozyme featuring microenvironment-programmed senolytic and senomorphic actions. Adv Mater. 2024; 36(29): e2401361.

[200]

Chen Z, Zhu Y, Chen S, et al. Immune patterns of cuproptosis in ischemic heart failure: a transcriptome analysis. J Cell Mol Med. 2024; 28(7): e18187.

[201]

Chen J, Yang X, Li W, et al. Potential molecular and cellular mechanisms of the effects of cuproptosis-related genes in the cardiomyocytes of patients with diabetic heart failure: a bioinformatics analysis. Front Endocrinol (Lausanne). 2024; 15: 1370387.

[202]

Huo S, Wang Q, Shi W, et al. ATF3/SPI1/SLC31A1 signaling promotes cuproptosis induced by advanced glycosylation end products in diabetic myocardial injury. Int J Mol Sci. 2023; 24(2): 1667.

[203]

Bian R, Wang Y, Li Z, Xu X. Identification of cuproptosis-related biomarkers in dilated cardiomyopathy and potential therapeutic prediction of herbal medicines. Front Mol Biosci. 2023; 10: 1154920.

[204]

Lin Y, Chen K, Guo J, et al. Identification of cuproptosis-related genes and immune infiltration in dilated cardiomyopathy. Int J Cardiol. 2024; 399: 131702.

[205]

Wang D, Tian Z, Zhang P, et al. The molecular mechanisms of cuproptosis and its relevance to cardiovascular disease. Biomed Pharmacother. 2023; 163: 114830.

[206]

Zhang S, Liu H, Amarsingh GV, et al. Restoration of myocellular copper-trafficking proteins and mitochondrial copper enzymes repairs cardiac function in rats with diabetes-evoked heart failure. Metallomics. 2020; 12(2): 259-272.

RIGHTS & PERMISSIONS

2024 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

231

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/