Copper Metabolism and Cuproptosis: Molecular Mechanisms and Therapeutic Perspectives in Neurodegenerative Diseases

Xiao-xia Ban, Hao Wan, Xin-xing Wan, Ya-ting Tan, Xi-min Hu, Hong-xia Ban, Xin-yu Chen, Kun Huang, Qi Zhang, Kun Xiong

Current Medical Science ›› 2024, Vol. 44 ›› Issue (1) : 28-50.

Current Medical Science All Journals
PDF
Current Medical Science ›› 2024, Vol. 44 ›› Issue (1) : 28-50. DOI: 10.1007/s11596-024-2832-z
Review Article

Copper Metabolism and Cuproptosis: Molecular Mechanisms and Therapeutic Perspectives in Neurodegenerative Diseases

Author information +
History +

Abstract

Copper is an essential trace element, and plays a vital role in numerous physiological processes within the human body. During normal metabolism, the human body maintains copper homeostasis. Copper deficiency or excess can adversely affect cellular function. Therefore, copper homeostasis is stringently regulated. Recent studies suggest that copper can trigger a specific form of cell death, namely, cuproptosis, which is triggered by excessive levels of intracellular copper. Cuproptosis induces the aggregation of mitochondrial lipoylated proteins, and the loss of iron-sulfur cluster proteins. In neurodegenerative diseases, the pathogenesis and progression of neurological disorders are linked to copper homeostasis. This review summarizes the advances in copper homeostasis and cuproptosis in the nervous system and neurodegenerative diseases. This offers research perspectives that provide new insights into the targeted treatment of neurodegenerative diseases based on cuproptosis.

Keywords

cuproptosis / copper metabolism / copper homeostasis / neurodegeneration / neurodegenerative disease

Cite this article

Download citation ▾
Xiao-xia Ban, Hao Wan, Xin-xing Wan, Ya-ting Tan, Xi-min Hu, Hong-xia Ban, Xin-yu Chen, Kun Huang, Qi Zhang, Kun Xiong. Copper Metabolism and Cuproptosis: Molecular Mechanisms and Therapeutic Perspectives in Neurodegenerative Diseases. Current Medical Science, 2024, 44(1): 28‒50 https://doi.org/10.1007/s11596-024-2832-z
This is a preview of subscription content, contact us for subscripton.

References

[1]
PfeifferCC, BravermanER. Zinc, the brain, and behavior. Biol Psychiatry, 1982, 17(4): 513-532
[2]
FestaRA, ThieleDJ. Copper: an essential metal in biology. Curr Biol, 2011, 21(21): R877-R883
CrossRef Google scholar
[3]
MaungMT, CarlsonA, Olea-FloresM, et al.. The molecular and cellular basis of copper dysregulation and its relationship with human pathologies. FASEB J, 2021, 35(9): e21810
CrossRef Google scholar
[4]
GromadzkaG, TarnackaB, FlagaA, et al.. Copper Dyshomeostasis in Neurodegenerative Diseases-Therapeutic Implications. Int J Mol Sci, 2020, 21(23): 9259
CrossRef Google scholar
[5]
ZhaoWJ, FanCL, HuXM, et al.. Regulated Cell Death of Retinal Ganglion Cells in Glaucoma: Molecular Insights and Therapeutic Potentials. Cell Mol Neurobiol, 2023, 43(7): 3161-3178
CrossRef Google scholar
[6]
ZhangQ, HuXM, ZhaoWJ, et al.. Targeting Necroptosis: A Novel Therapeutic Option for Retinal Degenerative Diseases. Int J Biol Sci, 2023, 19(2): 658-674
CrossRef Google scholar
[7]
WanH, YanYD, HuXM, et al.. Inhibition of mitochondrial VDAC1 oligomerization alleviates apoptosis and necroptosis of retinal neurons following OGD/R injury. Ann Anat, 2023, 247: 152049
CrossRef Google scholar
[8]
YanWT, ZhaoWJ, HuXM, et al.. PANoptosis-like cell death in ischemia/reperfusion injury of retinal neurons. Neural Regen Res, 2023, 18(2): 357-363
[9]
TangD, KangR, BergheTV, et al.. The molecular machinery of regulated cell death. Cell Res, 2019, 29(5): 347-364
CrossRef Google scholar
[10]
YangYD, LiZX, HuXM, et al.. Insight into Crosstalk Between Mitophagy and Apoptosis/Necroptosis: Mechanisms and Clinical Applications in Ischemic Stroke. Curr Med Sci, 2022, 42(2): 237-248
CrossRef Google scholar
[11]
YanWT, YangYD, HuXM, et al.. Do pyroptosis, apoptosis, and necroptosis (PANoptosis) exist in cerebral ischemia? Evidence from cell and rodent studies. Neural Regen Res, 2022, 17(8): 1761-1768
CrossRef Google scholar
[12]
ChenXY, DaiYH, WanXX, et al.. ZBP1-Mediated Necroptosis: Mechanisms and Therapeutic Implications. Molecules, 2022, 28(1): 52
CrossRef Google scholar
[13]
ChenJ, WangY, LiM, et al.. Netrin-1 Alleviates Early Brain Injury by Regulating Ferroptosis via the PPARy/Nrf2/GPX4 Signaling Pathway Following Subarachnoid Hemorrhage. Transl Stroke Res, 2024, 15(1): 219-237
CrossRef Google scholar
[14]
ZhangQ, WanXX, HuXM, et al.. Targeting Programmed Cell Death to Improve Stem Cell Therapy: Implications for Treating Diabetes and Diabetes-Related Diseases. Front Cell Dev Biol, 2021, 9: 809656
CrossRef Google scholar
[15]
HuXM, ZhangQ, ZhouRX, et al.. Programmed cell death in stem cell-based therapy: Mechanisms and clinical applications. World J Stem Cells, 2020, 12(8): 787-802
[16]
MouYH, WangJ, WuJC, et al.. Ferroptosis, a new form of cell death: opportunities and challenges in cancer. J Hematol Oncol, 2019, 29(1): 34 12
CrossRef Google scholar
[17]
TsvetkovP, CoyS, PetrovaB, et al.. Copper induces cell death by targeting lipoylated TCA cycle proteins. Science, 2022, 375(6586): 1254-1261
CrossRef Google scholar
[18]
AnY, LiS, HuangX, et al.. The Role of Copper Homeostasis in Brain Disease. Int J Mol Sci, 2022, 23(22): 13850
CrossRef Google scholar
[19]
PalA, RaniI, PawarA, et al.. Microglia and Astrocytes in Alzheimer’s Disease in the Context of the Aberrant Copper Homeostasis Hypothesis. Biomolecules, 2021, 11(11): 1598
CrossRef Google scholar
[20]
CobinePA, BradyDC. Cuproptosis: Cellular and molecular mechanisms underlying copper-induced cell death. Mol Cell, 2022, 82(10): 1786-1787
CrossRef Google scholar
[21]
ArredondoM, NúñezMT. Iron and copper metabolism. Mol Aspects Med, 2005, 26(4–5): 313-327
CrossRef Google scholar
[22]
Nutrition classics. The Journal of Biological Chemistry, Vol. LXXV II, 1928: Iron in nutrition. VII. Copper as a supplement to iron for hemoglobin building in the rat. By E.B. Hart, H. Steenbock, J. Waddell, and C.A. Elvehjem. Nutr Rev, 1987, 45(6): 181-183
[23]
KhoshbinK, CamilleriM. Effects of dietary components on intestinal permeability in health and disease. Am J Physiol Gastrointest Liver Physiol, 2020, 319(5): G589-G608
CrossRef Google scholar
[24]
NoseY, WoodLK, KimBE, et al.. Ctr1 is an apical copper transporter in mammalian intestinal epithelial cells in vivo that is controlled at the level of protein stability. J Biol Chem, 2010, 285(42): 32385-32392
CrossRef Google scholar
[25]
LaneDJR, BaeDH, MerlotAM, et al.. Duodenal cytochrome b (DCYTB) in iron metabolism: an update on function and regulation. Nutrients, 2015, 7(4): 2274-2296
CrossRef Google scholar
[26]
ZimnickaAM, MaryonEB, KaplanJH. Human copper transporter hCTR1 mediates basolateral uptake of copper into enterocytes: implications for copper homeostasis. J Biol Chem, 2007, 282(36): 26471-26480
CrossRef Google scholar
[27]
SchuchardtJP, HahnA. Intestinal Absorption and Factors Influencing Bioavailability of Magnesium-An Update. Curr Nutr Food Sci, 2017, 13(4): 260-278
CrossRef Google scholar
[28]
LutsenkoS. Dynamic and cell-specific transport networks for intracellular copper ions. J Cell Sci, 2021, 134(21): jcs240523
CrossRef Google scholar
[29]
NyasaeL, BustosR, BraitermanL, et al.. Dynamics of endogenous ATP7A (Menkes protein) in intestinal epithelial cells: copper-dependent redistribution between two intracellular sites. Am J Physiol Gastrointest Liver Physiol, 2007, 292(4): G1181-G1194
CrossRef Google scholar
[30]
LiangZD, TsaiWB, LeeMY, et al.. 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
CrossRef Google scholar
[31]
ZhaoJ, GuoS, SchrodiSJ, et al.. Cuproptosis and cuproptosis-related genes in rheumatoid arthritis: Implication, prospects, and perspectives. Front Immunol, 2022, 13: 930278
CrossRef Google scholar
[32]
LinderMC. Ceruloplasmin and other copper binding components of blood plasma and their functions: an update. Metallomics, 2016, 8(9): 887-905
CrossRef Google scholar
[33]
XieJ, YangY, GaoY, et al.. Cuproptosis: mechanisms and links with cancers. Mol Cancer, 2023, 22(1): 46
CrossRef Google scholar
[34]
CasarenoRL, WaggonerD, GitlinJD. The copper chaperone CCS directly interacts with copper/zinc superoxide dismutase. J Biol Chem, 1998, 273(37): 23625-23628
CrossRef Google scholar
[35]
PunterFA, AdamsDL, GlerumDM. Characterization and localization of human COX17, a gene involved in mitochondrial copper transport. Hum Genet, 2000, 107(1): 69-74
CrossRef Google scholar
[36]
PiersonH, MuchenditsiA, KimB-E, et al.. The Function of ATPase Copper Transporter ATP7B in Intestine. Gastroenterology, 2018, 154(1): 168-180
CrossRef Google scholar
[37]
TelianidisJ, HungYH, MateriaS, et al.. Role of the P-Type ATPases, ATP7A and ATP7B in brain copper homeostasis. Front Aging Neurosci, 2013, 5: 44
CrossRef Google scholar
[38]
LiuH, LaiW, LiuX, et al.. Exposure to copper oxide nanoparticles triggers oxidative stress and endoplasmic reticulum (ER)-stress induced toxicology and apoptosis in male rat liver and BRL-3A cell. J Hazard Mater, 2021, 401: 123349
CrossRef Google scholar
[39]
van RensburgMJ, van RooyM, BesterMJ, et al.. Oxidative and haemostatic effects of copper, manganese and mercury, alone and in combination at physiologically relevant levels: An ex vivo study. Hum Exp Toxicol, 2019, 38(4): 419-433
CrossRef Google scholar
[40]
ComesG, Fernandez-GayolO, MolineroA, et al.. Mouse metallothionein-1 and metallothionein-2 are not biologically interchangeable in an animal model of multiple sclerosis, EAE. Metallomics, 2019, 11(2): 327-337
CrossRef Google scholar
[41]
LearySC, WingeDR, CobinePA. “Pulling the plug” on cellular copper: the role of mitochondria in copper export. Biochim Biophys Acta, 2009, 1793(1): 146-153
CrossRef Google scholar
[42]
HornD, BarrientosA. Mitochondrial copper metabolism and delivery to cytochrome c oxidase. IUBMB Life, 2008, 60(7): 421-429
CrossRef Google scholar
[43]
Pacheu-GrauD, WasilewskiM, OeljeklausS, et al.. COA6 Facilitates Cytochrome c Oxidase Biogenesis as Thiol-reductase for Copper Metallochaperones in Mitochondria. J Mol Biol, 2020, 432(7): 2067-2079
CrossRef Google scholar
[44]
RobinsonNJ, WingeDR. Copper metallochaperones. Annu Rev Biochem, 2010, 79: 537-562
CrossRef Google scholar
[45]
JettKA, LearySC. Building the CuA site of cytochrome c oxidase: A complicated, redox-dependent process driven by a surprisingly large complement of accessory proteins. J Biol Chem, 2018, 293(13): 4644-4652
CrossRef Google scholar
[46]
LearySC. Redox regulation of SCO protein function: controlling copper at a mitochondrial crossroad. Antioxid Redox Signal, 2010, 13(9): 1403-1416
CrossRef Google scholar
[47]
MorgadaMN, AbriataLA, CefaroC, et al.. Loop recognition and copper-mediated disulfide reduction underpin meta l site assembly of CuA in human cytochrome oxidase. Proc Natl Acad Sci U S A, 2015, 112(38): 11771-11776
CrossRef Google scholar
[48]
ZhouJ, LiXY, LiuYJ, et al.. Full-coverage regulations of autophagy by ROS: from induction to maturation. Autophagy, 2022, 18(6): 1240-1255
CrossRef Google scholar
[49]
BompianiKM, TsaiCY, AchatzFP, et al.. Copper transporters and chaperones CTR1, CTR2, ATOX1, and CCS as determinants of cisplatin sensitivity. Metallomics, 2016, 8(9): 951-962
CrossRef Google scholar
[50]
SkoppA, BoydSD, UllrichMS, et al.. Copper-zinc superoxide dismutase (Sod1) activation terminates interact ion between its copper chaperone (Ccs) and the cytosolic metal-binding domain of the copper importer Ctr1. Biometals, 2019, 32(4): 695-705
CrossRef Google scholar
[51]
GuptaA, LutsenkoS. Human copper transporters: mechanism, role in human diseases and therapeutic potential. Future Med Chem, 2009, 1(6): 1125-1142
CrossRef Google scholar
[52]
HamzaI, FaisstA, ProhaskaJ, et al.. The metallochaperone Atox1 plays a critical role in perinatal copper homeostasis. Proc Natl Acad Sci USA, 2001, 98(12): 6848-6852
CrossRef Google scholar
[53]
ItohS, KimHW, NakagawaO, 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
CrossRef Google scholar
[54]
MattieMD, McElweeMK, FreedmanJH. Mechanism of copper-activated transcription: activation of AP-1, and the JNK/SAPK and p38 signal transduction pathways. J Mol Biol, 2008, 383(5): 1008-1018
CrossRef Google scholar
[55]
SatakeH, SuzukiK, AokiT, et al.. Cupric ion blocks NF kappa B activation through inhibiting the signal-induced phosphorylation of I kappa B alpha. Biochem Biophys Res Commun, 1995, 216(2): 568-573
CrossRef Google scholar
[56]
ChenL, LiN, ZhangM, et al.. APEX2-based Proximity Labeling of Atox1 Identifies CRIP2 as a Nuclear Copper-binding Protein that Regulates Autophagy Activation. Angew Chem Int Ed Engl, 2021, 60(48): 25346-25355
CrossRef Google scholar
[57]
AnY, LiS, HuangX, et al.. The Role of Copper Homeostasis in Brain Disease. Int J Mol Sci, 2022, 23(22): 13850
CrossRef Google scholar
[58]
TongX, TangR, XiaoM, et al.. Targeting cell death pathways for cancer therapy: recent developments in necroptosis, pyroptosis, ferroptosis, and cuproptosis research. J Hematol Oncol, 2022, 15(1): 174
CrossRef Google scholar
[59]
TümerZ, MøllerLB. Menkes disease. Eur J Hum Genet, 2010, 18(5): 511-518
CrossRef Google scholar
[60]
CzłonkowskaA, LitwinT, DusekP, et al.. Wilson disease. Nat Rev Dis Primers, 2018, 4(1): 21
CrossRef Google scholar
[61]
BushAI. Metals and neuroscience. Curr Opin Chem Biol, 2000, 4(2): 184-191
CrossRef Google scholar
[62]
DiDonatoM, NarindrasorasakS, ForbesJR, et al.. Expression, purification, and metal binding properties of_the N-terminal domain from the wilson disease putative copper-transporting ATPase (ATP7B). J Biol Chem, 1997, 272(52): 33279-33282
CrossRef Google scholar
[63]
LönnerdalB. Intestinal regulation of copper homeostasis: a developmental perspective. Am J Clin Nutr, 2008, 88(3): 846
CrossRef Google scholar
[64]
AisenP, EnnsC, Wessling-ResnickM. Chemistry and biology of eukaryotic iron metabolism. Int J Biochem Cell Biol, 2001, 33(10): 940-959
CrossRef Google scholar
[65]
AkilM, SchwartzJA, DutchakD, et al.. The psychiatric presentations of Wilson’s disease. J Neuropsychiatry Clin Neurosci, 1991, 3(4): 377-382
CrossRef Google scholar
[66]
Mairet-CoelloG, TuryA, Esnard-FeveA, et al.. FAD-linked sulfhydryl oxidase QSOX: topographic, cellular, and subcellular immunolocalization in adult rat central nervous system. J Comp Neurol, 2004, 473(3): 334-363
CrossRef Google scholar
[67]
TrombleyPQ, HorningMS, BlakemoreLJ. Interactions between carnosine and zinc and copper: implications for neuromodulation and neuroprotection. Biochemistry (Mosc), 2000, 65(7): 807-816
[68]
JohnsonKA, ConnPJ, NiswenderCM. Glutamate receptors as therapeutic targets for Parkinson’s disease. CNS Neurol Disord Drug Targets, 2009, 8(6): 475-491
CrossRef Google scholar
[69]
D’AmbrosiN, RossiL. Copper at synapse: Release, binding and modulation of neurotransmission. Neurochem Int, 2015, 90: 36-45
CrossRef Google scholar
[70]
MoriyaM, HoYH, GranaA, et al.. Copper is taken up efficiently from albumin and alpha2-macroglobulin by cultured human cells by more than one mechanism. Am J Physiol Cell Physiol, 2008, 295(3): C708-C721
CrossRef Google scholar
[71]
MontesS, Rivera-ManciaS, Diaz-RuizA, et al.. Copper and copper proteins in Parkinson’s disease. Oxid Med Cell Longev, 2014, 2014: 147251
CrossRef Google scholar
[72]
WuW, RuanX, GuC, et al.. Blood-cerebrospinal fluid barrier permeability of metals/metalloids and its determinants in pediatric patients. Ecotoxicol Environ Saf, 2023, 266: 115599
CrossRef Google scholar
[73]
ScheiberIF, MercerJFB, DringenR. Metabolism and functions of copper in brain. Prog Neurobiol, 2014, 116: 33-57
CrossRef Google scholar
[74]
DringenR, ScheiberIF, MercerJFB. Copper metabolism of astrocytes. Front Aging Neurosci, 2013, 5: 9
CrossRef Google scholar
[75]
HowellSB, SafaeiR, LarsonCA, et al.. Copper transporters and the cellular pharmacology of the platinum-containing cancer drugs. Mol Pharmacol, 2010, 77(6): 887-894
CrossRef Google scholar
[76]
Garza-LombóC, PosadasY, QuintanarL, et al.. Neurotoxicity Linked to Dysfunctional Metal Ion Homeostasis and Xenobiotic Metal Exposure: Redox Signaling and Oxidative Stress. Antioxid Redox Signal, 2018, 28(18): 1669-1703
CrossRef Google scholar
[77]
Varela-NallarL, ToledoEM, ChacónMA, et al.. The functional links between prion protein and copper. Biol Res, 2006, 39(1): 39-44
CrossRef Google scholar
[78]
StuerenburgHJ. CSF copper concentrations, blood-brain barrier function, and coeruloplasmin synthesis during the treatment of Wilson’s disease. J Neural Transm (Vienna), 2000, 107(3): 321-329
CrossRef Google scholar
[79]
ChoiBS, ZhengW. Copper transport to the brain by the blood-brain barrier and blood-CSF barrier. Brain Res, 2009, 1248: 14-21
CrossRef Google scholar
[80]
KalerSG. ATP7A-related copper transport diseases-emerging concepts and future trends. Nat Rev Neurol, 2011, 7(1): 15-29
CrossRef Google scholar
[81]
NishiharaE, FuruyamaT, YamashitaS, et al.. Expression of copper trafficking genes in the mouse brain. Neuroreport, 1998, 9(14): 3259-3263
CrossRef Google scholar
[82]
BarberRG, GrenierZA, BurkheadJL. Copper Toxicity Is Not Just Oxidative Damage: Zinc Systems and Insight from Wilson Disease. Biomedicines, 2021, 9(3): 316
CrossRef Google scholar
[83]
MercerJF, AmbrosiniL, HortonS, et al.. Animal models of Menkes disease. Adv Exp Med Biol, 1999, 448: 97-108
CrossRef Google scholar
[84]
MenkesJH, AlterM, SteiglederGK, et al.. A sex-linked recessive disorder with retardation of growth, peculiar hair, and focal cerebral and cerebellar degeneration. Pediatrics, 1962, 29: 764-779
[85]
MøllerLB, MogensenM, HornN. Molecular diagnosis of Menkes disease: genotype-phenotype correlation. Biochimie, 2009, 91(10): 1273-1277
CrossRef Google scholar
[86]
ShimH, HarrisZL. Genetic defects in copper metabolism. J Nutr, 2003, 133(5): 1527S-1531S Suppl 1
CrossRef Google scholar
[87]
KalerSG, GahlWA, BerrySA, et al.. Predictive value of plasma catecholamine levels in neonatal detection of Menkes disease. J Inherit Metab Dis, 1993, 16(5): 907-908
CrossRef Google scholar
[88]
SzauterKM, CaoT, BoydCD, et al.. Lysyl oxidase in development, aging and pathologies of the skin. Pathol Biol (Paris), 2005, 53(7): 448-456
CrossRef Google scholar
[89]
RoycePM, CamakarisJ, DanksDM. Reduced lysyl oxidase activity in skin fibroblasts from patients with Menkes’ syndrome. Biochem J, 1980, 192(2): 579-586
CrossRef Google scholar
[90]
SarkarB, Lingertat WalshK, ClarkeJT. Copper-histidine therapy for Menkes disease. J Pediatr, 1993, 123(5): 828-830
CrossRef Google scholar
[91]
GeorgeDH, CaseyRE. Menkes disease after copper histidine replacement therapy: case report. Pediatr Dev Pathol, 2001, 4(3): 281-288
CrossRef Google scholar
[92]
KimJH, LeeBH, KimYM, et al.. Novel mutations and clinical outcomes of copper-histidine therapy in Menkes disease patients. Metab Brain Dis, 2015, 30(1): 75-81
CrossRef Google scholar
[93]
CumingsJN. The copper and iron content of brain and liver in the normal and in hepato-lenticular degeneration. Brain, 1948, 71: 410-415 Pt. 4
CrossRef Google scholar
[94]
PrzybyłkowskiA, GromadzkaG, ChabikG, et al.. Liver cirrhosis in patients newly diagnosed with neurological phenotype of Wilson’s disease. Funct Neurol, 2014, 29(1): 23-29
[95]
WalsheJM, PotterG. The pattern of the wholebody distribution of radioactive copper (67Cu, 64Cu) in Wilson’s Disease and various control groups. Q J Med, 1977, 46(184): 445-462
[96]
HoroupianDS, SternliebI, ScheinbergIH. Neuropathological findings in penicillamine-treated patients with Wilson’s disease. Clin Neuropathol, 1988, 7(2): 62-67
[97]
BertrandE, LewandowskaE, SzpakGM, et al.. Neuropathological analysis of pathological forms of astroglia in Wilson’s disease. Folia Neuropathol, 2001, 39(2): 73-79
[98]
Langwińska-WośkoE, LitwinT, SzulborskiK, et al.. Optical coherence tomography and electrophysiology of retinal and visual pathways in Wilson’s disease. Metab Brain Dis, 2016, 31(2): 405-415
CrossRef Google scholar
[99]
CzłonkowskaA, LitwinT, KarlińskiM, et al.. D-penicillamine versus zinc sulfate as first-line therapy for Wilson’s disease. Eur J Neurol, 2014, 21(4): 599-606
CrossRef Google scholar
[100]
European Association for Study of L. EASL Clinical Practice Guidelines: Wilson’s disease. J Hepatol, 2012, 56(3): 671-685
CrossRef Google scholar
[101]
BarnesDE, YaffeK. The projected effect of risk factor reduction on Alzheimer’s disease prevalence. Lancet Neurol, 2011, 10(9): 819-828
CrossRef Google scholar
[102]
MorrisGP, ClarkIA, VisselB. Inconsistencies and controversies surrounding the amyloid hypothesis of Alzheimer’s disease. Acta Neuropathol Commun, 2014, 2: 135
[103]
ZhangYL, WangJ, ZhangZN, et al.. The relationship between amyloid-beta and brain capillary endothelial cells in Alzheimer’s disease. Neural Regen Res, 2022, 17(11): 2355-2363
CrossRef Google scholar
[104]
GaggelliE, KozlowskiH, ValensinD, et al.. Copper homeostasis and neurodegenerative disorders (Alzheimer’s, prion, and Parkinson’s diseases and amyotrophic lateral sclerosis). Chem Rev, 2006, 106(6): 1995-2044
CrossRef Google scholar
[105]
AtwoodCS, ScarpaRC, HuangX, et al.. Characterization of copper interactions with alzheimer amyloid beta peptides: identification of an attomolar-affinity copper binding site on amyloid beta1-42. J Neurochem, 2000, 75(3): 1219-1233
CrossRef Google scholar
[106]
NewcombeEA, Camats-PernaJ, SilvaML, et al.. Inflammation: the link between comorbidities, genetics, and Alzheimer’s disease. J Neuroinflammation, 2018, 15(1): 276
CrossRef Google scholar
[107]
LovellMA, RobertsonJD, TeesdaleWJ, et al.. Copper, iron and zinc in Alzheimer’s disease senile plaques. J Neurol Sci, 1998, 158(1): 47-52
CrossRef Google scholar
[108]
SuXY, WuWH, HuangZP, et al.. Hydrogen peroxide can be generated by tau in the presence of Cu(II). Biochem Biophys Res Commun, 2007, 358(2): 661-665
CrossRef Google scholar
[109]
YuJ, LuoX, XuH, et al.. Identification of the key molecules involved in chronic copper exposure-aggravated memory impairment in transgenic mice of Alzheimer’s disease using proteomic analysis. J Alzheimers Dis, 2015, 44(2): 455-469
CrossRef Google scholar
[110]
JamesSA, VolitakisI, AdlardPA, et al.. Elevated labile Cu is associated with oxidative pathology in Alzheimer disease. Free Radic Biol Med, 2012, 52(2): 298-302
CrossRef Google scholar
[111]
YuF, GongP, HuZ, et al.. Cu (II) enhances the effect of Alzheimer’s amyloid-β peptide on microglial activation. J Neuroinflammation, 2015, 12: 122
CrossRef Google scholar
[112]
LuJ, WuD, ZhengY, et al.. Trace amounts of copper exacerbate beta amyloid-induced neurotoxicity in the cholesterol-fed mice through TNF-mediated inflammatory pathway. Brain Behav Immun, 2009, 23(2): 193-203
CrossRef Google scholar
[113]
Rossi-GeorgeA, GuoCJ. Copper disrupts S-nitrosothiol signaling in activated BV2 microglia. Neurochem Int, 2016, 99: 1-8
CrossRef Google scholar
[114]
KrasemannS, MadoreC, CialicR, et al.. The TREM2-APOE Pathway Drives the Transcriptional Phenotype of Dysfunctional Microglia in Neurodegenerative Diseases. Immunity, 2017, 47(3): 566-581
CrossRef Google scholar
[115]
ChenY, ChenJ, WeiH, et al.. Akkermansia muciniphila-Nlrp3 is involved in the neuroprotection of phosphoglycerate mutase 5 deficiency in traumatic brain injury mice. Front Immunol, 2023, 14: 1172710
CrossRef Google scholar
[116]
ZhouZ, ShangL, ZhangQ, et al.. DTX3L induced NLRP3 ubiquitination inhibit R28 cell pyroptosis in OGD/R injury. Biochim Biophys Acta Mol Cell Res, 2023, 1870(3): 119433
CrossRef Google scholar
[117]
HeYF, HuXM, KhanMA, et al.. HSF1 Alleviates Brain Injury by Inhibiting NLRP3-Induced Pyroptosis in a Sepsis Model. Mediators Inflamm, 2023, 2023: 2252255
CrossRef Google scholar
[118]
HuangY, WangS, HuangF, et al.. c-FLIP regulates pyroptosis in retinal neurons following oxygen-glucose deprivation/recovery via a GSDMD-mediated pathway. Ann Anat, 2021, 235: 151672
CrossRef Google scholar
[119]
LiaoLS, LuS, YanWT, et al.. The Role of HSP90α in Methamphetamine/Hyperthermia-Induced Necroptosis in Rat Striatal Neurons. Front Pharmacol, 2021, 12: 716394
CrossRef Google scholar
[120]
YanWT, LuS, YangYD, et al.. Research trends, hot spots and prospects for necroptosis in the field of neuroscience. Neural Regen Res, 2021, 16(8): 1628-1637
CrossRef Google scholar
[121]
HuXM, LiZX, LinRH, et al.. Guidelines for Regulated Cell Death Assays: A Systematic Summary, A Categorical Comparison, A Prospective. Front Cell Dev Biol, 2021, 9: 634690
CrossRef Google scholar
[122]
WakhlooD, OberhauserJ, MadiraA, et al.. From cradle to grave: neurogenesis, neuroregeneration and neurodegeneration in Alzheimer’s and Parkinson’s diseases. Neural Regen Res, 2022, 17(12): 2606-2614
CrossRef Google scholar
[123]
BanjaraM, GhoshC. Sterile Neuroinflammation and Strategies for Therapeutic Intervention. Int J Inflam, 2017, 2017: 8385961
[124]
ScheiberIF, DringenR. Astrocyte functions in the copper homeostasis of the brain. Neurochem Int, 2013, 62(5): 556-565
CrossRef Google scholar
[125]
PalA, VasishtaR, PrasadR. Hepatic and hippocampus iron status is not altered in response to increased serum ceruloplasmin and serum “free” copper in Wistar rat model for non-Wilsonian brain copper toxicosis. Biol Trace Elem Res, 2013, 154(3): 403-411
CrossRef Google scholar
[126]
QianY, ZhengY, TaylorR, et al.. Involvement of the molecular chaperone Hspa5 in copper homeostasis in astrocytes. Brain Res, 2012, 1447: 9-19
CrossRef Google scholar
[127]
PikeCJ, CummingsBJ, MonzaviR, et al.. Betaamyloid-induced changes in cultured astrocytes parallel reactive astrocytosis associated with senile plaques in Alzheimer’s disease. Neuroscience, 1994, 63(2): 517-531
CrossRef Google scholar
[128]
DeWittDA, PerryG, CohenM, et al.. Astrocytes regulate microglial phagocytosis of senile plaque cores of Alzheimer’s disease. Exp Neurol, 1998, 149(2): 329-340
CrossRef Google scholar
[129]
ChooXY, LiddellJR, HuuskonenMT, et al.. CuII(atsm) Attenuates Neuroinflammation. Front Neurosci, 2018, 12: 668
CrossRef Google scholar
[130]
MandalPK, SaharanS, TripathiM, et al.. Brain glutathione levels—a novel biomarker for mild cognitive impairment and Alzheimer’s disease. Biol psychiatry, 2015, 78(10): 702-710
CrossRef Google scholar
[131]
UttamsinghV, KellerDA, AndersMW. Acylase I-catalyzed deacetylation of N-acetyl-L-cysteine and S-alkyl-N -acetyl-L-cysteines. Chem Res Toxicol, 1998, 11(7): 800-809
CrossRef Google scholar
[132]
AshrafA, SoPW. Spotlight on Ferroptosis: Iron-Dependent Cell Death in Alzheimer’s Disease. Front Aging Neurosci, 2020, 12: 196
CrossRef Google scholar
[133]
DerryPJ, HegdeML, JacksonGR, et al.. Revisiting the intersection of amyloid, pathologically modified tau and iron in Alzheimer’s disease from a ferroptosis perspective. Prog Neurobiol, 2020, 184: 101716
CrossRef Google scholar
[134]
BushAI. Drug development based on the metals hypothesis of Alzheimer’s disease. J Alzheimers Dis, 2008, 15(2): 223-240
CrossRef Google scholar
[135]
WalkerFO. Huntington’s disease. Lancet, 2007, 369(9557): 218-228
CrossRef Google scholar
[136]
ArrasateM, FinkbeinerS. Protein aggregates in Huntington’s disease. Exp Neurol, 2012, 238(1): 1-11
CrossRef Google scholar
[137]
DexterDT, CarayonA, Javoy-AgidF, et al.. Alterations in the levels of iron, ferritin and other trace metals in Parkinson’s disease and other neurodegenerative diseases affecting the basal ganglia. Brain, 1991, 114: 1953-1975 Pt4
CrossRef Google scholar
[138]
TabriziSJ, GhoshR, LeavittBR. Huntingtin Lowering Strategies for Disease Modification in Huntington’s Disease. Neuron, 2019, 101(5): 801-819
CrossRef Google scholar
[139]
XiaoG, FanQ, WangX, et al.. Huntington disease arises from a combinatory toxicity of polyglutamine and copper binding. Proc Natl Acad Sci U S A, 2013, 110(37): 14995-15000
CrossRef Google scholar
[140]
FoxJH, KamaJA, LiebermanG, et al.. Mechanisms of copper ion mediated Huntington’s disease progression. PLoS One, 2007, 2(3): e334
CrossRef Google scholar
[141]
PampK, BrameyT, KirschM, et al.. NAD(H) enhances the Cu (II)-mediated inactivation of lactate dehydrogenase by increasing the accessibility of sulfhydryl groups. Free Radic Res, 2005, 39(1): 31-40
CrossRef Google scholar
[142]
HarmsL, MeierkordH, TimmG, et al.. Decreased N-acetyl-aspartate/choline ratio and increased lactate in the frontal lobe of patients with Huntington’s disease: a proton magnetic resonance spectroscopy study. J Neurol Neurosurg Psychiatry, 1997, 62(1): 27-30
CrossRef Google scholar
[143]
ShelineCT, ChoiDW. Cu2+ toxicity inhibition of mitochondrial dehydrogenases in vitro and in vivo. Ann Neurol, 2004, 55(5): 645-653
CrossRef Google scholar
[144]
ChernyRA, AytonS, FinkelsteinDI, et al.. PBT2 Reduces Toxicity in a C. elegans Model of polyQ Aggregation and Extends Lifespan, Reduces Striatal Atrophy and Improves Motor Performance in the R6/2 Mouse Model of Huntington’s Disease. J Huntingtons Dis, 2012, 1(2): 211-219
CrossRef Google scholar
[145]
BoilléeS, Vande VeldeC, ClevelandDW. ALS: a disease of motor neurons and their nonneuronal neighbors. Neuron, 2006, 52(1): 39-59
CrossRef Google scholar
[146]
SwinnenB, RobberechtW. The phenotypic variability of amyotrophic lateral sclerosis. Nat Rev Neurol, 2014, 10(11): 661-670
CrossRef Google scholar
[147]
FeldmanEL, GoutmanSA, PetriS, et al.. Amyotrophic lateral sclerosis. Lancet, 2022, 400(10360): 1363-1380
CrossRef Google scholar
[148]
HardimanO, Al-ChalabiA, ChioA, et al.. Amyotrophic lateral sclerosis. Nat Rev Dis Primers, 2017, 3: 17071
CrossRef Google scholar
[149]
Gil-BeaFJ, AldanondoG, Lasa-FernándezH, et al.. Insights into the mechanisms of copper dyshomeostasis in amyotrophic lateral sclerosis. Expert Rev Mol Med, 2017, 19: e7
CrossRef Google scholar
[150]
SonM, PuttaparthiK, KawamataH, et al.. Overexpression of CCS in G93A-SOD1 mice leads to accelerated neurologi cal deficits with severe mitochondrial pathology. Proc Natl Acad Sci USA, 2007, 104(14): 6072-6077
CrossRef Google scholar
[151]
WilliamsJR, TriasE, BeilbyPR, et al.. Copper delivery to the CNS by CuATSM effectively treats motor neuron disease in SOD(G93A) mice co-expressing the Copper-Chaperone-for-SOD. Neurobiol Dis, 2016, 89: 1-9
CrossRef Google scholar
[152]
CabreiroF, AckermanD, DoonanR, et al.. Increased life span from overexpression of superoxide dismutase in Caenorhabditis elegans is not caused by decreased oxidative damage. Free Radic Biol Med, 2011, 51(8): 1575-1582
CrossRef Google scholar
[153]
EngeTG, EcroydH, JolleyDF, et al.. Longitudinal assessment of metal concentrations and copper isotope ratios in the G93A SOD1 mouse model of amyotrophic lateral sclerosis. Metallomics, 2017, 9(2): 161-174
CrossRef Google scholar
[154]
RoosPM, VesterbergO, SyversenT, et al.. Metal concentrations in cerebrospinal fluid and blood plasma from patients with amyotrophic lateral sclerosis. Biol Trace Elem Res, 2013, 151(2): 159-170
CrossRef Google scholar
[155]
HiltonJB, KyseniusK, WhiteAR, et al.. The accumulation of enzymatically inactive cuproenzymes is a CNS-specific phenomenon of the SOD1G37R mouse model of ALS and can be restored by overexpressing the human copper transporter hCTR1. Exp Neurol, 2018, 307: 118-128
CrossRef Google scholar
[156]
TokudaE, OkawaE, OnoS. Dysregulation of intracellular copper trafficking pathway in a mouse model of mutant copper/zinc superoxide dismutase-linked familial amyotrophic lateral sclerosis. J Neurochem, 2009, 111(1): 181-191
CrossRef Google scholar
[157]
HottingerAF, FineEG, GurneyME, et al.. The copper chelator d-penicillamine delays onset of disease and extend s survival in a transgenic mouse model of familial amyotrophic lateral sclerosis. Eur J Neurosci, 1997, 9(7): 1548-1551
CrossRef Google scholar
[158]
TokudaE, OnoS, IshigeK, et al.. Ammonium tetrathiomolybdate delays onset, prolongs survival, and slows progression of disease in a mouse model for amyotrophic lateral sclerosis. Exp Neurol, 2008, 213(1): 122-128
CrossRef Google scholar
[159]
RobertsBR, LimNKH, McAllumEJ, et al.. Oral treatment with Cu (II)(atsm) increases mutant SOD1 in vivo but protects motor neurons and improves the phenotype of a transgenic mouse model of amyotrophic lateral sclerosis. J Neurosci, 2014, 34(23): 8021-8031
CrossRef Google scholar
[160]
HiltonJB, MercerSW, LimNKH, et al.. CuII(atsm) improves the neurological phenotype and survival of SOD1G93A mice and selectively increases enzymatically active SOD1 in the spinal cord. Sci Rep, 2017, 7: 42292
CrossRef Google scholar
[161]
TokudaE, OkawaE, WatanabeS, 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
CrossRef Google scholar
[162]
De LazzariF, BubaccoL, WhitworthAJ, et al.. Superoxide Radical Dismutation as New Therapeutic Strategy in Parkinson’s Disease. Aging Dis, 2018, 9(4): 716-728
CrossRef Google scholar
[163]
ReichSG, SavittJM. Parkinson’s Disease. Med Clin North Am, 2019, 103(2): 337-350
CrossRef Google scholar
[164]
Karimi-MoghadamA, CharsoueiS, BellB, et al.. Parkinson Disease from Mendelian Forms to Genetic Susceptibility: New Molecular Insights into the Neurodegeneration Process. Cell Mol Neurobiol, 2018, 38(6): 1153-1178
CrossRef Google scholar
[165]
TolosaE, GarridoA, ScholzSW, et al.. Challenges in the diagnosis of Parkinson’s disease. Lancet Neurol, 2021, 20(5): 385-397
CrossRef Google scholar
[166]
Youdim MB, Ben-Shachar D, Riederer P. Is Parkinson’s disease a progressive siderosis of substantia nigra resulting in iron and melanin induced neurodegeneration? Acta Neurol Scand Suppl, 1989,126:47–54
[167]
RiedererP, SoficE, RauschWD, et al.. Transition metals, ferritin, glutathione, and ascorbic acid in parkinsonian brains. J Neurochem, 1989, 52(2): 515-520
CrossRef Google scholar
[168]
Perry TL, Godin DV, Hansen S. Parkinson’s disease: a disorder due to nigral glutathione deficiency? Neurosci Lett, 1982,33(3):305–310
[169]
JennerP. Oxidative stress in Parkinson’s disease. Ann Neurol, 2003, 53: S26-S36
CrossRef Google scholar
[170]
BisagliaM, MammiS, BubaccoL. Structural insights on physiological functions and pathological effects of alpha-synuclein. FASEB J, 2009, 23(2): 329-340
CrossRef Google scholar
[171]
BinolfiA, RasiaRM, BertonciniCW, et al.. Interaction of alpha-synuclein with divalent metal ions reveals key differences: a link between structure, binding specificity and fibrillation enhancement. J Am Chem Soc, 2006, 128(30): 9893-9901
CrossRef Google scholar
[172]
DudzikCG, WalterED, MillhauserGL. Coordination features and affinity of the Cu2+ site in the α-synuclein protein of Parkinson’s disease. Biochemistry, 2011, 50(11): 1771-1777
CrossRef Google scholar
[173]
McDowallJS, BrownDR. Alpha-synuclein: relating metals to structure, function and inhibition. Metallomics, 2016, 8(4): 385-397
CrossRef Google scholar
[174]
AndersonJP, WalkerDE, GoldsteinJM, et al.. Phosphorylation of Ser-129 is the dominant pathological modification of alpha-synuclein in familial and sporadic Lewy body disease. J Biol Chem, 2006, 281(40): 29739-29752
CrossRef Google scholar
[175]
DikiyI, EliezerD. N-terminal acetylation stabilizes N-terminal helicity in lipid- and micelle-bound α-synuclein and increases its affinity for physiological membranes. J Biol Chem, 2014, 289(6): 3652-3665
CrossRef Google scholar
[176]
MasonRJ, PaskinsAR, DaltonCF, et al.. Copper Binding and Subsequent Aggregation of α-Synuclein Are Modulated by N-Terminal Acetylation and Ablated by the H50Q Missense Mutation. Biochem, 2016, 55(34): 4737-4741
CrossRef Google scholar
[177]
Bisaglia M, Bubacco L. Copper Ions and Parkinson’s Disease: Why Is Homeostasis So Relevant? Biomolecules, 2020,10(2):195
[178]
UittiRJ, RajputAH, RozdilskyB, et al.. Regional metal concentrations in Parkinson’s disease, other chronic neurological diseases, and control brains. Can J Neurol Sci, 1989, 16(3): 310-314
CrossRef Google scholar
[179]
de FreitasLV, da SilvaCCP, EllenaJ, et al.. Structural and vibrational study of 8-hydroxyquinoline-2-carboxaldehyde isonicotinoyl hydrazine-a potential metal-protein attenuating compound (MPAC) for the treatment of Alzheimer’s disease. Spectrochim Acta A Mol Biomol Spectrosc, 2013, 116: 41-48
CrossRef Google scholar
[180]
McAllumEJ, LimNKH, HickeyJL, et al.. Therapeutic effects of CuII(atsm) in the SOD1-G37R mouse model of amyotrophic lateral sclerosis. Amyotroph Lateral Scler Frontotemporal Degener, 2013, 14(7–8): 586-590
CrossRef Google scholar
[181]
WangM, WanH, WangS, et al.. RSK3 mediates necroptosis by regulating phosphorylation of RIP3 in rat retinal ganglion cells. J Anat, 2020, 237(1): 29-47
CrossRef Google scholar
[182]
GuoLM, WangZ, LiSP, et al.. RIP3/MLKL-mediated neuronal necroptosis induced by methamphetamine at 39°C. Neural Regen Res, 2020, 15(5): 865-874
CrossRef Google scholar
[183]
WangZ, GuoLM, WangY, et al.. Inhibition of HSP90α protects cultured neurons from oxygen-glucose deprivation induced necroptosis by decreasing RIP3 expression. J Cell Physiol, 2018, 233(6): 4864-4884
CrossRef Google scholar
[184]
ChenJ, LiM, LiuZ, et al.. Molecular mechanisms of neuronal death in brain injury after subarachnoid hemorrhage. Front Cell Neurosci, 2022, 16: 1025708
CrossRef Google scholar
[185]
HunsakerEW, FranzKJ. Emerging Opportunities To Manipulate Metal Trafficking for Therapeutic Benefit. Inorg Chem, 2019, 58(20): 13528-13545
CrossRef Google scholar
[186]
ZhengP, ZhouC, LuL, et al.. Elesclomol: a copper ionophore targeting mitochondrial metabolism for cancer therapy. J Exp Clin Cancer Res, 2022, 41(1): 271
CrossRef Google scholar
[187]
HasinoffBB, YadavAA, PatelD, et al.. The cytotoxicity of the anticancer drug elesclomol is due to oxidative stress indirectly mediated through its complex with Cu (II). J Inorg Biochem, 2014, 137: 22-30
CrossRef Google scholar
[188]
YangW, WangY, HuangY, et al.. 4-Octyl itaconate inhibits aerobic glycolysis by targeting GAPDH to promote cuproptosis in colorectal cancer. Biomed Pharmacother, 2023, 159: 114301
CrossRef Google scholar
[189]
TsvetkovP, DetappeA, CaiK, et al.. Mitochondrial metabolism promotes adaptation to proteotoxic stress. Nat Chem Biol, 2019, 15(7): 681-689
CrossRef Google scholar
[190]
SolmonsonA, DeBerardinisRJ. Lipoic acid metabolism and mitochondrial redox regulation. J Biol Chem, 2018, 293(20): 7522-7530
CrossRef Google scholar
[191]
LutsenkoS. Atp7b−/− mice as a model for studies of Wilson’s disease. Biochem Soc Trans, 2008, 36: 1233-1238 Pt 6
CrossRef Google scholar
[192]
LingP, YangP, GaoX, et al.. ROS generation strategy based on biomimetic nanosheets by self-assembly of nanozymes. J Mater Chem B, 2022, 10(46): 9607-9612
CrossRef Google scholar
[193]
XueJ, YangG, DingH, et al.. Role of NSC319726 in ovarian cancer based on the bioinformatics analyses. Onco Targets Ther, 2015, 8: 3757-3765
[194]
ShimadaK, ReznikE, StokesME, et al.. Copper-Binding Small Molecule Induces Oxidative Stress and Cell-Cycle Arrest in Glioblastoma-Patient-Derived Cells. Cell Chem Biol, 2018, 25(5): 585-594
CrossRef Google scholar
[195]
PolishchukEV, MerollaA, LichtmanneggerJ, 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
CrossRef Google scholar
[196]
GuoJ, ChengJ, ZhengN, et al.. Copper Promotes Tumorigenesis by Activating the PDK1-AKT Oncogenic Pathway in a Copper Transporter 1 Dependent Manner. Adv Sci (Weinh), 2021, 8(18): e2004303
CrossRef Google scholar
[197]
ChenGH, LvW, XuYH, et al.. Functional analysis of MTF-1 and MT promoters and their transcriptional response to zinc (Zn) and copper (Cu) in yellow catfish Pelteobagrus fulvidraco. Chemosphere, 2020, 246: 125792
CrossRef Google scholar
[198]
HuW, ZhangC, WuR, et al.. Glutaminase 2, a novel p53 target gene regulating energy metabolism and antioxidant function. Proc Natl Acad Sci U S A, 2010, 107(16): 7455-7460
CrossRef Google scholar
[199]
LiuJ, LiuY, WangY, et al.. HMGB1 is a mediator of cuproptosis-related sterile inflammation. Front Cell Dev Biol, 2022, 10: 996307
CrossRef Google scholar
[200]
LuH, ZhouL, ZhangB, et al.. Cuproptosis key gene FDX1 is a prognostic biomarker and associated with immune infiltration in glioma. Front Med (Lausanne), 2022, 9: 939776
CrossRef Google scholar
[201]
ZhouY, XiaoD, JiangX, et al.. EREG is the core oncoimmunological biomarker of cuproptosis and mediates the cross-talk between VEGF and CD99 signaling in glioblastoma. J Transl Med, 2023, 21(1): 28
CrossRef Google scholar
[202]
LiY, YangJ, ZhangQ, et al.. Copper ionophore elesclomol selectively targets GNAQ/11-mutant uveal melanoma. Oncogene, 2022, 41(27): 3539-3553
CrossRef Google scholar
[203]
LvH, LiuX, ZengX, et al.. Comprehensive Analysis of Cuproptosis-Related Genes in Immune Infiltration and Prognosis in Melanoma. Front Pharmacol, 2022, 13: 930041
CrossRef Google scholar
[204]
XuS, LiuD, ChangT, et al.. Cuproptosis-Associated lncRNA Establishes New Prognostic Profile and Predicts Immunotherapy Response in Clear Cell Renal Cell Carcinoma. Front Genet, 2022, 13: 938259
CrossRef Google scholar
[205]
ZhangZ, ZengX, WuY, et al.. Cuproptosis-Related Risk Score Predicts Prognosis and Characterizes the Tumor Microenvironment in Hepatocellular Carcinoma. Front Immunol, 2022, 13: 925618
CrossRef Google scholar
[206]
YanC, NiuY, MaL, et al.. System analysis based on the cuproptosis-related genes identifies LIPT 1 as a novel therapy target for liver hepatocellular carcinoma. J Transl Med, 2022, 20(1): 452
CrossRef Google scholar
[207]
BaoJH, LuWC, DuanH, et al.. Identification of a novel cuproptosis-related gene signature and integrative analyses in patients with lower-grade gliomas. Front Immunol, 2022, 13: 933973
CrossRef Google scholar
[208]
LiuH, TangT. Pan-cancer genetic analysis of cuproptosis and copper metabolism-related gene set. Front Oncol, 2022, 12: 952290
CrossRef Google scholar
[209]
GuoB, YangF, ZhangL, 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
CrossRef Google scholar
[210]
XuY, LiuSY, ZengL, et al.. An Enzyme-Engineered Nonporous Copper(I) Coordination Polymer Nanoplat form for Cuproptosis-Based Synergistic Cancer Therapy. Adv Mater, 2022, 34(43): e2204733
CrossRef Google scholar
[211]
LiT, WangD, MengM, et al.. Copper-Coordinated Covalent Organic Framework Produced a Robust Fenton-Like Effect Inducing Immunogenic Cell Death of Tumors. Macromol Rapid Commun, 2023, 44(11): e2200929
CrossRef Google scholar
[212]
CollinsJF, ProhaskaJR, KnutsonMD. Metabolic crossroads of iron and copper. Nutr Rev, 2010, 68(3): 133-147
CrossRef Google scholar
[213]
JhelumP, DavidS. Ferroptosis: copper-iron connection in cuprizone-induced demyelination. Neural Regen Res, 2022, 17(1): 89-90
CrossRef Google scholar
[214]
GulecS, CollinsJF. Molecular mediators governing iron-copper interactions. Annu Rev Nutr, 2014, 34: 95-116
CrossRef Google scholar
[215]
TangD, ChenX, KangR, et al.. Ferroptosis: molecular mechanisms and health implications. Cell Res, 2021, 31(2): 107-125
CrossRef Google scholar
[216]
JiangX, StockwellBR, ConradM. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol, 2021, 22(4): 266-282
CrossRef Google scholar
[217]
FlemingMD, TrenorCC3rd, SuMA, et al.. Microcytic anaemia mice have a mutation in Nramp2, a candidate iron transporter gene. Nat Genet, 1997, 16(4): 383-386
CrossRef Google scholar
[218]
PatelBN, DavidS. Anovel glycosylphosphatidylinositol-anchored form of ceruloplasmin is expressed by mammalian astrocytes. J Biol Chem, 1997, 272(32): 20185-20190
CrossRef Google scholar
[219]
MastrogiannakiM, MatakP, KeithB, et al.. HIF-2alpha, but not HIF-1alpha, promotes iron absorption in mice. J Clin Invest, 2009, 119(5): 1159-1166
CrossRef Google scholar
[220]
RaviaJJ, StephenRM, GhishanFK, et al.. Menkes Copper ATPase (Atp7a) is a novel metal-responsive gene in rat duodenum, and immunoreactive protein is present on brush-border and basolateral membrane domains. J Biol Chem, 2005, 280(43): 36221-36227
CrossRef Google scholar
[221]
HaJH, DoguerC, CollinsJF. Consumption of a High-Iron Diet Disrupts Homeostatic Regulation of Intestinal Copper Absorption in Adolescent Mice. Am J Physiol Gastrointest Liver Physiol, 2017, 313(4): G535-G360
CrossRef Google scholar
[222]
JhelumP, Santos-NogueiraE, TeoW, et al.. Ferroptosis Mediates Cuprizone-Induced Loss of Oligodendrocytes and Demyelination. J Neurosci, 2020, 40(48): 9327-9341
CrossRef Google scholar
[223]
YangM, WuX, HuJ, et al.. COMMD10 inhibits HIFlα/CP loop to enhance ferroptosis and radiosensitivity by disrupting Cu-Fe balance in hepatocellular carcinoma. J Hepatol, 2022, 76(5): 1138-1150
CrossRef Google scholar
[224]
RenX, LiY, ZhouY, et al.. Overcoming the compensatory elevation of NRF2 renders hepatocellular carcinoma cells more vulnerable to disulfiram/copper-induced ferroptosis. Redox Biol, 2021, 46: 102122
CrossRef Google scholar
[225]
XueQ, YanD, ChenX, et al.. Copper-dependent autophagic degradation of GPX4 drives ferroptosis. Autophagy, 2023, 19(7): 1982-1996
CrossRef Google scholar
[226]
ShenY, LiD, LiangQ, et al.. Cross-talk between cuproptosis and ferroptosis regulators defines the tumor microenvironment for the prediction of prognosis and therapies in lung adenocarcinoma. Front Immunol, 2023, 13: 1029092
CrossRef Google scholar
[227]
LiY, WangRY, DengYJ, et al.. Molecular characteristics, clinical significance, and cancer immune interactions of cuproptosis and ferroptosis-associated genes in colorectal cancer. Front Oncol, 2022, 12: 975859
CrossRef Google scholar
[228]
ZhaoC, ZhangZ, JingT. A novel signature of combing cuproptosis-with ferroptosis-related genes for prediction of prognosis, immunologic therapy responses and drug sensitivity in hepatocellular carcinoma. Front Oncol, 2022, 12: 1000993
CrossRef Google scholar
[229]
GromadzkaG, TarnackaB, FlagaA, et al.. Copper Dyshomeostasis in Neurodegenerative Diseases-Therapeutic Implications. Int J Mol Sci, 2020, 21(23): 9259
CrossRef Google scholar
[230]
ChenL, MinJ, WangF. Copper homeostasis and cuproptosis in health and disease. Signal Transduct Target Ther, 2022, 7(1): 378
CrossRef Google scholar
[231]
ZlaticSA, Vrailas-MortimerA, GokhaleA, et al.. Rare Disease Mechanisms Identified by Genealogical Proteomics of Copper Homeostasis Mutant Pedigrees. Cell Syst, 2018, 6(3): 368-380
CrossRef Google scholar
[232]
BakkarN, StarrA, RabichowBE, et al.. The M1311V variant of ATP7A is associated with impaired trafficking and copper homeostasis in models of motor neuron disease. Neurobiol Dis, 2021, 149: 105228
CrossRef Google scholar
[233]
HartwigC, MéndezGM, BhattacharjeeS, et al.. Golgi-Dependent Copper Homeostasis Sustains Synaptic Development and Mitochondrial Content. J Neurosci, 2021, 41(2): 215-233
CrossRef Google scholar
[234]
ChoiBY, JangBG, KimJH, et al.. Copper/zinc chelation by clioquinol reduces spinal cord white matter damage and behavioral deficits in a murine MOG-induced multiple sclerosis model. Neurobiol Dis, 2013, 54: 382-391
CrossRef Google scholar
[235]
LaiY, LinC, LinX, et al.. Identification and immunological characterization of cuproptosis-related molecular clusters in Alzheimer’s disease. Front Aging Neurosci, 2022, 14: 932676
CrossRef Google scholar
[236]
GawandeMB, GoswamiA, FelpinFX, et al.. Cu and Cu-Based Nanoparticles: Synthesis and Applications in Catalysis. Chem Rev, 2016, 116(6): 3722-3811
CrossRef Google scholar
[237]
VermaN, KumarN. Synthesis and Biomedical Applications of Copper Oxide Nanoparticles: An Expanding Horizon. ACS Biomater Sci Eng, 2019, 5(3): 1170-1188
CrossRef Google scholar
[238]
ManiVM, KalaivaniS, SabarathinamS, et al.. Copper oxide nanoparticles synthesized from an endophytic fungus Aspergillus terreus: Bioactivity and anti-cancer evaluations. Environ Res, 2021, 201: 111502
CrossRef Google scholar
[239]
ImaniSM, LadouceurL, MarshallT, et al.. Antimicrobial Nanomaterials and Coatings: Current Mechanisms and Future Perspectives to Control the Spread of Viruses Including SARS-CoV-2. ACS Nano, 2020, 14(10): 12341-12369
CrossRef Google scholar
[240]
BrewerGJ. Copper-2 Hypothesis for Causation of the Current Alzheimer’s Disease Epidemic Together with Dietary Changes That Enhance the Epidemic. Chem Res Toxicol, 2017, 30(3): 763-768
CrossRef Google scholar
[241]
McCannCJ, JayakanthanS, SiottoM, et al.. Single nucleotide polymorphisms in the human ATP7B gene modify the properties of the ATP7B protein. Metallomics, 2019, 11(6): 1128-1139
CrossRef Google scholar
[242]
CliffordRJ, MaryonEB, KaplanJH. Dynamic internalization and recycling of a metal ion transporter: Cu homeostasis and CTR1, the human Cu+ uptake system. J Cell Sci, 2016, 129(8): 1711-1721
[243]
NarindrasorasakS, KulkarniP, DeschampsP, et al.. Characterization and copper binding properties of human COMMD1 (MURR1). Biochemistry, 2007, 46(11): 3116-3128
CrossRef Google scholar
[244]
HuXM, ZhengSY, ZhangQ, et al.. PANoptosis signaling enables broad immune response in psoriasis: From pathogenesis to new therapeutic strategies. Comput Struct Biotechnol J, 2023, 23: 64-76
CrossRef Google scholar
[245]
YangGJ, LiuH, MaDL, et al.. Rebalancing metal dyshomeostasis for Alzheimer’s disease therapy. J Biol Inorg Chem, 2019, 24(8): 1159-1170
CrossRef Google scholar
[246]
TulinskaJ, MikusovaML, LiskovaA, et al.. Copper Oxide Nanoparticles Stimulate the Immune Response and Decrease Antioxidant Defense in Mice After Six-Week Inhalation. Front Immunol, 2022, 13: 874253
CrossRef Google scholar
[247]
StamenkovićS, DučićT, StamenkovićV, et al.. Imaging of glial cell morphology, SOD1 distribution and elemental composition in the brainstem and hippocampus of the ALS hSOD1G93A rat. Neuroscience, 2017, 357: 37-55
CrossRef Google scholar
[248]
WenMH, XieX, TuJ, et al.. Generation of a genetically modified human embryonic stem cells expressing fluorescence tagged ATOX1. Stem Cell Res, 2019, 41: 101631
CrossRef Google scholar
[249]
ChenH, XieX, ChenTY. Single-molecule microscopy for in-cell quantification of protein oligomeric stoichiometry. Curr Opin Struct Biol, 2021, 66: 112-118
CrossRef Google scholar
[250]
GuptaD, BhattacharjeeO, MandalD, et al.. CRISPR-Cas9 system: A new-fangled dawn in gene editing. Life Sci, 2019, 232: 116636
CrossRef Google scholar
[251]
WangX, ZhouM, LiuY, et al.. Cope with copper: From copper linked mechanisms to copper-based clinical cancer therapies. Cancer Lett, 2023, 561: 216157
CrossRef Google scholar
[252]
ParpuraV, HenekaMT, MontanaV, et al.. Glial cells in (patho)physiology. J Neurochem, 2012, 121(1): 4-27
CrossRef Google scholar
[253]
XuMB, RongPQ, JinTY, et al.. Chinese Herbal Medicine for Wilson’s Disease: A Systematic Review and Meta-Analysis. Front Pharmacol, 2019, 10: 277
CrossRef Google scholar
[254]
SimunkovaM, AlwaselSH, AlhazzaIM, et al.. Management of oxidative stress and other pathologies in Alzheimer’s disease. Arch Toxicol, 2019, 93(9): 2491-2513
CrossRef Google scholar
[255]
AiroldiC, La FerlaB, D OrazioG, et al.. Flavonoids in the Treatment of Alzheimer’s and Other Neurodegenerative Diseases. Curr Med Chem, 2018, 25(27): 3228-3246
CrossRef Google scholar
[256]
WangD, TianZ, ZhangP, et al.. The molecular mechanisms of cuproptosis and its relevance to cardiovascular disease. Biomed Pharmacother, 2023, 163: 114830
CrossRef Google scholar
PDF

110

Accesses

14

Citations

5

Altmetric

Detail

Sections
Recommended

/