Skip to main content
Top
Published in: Translational Neurodegeneration 1/2020

01-12-2020 | Alzheimer's Disease | Review

Current understanding of metal ions in the pathogenesis of Alzheimer’s disease

Authors: Lu Wang, Ya-Ling Yin, Xin-Zi Liu, Peng Shen, Yan-Ge Zheng, Xin-Rui Lan, Cheng-Biao Lu, Jian-Zhi Wang

Published in: Translational Neurodegeneration | Issue 1/2020

Login to get access

Abstract

Background

The homeostasis of metal ions, such as iron, copper, zinc and calcium, in the brain is crucial for maintaining normal physiological functions. Studies have shown that imbalance of these metal ions in the brain is closely related to the onset and progression of Alzheimer’s disease (AD), the most common neurodegenerative disorder in the elderly.

Main body

Erroneous deposition/distribution of the metal ions in different brain regions induces oxidative stress. The metal ions imbalance and oxidative stress together or independently promote amyloid-β (Aβ) overproduction by activating β- or γ-secretases and inhibiting α-secretase, it also causes tau hyperphosphorylation by activating protein kinases, such as glycogen synthase kinase-3β (GSK-3β), cyclin-dependent protein kinase-5 (CDK5), mitogen-activated protein kinases (MAPKs), etc., and inhibiting protein phosphatase 2A (PP2A). The metal ions imbalances can also directly or indirectly disrupt organelles, causing endoplasmic reticulum (ER) stress; mitochondrial and autophagic dysfunctions, which can cause or aggravate Aβ and tau aggregation/accumulation, and impair synaptic functions. Even worse, the metal ions imbalance-induced alterations can reversely exacerbate metal ions misdistribution and deposition. The vicious cycles between metal ions imbalances and Aβ/tau abnormalities will eventually lead to a chronic neurodegeneration and cognitive deficits, such as seen in AD patients.

Conclusion

The metal ions imbalance induces Aβ and tau pathologies by directly or indirectly affecting multiple cellular/subcellular pathways, and the disrupted homeostasis can reversely aggravate the abnormalities of metal ions transportation/deposition. Therefore, adjusting metal balance by supplementing or chelating the metal ions may be potential in ameliorating AD pathologies, which provides new research directions for AD treatment.
Literature
2.
go back to reference Grundke-Iqbal I, Iqbal K, Tung YC, Wisniewski HM. Alzheimer paired helical filaments: immunochemical identification of polypeptides. Acta Neuropathol. 1984;62(4):259–67.PubMedCrossRef Grundke-Iqbal I, Iqbal K, Tung YC, Wisniewski HM. Alzheimer paired helical filaments: immunochemical identification of polypeptides. Acta Neuropathol. 1984;62(4):259–67.PubMedCrossRef
3.
go back to reference Elmaleh DR, Farlow MR, Conti PS, Tompkins RG, Kundakovic L, Tanzi RE. Developing effective Alzheimer's disease therapies: clinical experience and future directions. J Alzheimers Dis. 2019;71(3):715–32.PubMedPubMedCentralCrossRef Elmaleh DR, Farlow MR, Conti PS, Tompkins RG, Kundakovic L, Tanzi RE. Developing effective Alzheimer's disease therapies: clinical experience and future directions. J Alzheimers Dis. 2019;71(3):715–32.PubMedPubMedCentralCrossRef
4.
go back to reference Dubois B, Hampel H, Feldman HH, Scheltens P, Aisen P, Andrieu S, et al. Preclinical Alzheimer's disease: definition, natural history, and diagnostic criteria. Alzheimers Dement. 2016;12(3):292–323.PubMedPubMedCentralCrossRef Dubois B, Hampel H, Feldman HH, Scheltens P, Aisen P, Andrieu S, et al. Preclinical Alzheimer's disease: definition, natural history, and diagnostic criteria. Alzheimers Dement. 2016;12(3):292–323.PubMedPubMedCentralCrossRef
5.
go back to reference Yin Y, Gao D, Wang Y, Wang ZH, Wang X, Ye J, et al. Tau accumulation induces synaptic impairment and memory deficit by calcineurin-mediated inactivation of nuclear CaMKIV/CREB signaling. Proc Natl Acad Sci U S A. 2016;113(26):E3773–81.PubMedPubMedCentralCrossRef Yin Y, Gao D, Wang Y, Wang ZH, Wang X, Ye J, et al. Tau accumulation induces synaptic impairment and memory deficit by calcineurin-mediated inactivation of nuclear CaMKIV/CREB signaling. Proc Natl Acad Sci U S A. 2016;113(26):E3773–81.PubMedPubMedCentralCrossRef
6.
go back to reference Luo HB, Xia YY, Shu XJ, Liu ZC, Feng Y, Liu XH, et al. SUMOylation at K340 inhibits tau degradation through deregulating its phosphorylation and ubiquitination. Proc Natl Acad Sci U S A. 2014;111(46):16586–91.PubMedPubMedCentralCrossRef Luo HB, Xia YY, Shu XJ, Liu ZC, Feng Y, Liu XH, et al. SUMOylation at K340 inhibits tau degradation through deregulating its phosphorylation and ubiquitination. Proc Natl Acad Sci U S A. 2014;111(46):16586–91.PubMedPubMedCentralCrossRef
7.
go back to reference Wang JZ, Grundke-Iqbal I, Iqbal K. Glycosylation of microtubule-associated protein tau: an abnormal posttranslational modification in Alzheimer's disease. Nat Med. 1996;2(8):871–5.PubMedCrossRef Wang JZ, Grundke-Iqbal I, Iqbal K. Glycosylation of microtubule-associated protein tau: an abnormal posttranslational modification in Alzheimer's disease. Nat Med. 1996;2(8):871–5.PubMedCrossRef
8.
go back to reference Wang JZ, Liu F. Microtubule-associated protein tau in development, degeneration and protection of neurons. Prog Neurobiol. 2008;85(2):148–75.PubMedCrossRef Wang JZ, Liu F. Microtubule-associated protein tau in development, degeneration and protection of neurons. Prog Neurobiol. 2008;85(2):148–75.PubMedCrossRef
9.
go back to reference Wang JZ, Wang ZH, Tian Q. Tau hyperphosphorylation induces apoptotic escape and triggers neurodegeneration in Alzheimer's disease. Neurosci Bull. 2014;30(2):359–66.PubMedPubMedCentralCrossRef Wang JZ, Wang ZH, Tian Q. Tau hyperphosphorylation induces apoptotic escape and triggers neurodegeneration in Alzheimer's disease. Neurosci Bull. 2014;30(2):359–66.PubMedPubMedCentralCrossRef
10.
go back to reference Wang JZ, Gao X, Wang ZH. The physiology and pathology of microtubule-associated protein tau. Essays Biochem. 2014;56:111–23.PubMedCrossRef Wang JZ, Gao X, Wang ZH. The physiology and pathology of microtubule-associated protein tau. Essays Biochem. 2014;56:111–23.PubMedCrossRef
11.
go back to reference Wang JZ, Wang ZH. Senescence may mediate conversion of tau phosphorylation-induced apoptotic escape to neurodegeneration. Exp Gerontol. 2015;68:82–6.PubMedCrossRef Wang JZ, Wang ZH. Senescence may mediate conversion of tau phosphorylation-induced apoptotic escape to neurodegeneration. Exp Gerontol. 2015;68:82–6.PubMedCrossRef
13.
go back to reference Feng Q, Luo Y, Zhang XN, Yang XF, Hong XY, Sun DS, et al. MAPT/tau accumulation represses autophagy flux by disrupting IST1-regulated ESCRT-III complex formation: a vicious cycle in Alzheimer neurodegeneration. Autophagy. 2019;28:1–18. Feng Q, Luo Y, Zhang XN, Yang XF, Hong XY, Sun DS, et al. MAPT/tau accumulation represses autophagy flux by disrupting IST1-regulated ESCRT-III complex formation: a vicious cycle in Alzheimer neurodegeneration. Autophagy. 2019;28:1–18.
14.
go back to reference Li XG, Hong XY, Wang YL, Zhang SJ, Zhang JF, Li XC, et al. Tau accumulation triggers STAT1-dependent memory deficits by suppressing NMDA receptor expression. EMBO Rep. 2019;20(6). Li XG, Hong XY, Wang YL, Zhang SJ, Zhang JF, Li XC, et al. Tau accumulation triggers STAT1-dependent memory deficits by suppressing NMDA receptor expression. EMBO Rep. 2019;20(6).
15.
go back to reference Liu E, Zhou Q, Xie AJ, Li M, Zhang S, Huang H, et al. Enriched gestation activates the IGF pathway to evoke embryo-adult benefits to prevent Alzheimer's disease. Transl Neurodegener. 2019;8:8.PubMedPubMedCentralCrossRef Liu E, Zhou Q, Xie AJ, Li M, Zhang S, Huang H, et al. Enriched gestation activates the IGF pathway to evoke embryo-adult benefits to prevent Alzheimer's disease. Transl Neurodegener. 2019;8:8.PubMedPubMedCentralCrossRef
16.
go back to reference Guo J, Xu C, Ni S, Zhang S, Li Q, Zeng P, et al. Elevation of pS262-tau and Demethylated PP2A in retina occurs earlier than in Hippocampus during Hyperhomocysteinemia. J Alzheimers Dis. 2019;68(1):367–81.PubMedCrossRef Guo J, Xu C, Ni S, Zhang S, Li Q, Zeng P, et al. Elevation of pS262-tau and Demethylated PP2A in retina occurs earlier than in Hippocampus during Hyperhomocysteinemia. J Alzheimers Dis. 2019;68(1):367–81.PubMedCrossRef
17.
go back to reference Xu ZP, Yang SL, Zhao S, Zheng CH, Li HH, Zhang Y, et al. Biomarkers for early diagnostic of mild cognitive impairment in Type-2 diabetes patients: a multicentre, retrospective, Nested Case-Control Study. EBioMedicine. 2016;5:105–13.PubMedPubMedCentralCrossRef Xu ZP, Yang SL, Zhao S, Zheng CH, Li HH, Zhang Y, et al. Biomarkers for early diagnostic of mild cognitive impairment in Type-2 diabetes patients: a multicentre, retrospective, Nested Case-Control Study. EBioMedicine. 2016;5:105–13.PubMedPubMedCentralCrossRef
18.
go back to reference Zhang Y, Xie JZ, Xu XY, Hu J, Xu T, Jin S, et al. Liraglutide ameliorates Hyperhomocysteinemia-induced Alzheimer-like pathology and memory deficits in rats via multi-molecular targeting. Neurosci Bull. 2019;35(4):724–34.PubMedCrossRefPubMedCentral Zhang Y, Xie JZ, Xu XY, Hu J, Xu T, Jin S, et al. Liraglutide ameliorates Hyperhomocysteinemia-induced Alzheimer-like pathology and memory deficits in rats via multi-molecular targeting. Neurosci Bull. 2019;35(4):724–34.PubMedCrossRefPubMedCentral
20.
go back to reference Drayer B, Burger P, Darwin R, Riederer S, Herfkens R, Johnson GA. MRI of brain iron. AJR Am J Roentgenol. 1986;147(1):103–10.PubMedCrossRef Drayer B, Burger P, Darwin R, Riederer S, Herfkens R, Johnson GA. MRI of brain iron. AJR Am J Roentgenol. 1986;147(1):103–10.PubMedCrossRef
21.
go back to reference Moos T, Trinder D, Morgan EH. Cellular distribution of ferric iron, ferritin, transferrin and divalent metal transporter 1 (DMT1) in substantia nigra and basal ganglia of normal and beta2-microglobulin deficient mouse brain. Cell Mol Biol (Noisy-le-Grand). 2000;46(3):549–61. Moos T, Trinder D, Morgan EH. Cellular distribution of ferric iron, ferritin, transferrin and divalent metal transporter 1 (DMT1) in substantia nigra and basal ganglia of normal and beta2-microglobulin deficient mouse brain. Cell Mol Biol (Noisy-le-Grand). 2000;46(3):549–61.
22.
go back to reference Morris CM, Candy JM, Oakley AE, Bloxham CA, Edwardson JA. Histochemical distribution of non-haem iron in the human brain. Acta Anat. 1992;144(3):235–57.PubMedCrossRef Morris CM, Candy JM, Oakley AE, Bloxham CA, Edwardson JA. Histochemical distribution of non-haem iron in the human brain. Acta Anat. 1992;144(3):235–57.PubMedCrossRef
24.
go back to reference Paoletti P, Vergnano AM, Barbour B, Casado M. Zinc at glutamatergic synapses. Neuroscience. 2009;158(1):126–36.PubMedCrossRef Paoletti P, Vergnano AM, Barbour B, Casado M. Zinc at glutamatergic synapses. Neuroscience. 2009;158(1):126–36.PubMedCrossRef
25.
go back to reference Sensi SL, Paoletti P, Bush AI, Sekler I. Zinc in the physiology and pathology of the CNS. Nat Rev Neurosci. 2009;10(11):780–91.PubMedCrossRef Sensi SL, Paoletti P, Bush AI, Sekler I. Zinc in the physiology and pathology of the CNS. Nat Rev Neurosci. 2009;10(11):780–91.PubMedCrossRef
26.
go back to reference Olesen RH, Hyde TM, Kleinman JE, Smidt K, Rungby J, Larsen A. Obesity and age-related alterations in the gene expression of zinc-transporter proteins in the human brain. Transl Psychiatry. 2016;6(6):e838.PubMedPubMedCentralCrossRef Olesen RH, Hyde TM, Kleinman JE, Smidt K, Rungby J, Larsen A. Obesity and age-related alterations in the gene expression of zinc-transporter proteins in the human brain. Transl Psychiatry. 2016;6(6):e838.PubMedPubMedCentralCrossRef
27.
go back to reference Davies KM, Hare DJ, Cottam V, Chen N, Hilgers L, Halliday G, et al. Localization of copper and copper transporters in the human brain. Metallomics. 2013;5(1):43–51.PubMedCrossRef Davies KM, Hare DJ, Cottam V, Chen N, Hilgers L, Halliday G, et al. Localization of copper and copper transporters in the human brain. Metallomics. 2013;5(1):43–51.PubMedCrossRef
28.
go back to reference Rembach A, Hare DJ, Lind M, Fowler CJ, Cherny RA, McLean C, et al. Decreased copper in Alzheimer’s disease brain is predominantly in the soluble extractable fraction. Int J Alzheimers Dis. 2013;2013:623241.PubMedPubMedCentral Rembach A, Hare DJ, Lind M, Fowler CJ, Cherny RA, McLean C, et al. Decreased copper in Alzheimer’s disease brain is predominantly in the soluble extractable fraction. Int J Alzheimers Dis. 2013;2013:623241.PubMedPubMedCentral
29.
go back to reference Xu J, Church SJ, Patassini S, Begley P, Waldvogel HJ, Curtis MA, et al. Evidence for widespread, severe brain copper deficiency in Alzheimer's dementia. Metallomics. 2017;9(8):1106–19.PubMedCrossRef Xu J, Church SJ, Patassini S, Begley P, Waldvogel HJ, Curtis MA, et al. Evidence for widespread, severe brain copper deficiency in Alzheimer's dementia. Metallomics. 2017;9(8):1106–19.PubMedCrossRef
30.
go back to reference Alzheimer's Association Calcium Hypothesis Workgroup. Calcium Hypothesis of Alzheimer's disease and brain aging: A framework for integrating new evidence into a comprehensive theory of pathogenesis. Alzheimers Dement. 2017;13(2):178–82 e17.CrossRef Alzheimer's Association Calcium Hypothesis Workgroup. Calcium Hypothesis of Alzheimer's disease and brain aging: A framework for integrating new evidence into a comprehensive theory of pathogenesis. Alzheimers Dement. 2017;13(2):178–82 e17.CrossRef
31.
go back to reference van Duijn S, Bulk M, van Duinen SG, Nabuurs RJA, van Buchem MA, van der Weerd L, et al. Cortical Iron Reflects Severity of Alzheimer’s Disease. J Alzheimers Dis. 2017;60(4):1533–45.PubMedPubMedCentralCrossRef van Duijn S, Bulk M, van Duinen SG, Nabuurs RJA, van Buchem MA, van der Weerd L, et al. Cortical Iron Reflects Severity of Alzheimer’s Disease. J Alzheimers Dis. 2017;60(4):1533–45.PubMedPubMedCentralCrossRef
32.
go back to reference Liu B, Moloney A, Meehan S, Morris K, Thomas SE, Serpell LC, et al. Iron promotes the toxicity of amyloid beta peptide by impeding its ordered aggregation. J Biol Chem. 2011;286(6):4248–56.PubMedCrossRef Liu B, Moloney A, Meehan S, Morris K, Thomas SE, Serpell LC, et al. Iron promotes the toxicity of amyloid beta peptide by impeding its ordered aggregation. J Biol Chem. 2011;286(6):4248–56.PubMedCrossRef
33.
go back to reference Rogers JT, Xia N, Wong A, Bakshi R, Cahill CM. Targeting the Iron-Response Elements of the mRNAs for the Alzheimer's Amyloid Precursor Protein and Ferritin to Treat Acute Lead and Manganese Neurotoxicity. Int J Mol Sci. 2019;20(4). Rogers JT, Xia N, Wong A, Bakshi R, Cahill CM. Targeting the Iron-Response Elements of the mRNAs for the Alzheimer's Amyloid Precursor Protein and Ferritin to Treat Acute Lead and Manganese Neurotoxicity. Int J Mol Sci. 2019;20(4).
34.
35.
go back to reference Wärmländer SKTS, Österlund N, Wallin C, Wu J, Luo J, Tiiman A, et al. Metal binding to the amyloid-β peptides in the presence of biomembranes: potential mechanisms of cell toxicity. J Biol Inorg Chem. 2019;24(8):1189–96.PubMedCrossRef Wärmländer SKTS, Österlund N, Wallin C, Wu J, Luo J, Tiiman A, et al. Metal binding to the amyloid-β peptides in the presence of biomembranes: potential mechanisms of cell toxicity. J Biol Inorg Chem. 2019;24(8):1189–96.PubMedCrossRef
36.
go back to reference Banerjee P, Sahoo A, Anand S, Ganguly A, Righi G, et al. Multiple mechanisms of iron-induced amyloid beta-peptide accumulation in SHSY5Y cells: protective action of negletein. NeuroMolecular Med. 2014;16(4):787–98.PubMedCrossRef Banerjee P, Sahoo A, Anand S, Ganguly A, Righi G, et al. Multiple mechanisms of iron-induced amyloid beta-peptide accumulation in SHSY5Y cells: protective action of negletein. NeuroMolecular Med. 2014;16(4):787–98.PubMedCrossRef
37.
go back to reference Huang XT, Qian ZM, He X, Gong Q, Wu KC, et al. Reducing iron in the brain: a novel pharmacologic mechanism of huperzine A in the treatment of Alzheimer's disease. Neurobiol Aging. 2014;35(5):1045–54.PubMedCrossRef Huang XT, Qian ZM, He X, Gong Q, Wu KC, et al. Reducing iron in the brain: a novel pharmacologic mechanism of huperzine A in the treatment of Alzheimer's disease. Neurobiol Aging. 2014;35(5):1045–54.PubMedCrossRef
39.
go back to reference Bandyopadhyay S, Rogers JT. Alzheimer's disease therapeutics targeted to the control of amyloidprecursor protein translation: maintenance of brain iron homeostasis. Biochem Pharmacol. 2014;88(4):486–94.PubMedPubMedCentralCrossRef Bandyopadhyay S, Rogers JT. Alzheimer's disease therapeutics targeted to the control of amyloidprecursor protein translation: maintenance of brain iron homeostasis. Biochem Pharmacol. 2014;88(4):486–94.PubMedPubMedCentralCrossRef
40.
go back to reference Rogers JT, Venkataramani V, Washburn C, Liu Y, Tummala V, Jiang H, et al. A role for amyloid precursor protein translation to restore iron homeostasis and ameliorate lead (Pb) neurotoxicity. J Neurochem. 2016;138(3):479–94.PubMedCrossRef Rogers JT, Venkataramani V, Washburn C, Liu Y, Tummala V, Jiang H, et al. A role for amyloid precursor protein translation to restore iron homeostasis and ameliorate lead (Pb) neurotoxicity. J Neurochem. 2016;138(3):479–94.PubMedCrossRef
41.
go back to reference Belaidi AA, Gunn AP, Wong BX, Ayton S, Appukuttan AT, Roberts BR, et al. Marked Age-Related Changes in Brain Iron Homeostasis in Amyloid Protein Precursor Knockout Mice. Neurotherapeutics. 2018;15(4):1055–62.PubMedPubMedCentralCrossRef Belaidi AA, Gunn AP, Wong BX, Ayton S, Appukuttan AT, Roberts BR, et al. Marked Age-Related Changes in Brain Iron Homeostasis in Amyloid Protein Precursor Knockout Mice. Neurotherapeutics. 2018;15(4):1055–62.PubMedPubMedCentralCrossRef
42.
go back to reference Guo C, Wang P, Zhong ML, Wang T, Huang XS, Li JY, et al. Deferoxamine inhibits iron induced hippocampal tau phosphorylation in the Alzheimer transgenic mouse brain. Neurochem Int. 2013;62(2):165–72.PubMedCrossRef Guo C, Wang P, Zhong ML, Wang T, Huang XS, Li JY, et al. Deferoxamine inhibits iron induced hippocampal tau phosphorylation in the Alzheimer transgenic mouse brain. Neurochem Int. 2013;62(2):165–72.PubMedCrossRef
43.
go back to reference Muñoz P, Zavala G, Castillo K, Aguirre P, Hidalgo C, Núñez MT. Effect of iron on the activation of the MAPK/ERK pathway in PC12 neuroblastoma cells. BiolRes. 2006;39(1):189–90. Muñoz P, Zavala G, Castillo K, Aguirre P, Hidalgo C, Núñez MT. Effect of iron on the activation of the MAPK/ERK pathway in PC12 neuroblastoma cells. BiolRes. 2006;39(1):189–90.
44.
go back to reference Huang X, Dai J, Huang C, Zhang Q, Bhanot O, Pelle E. Deferoxamine synergistically enhances iron-mediated AP-1 activation: a showcase of the interplay between extracellular-signal-regulated kinase and tyrosine phosphatase. Free Radic Res. 2007;41(10):1135–42.PubMedCrossRef Huang X, Dai J, Huang C, Zhang Q, Bhanot O, Pelle E. Deferoxamine synergistically enhances iron-mediated AP-1 activation: a showcase of the interplay between extracellular-signal-regulated kinase and tyrosine phosphatase. Free Radic Res. 2007;41(10):1135–42.PubMedCrossRef
45.
go back to reference Egaña JT, Zambrano C, Nuñez MT, Gonzalez-Billault C, Maccioni RB. Iron-induced oxidative stress modififies tau phosphorylation patterns in hippocampal cell cultures. Biometals. 2003;16(1):215–23.PubMedCrossRef Egaña JT, Zambrano C, Nuñez MT, Gonzalez-Billault C, Maccioni RB. Iron-induced oxidative stress modififies tau phosphorylation patterns in hippocampal cell cultures. Biometals. 2003;16(1):215–23.PubMedCrossRef
46.
go back to reference Tuo QZ, Lei P, Jackman KA, Li XL, Xiong H, Li XL, et al. Tau-mediated iron export prevents ferroptotic damage after ischemic stroke. Mol Psychiatry. 2017;22(11):1520–30.PubMedCrossRef Tuo QZ, Lei P, Jackman KA, Li XL, Xiong H, Li XL, et al. Tau-mediated iron export prevents ferroptotic damage after ischemic stroke. Mol Psychiatry. 2017;22(11):1520–30.PubMedCrossRef
47.
go back to reference Matheou CJ, Younan ND, Viles JH. Cu2+ accentuates distinct misfolding of Aβ (1-40) and Aβ (1-42) peptides, and potentiates membrane disruption. BiochemJ. 2015;466(2):233–42.CrossRef Matheou CJ, Younan ND, Viles JH. Cu2+ accentuates distinct misfolding of Aβ (1-40) and Aβ (1-42) peptides, and potentiates membrane disruption. BiochemJ. 2015;466(2):233–42.CrossRef
48.
go back to reference Zhao J, Gao W, Yang Z, Li H, Gao Z. Nitration of amyloid-β peptide (1-42) as a protective mechanism for the amyloid-β peptide (1-42) against copper ion toxicity. J Inorg Biochem. 2019;190:15–23.PubMedCrossRef Zhao J, Gao W, Yang Z, Li H, Gao Z. Nitration of amyloid-β peptide (1-42) as a protective mechanism for the amyloid-β peptide (1-42) against copper ion toxicity. J Inorg Biochem. 2019;190:15–23.PubMedCrossRef
49.
go back to reference Multhaup G, Schlicksupp A, Hesse L, Beher D, Ruppert T, Masters CL. The amyloid precursor protein of Alzheimer’s disease in the reduction of copper (II) to copper(I). Science. 1996;271(5254):1406–9.PubMedCrossRef Multhaup G, Schlicksupp A, Hesse L, Beher D, Ruppert T, Masters CL. The amyloid precursor protein of Alzheimer’s disease in the reduction of copper (II) to copper(I). Science. 1996;271(5254):1406–9.PubMedCrossRef
50.
go back to reference Wang Z, Zhang YH, Zhang W, Gao HL, Zhong ML, Huang TT, et al. Copper chelators promote nonamyloidogenic processing of AβPP via MT1/2/CREB-dependent signaling pathways in AβPP/PS1 transgenic mice. J Pineal Res. 2018;65(3):e12502.PubMedCrossRef Wang Z, Zhang YH, Zhang W, Gao HL, Zhong ML, Huang TT, et al. Copper chelators promote nonamyloidogenic processing of AβPP via MT1/2/CREB-dependent signaling pathways in AβPP/PS1 transgenic mice. J Pineal Res. 2018;65(3):e12502.PubMedCrossRef
51.
go back to reference Sparks DL, Schreurs BG. Trace amounts of copper in water induce beta-amyloid plaques and learning deficits in a rabbit model of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2003;100(19):11065–9.PubMedPubMedCentralCrossRef Sparks DL, Schreurs BG. Trace amounts of copper in water induce beta-amyloid plaques and learning deficits in a rabbit model of Alzheimer’s disease. Proc Natl Acad Sci U S A. 2003;100(19):11065–9.PubMedPubMedCentralCrossRef
52.
go back to reference Kenche VB, Zawisza I, Masters CL, Bal W, Barnham KJ, Drew SC, et al. Mixed ligand Cu2+ complexes of a model therapeutic with Alzheimer's amyloid-β peptide and monoamine neurotransmitters. Inorg Chem. 2013;52(8):4303–18.PubMedCrossRef Kenche VB, Zawisza I, Masters CL, Bal W, Barnham KJ, Drew SC, et al. Mixed ligand Cu2+ complexes of a model therapeutic with Alzheimer's amyloid-β peptide and monoamine neurotransmitters. Inorg Chem. 2013;52(8):4303–18.PubMedCrossRef
53.
go back to reference Voss K, Harris C, Ralle M, Duffy M, Murchison C, Quinn JF. Modulation of tau phosphorylation by environmental copper. Transl Neurodegener. 2014;3(1):24.PubMedPubMedCentralCrossRef Voss K, Harris C, Ralle M, Duffy M, Murchison C, Quinn JF. Modulation of tau phosphorylation by environmental copper. Transl Neurodegener. 2014;3(1):24.PubMedPubMedCentralCrossRef
54.
go back to reference Heicklen-Klein A, Ginzburg I. Tau promoter confers neuronal specificity and binds Sp1 and AP-2. J Neurochem. 2000;75(4):1408.PubMedCrossRef Heicklen-Klein A, Ginzburg I. Tau promoter confers neuronal specificity and binds Sp1 and AP-2. J Neurochem. 2000;75(4):1408.PubMedCrossRef
55.
go back to reference James SA, Churches QI, de Jonge MD, Birchall IE, Streltsov V, McColl G, et al. Iron, copper, and zinc concentration in Aβ plaques in the APP/PS1 mouse model of Alzheimer's disease correlates with metal levels in the surrounding Neuropil. ACS Chem Neurosci. 2017;8(3):629–37.PubMedCrossRef James SA, Churches QI, de Jonge MD, Birchall IE, Streltsov V, McColl G, et al. Iron, copper, and zinc concentration in Aβ plaques in the APP/PS1 mouse model of Alzheimer's disease correlates with metal levels in the surrounding Neuropil. ACS Chem Neurosci. 2017;8(3):629–37.PubMedCrossRef
56.
go back to reference Miller Y, Ma B, Nussinov R. Zinc ions promote Alzheimer Abeta aggregation via population shift of polymorphic states. Proc Natl Acad Sci U S A. 2010;107(21):9490–5.PubMedPubMedCentralCrossRef Miller Y, Ma B, Nussinov R. Zinc ions promote Alzheimer Abeta aggregation via population shift of polymorphic states. Proc Natl Acad Sci U S A. 2010;107(21):9490–5.PubMedPubMedCentralCrossRef
57.
go back to reference Sciacca MFM, Kotler SA, Brender JR, Chen J, Lee DK, Ramamoorthy A. Two-step mechanism of membrane disruption by Aβ through membrane fragmentation and pore formation. Biophys J. 2012;103(4):702–10.PubMedPubMedCentralCrossRef Sciacca MFM, Kotler SA, Brender JR, Chen J, Lee DK, Ramamoorthy A. Two-step mechanism of membrane disruption by Aβ through membrane fragmentation and pore formation. Biophys J. 2012;103(4):702–10.PubMedPubMedCentralCrossRef
58.
go back to reference Seegar TCM, Killingsworth LB, Saha N, Meyer PA, Patra D, Zimmerman B, et al. Structural Basis for Regulated Proteolysis by the α-Secretase ADAM10. Cell. 2017;171(7):1638–48 e7.PubMedPubMedCentralCrossRef Seegar TCM, Killingsworth LB, Saha N, Meyer PA, Patra D, Zimmerman B, et al. Structural Basis for Regulated Proteolysis by the α-Secretase ADAM10. Cell. 2017;171(7):1638–48 e7.PubMedPubMedCentralCrossRef
60.
go back to reference An WL, Bjorkdahl C, Liu R, Cowburn RF, Winblad B, Pei JJ. Mechanism of zinc-induced phosphorylation of p70 S6 kinase and glycogen synthase kinase 3beta in SH-SY5Y neuroblastoma cells. J Neurochem. 2005;92(5):1104–15.PubMedCrossRef An WL, Bjorkdahl C, Liu R, Cowburn RF, Winblad B, Pei JJ. Mechanism of zinc-induced phosphorylation of p70 S6 kinase and glycogen synthase kinase 3beta in SH-SY5Y neuroblastoma cells. J Neurochem. 2005;92(5):1104–15.PubMedCrossRef
61.
go back to reference Kim I, Park EJ, Seo J, Ko SJ, Lee J, Kim CH. Zinc stimulates tau S214 phosphorylation by the activation of Raf/mitogen-activated protein kinase-kinase/extracellular signal-regulated kinase pathway. Neuroreport. 2011;22(16):839–44.PubMed Kim I, Park EJ, Seo J, Ko SJ, Lee J, Kim CH. Zinc stimulates tau S214 phosphorylation by the activation of Raf/mitogen-activated protein kinase-kinase/extracellular signal-regulated kinase pathway. Neuroreport. 2011;22(16):839–44.PubMed
62.
go back to reference Sun XY, Wei YP, Xiong Y, Wang XC, Xie AJ, Wang XL, et al. Synaptic released zinc promotes tau hyperphosphorylation by inhibition of protein phosphatase 2A (PP2A). J Biol Chem. 2012;287(14):11174–82.PubMedPubMedCentralCrossRef Sun XY, Wei YP, Xiong Y, Wang XC, Xie AJ, Wang XL, et al. Synaptic released zinc promotes tau hyperphosphorylation by inhibition of protein phosphatase 2A (PP2A). J Biol Chem. 2012;287(14):11174–82.PubMedPubMedCentralCrossRef
63.
go back to reference Xiong Y, Jing XP, Zhou XW, Wang XL, Yang Y, Sun XY, et al. Zinc induces protein phosphatase 2A inactivation and tau hyperphosphorylation through Src dependent PP2A (tyrosine 307) phosphorylation. Neurobiol Aging. 2013;34(3):745–56.PubMedCrossRef Xiong Y, Jing XP, Zhou XW, Wang XL, Yang Y, Sun XY, et al. Zinc induces protein phosphatase 2A inactivation and tau hyperphosphorylation through Src dependent PP2A (tyrosine 307) phosphorylation. Neurobiol Aging. 2013;34(3):745–56.PubMedCrossRef
65.
go back to reference Kuchibhotla KV, Goldman ST, Lattarulo CR, Wu HY, Hyman BT, Bacskai BJ. Abeta plaques lead to aberrant regulation of calcium homeostasis in vivo resulting in structural and functional disruption of neuronal networks. Neuron. 2008;59(2):214–25.PubMedPubMedCentralCrossRef Kuchibhotla KV, Goldman ST, Lattarulo CR, Wu HY, Hyman BT, Bacskai BJ. Abeta plaques lead to aberrant regulation of calcium homeostasis in vivo resulting in structural and functional disruption of neuronal networks. Neuron. 2008;59(2):214–25.PubMedPubMedCentralCrossRef
66.
go back to reference Demuro A, Mina E, Kayed R, Milton SC, Parker I, Glabe CG. Calcium dysregulation and membrane disruption as a ubiquitous neurotoxic mechanism of soluble amyloid oligomers. J Biol Chem. 2005;280(17):17294–300.PubMedCrossRef Demuro A, Mina E, Kayed R, Milton SC, Parker I, Glabe CG. Calcium dysregulation and membrane disruption as a ubiquitous neurotoxic mechanism of soluble amyloid oligomers. J Biol Chem. 2005;280(17):17294–300.PubMedCrossRef
67.
go back to reference Ranjan B, Chong KH, Zheng J. Composite mathematical modeling of calcium signaling behind neuronal cell death in Alzheimer's disease. BMC Syst Biol. 2018;12(Suppl 1):10.PubMedPubMedCentralCrossRef Ranjan B, Chong KH, Zheng J. Composite mathematical modeling of calcium signaling behind neuronal cell death in Alzheimer's disease. BMC Syst Biol. 2018;12(Suppl 1):10.PubMedPubMedCentralCrossRef
68.
go back to reference Yang L, Wang Z, Wang B, Justice NJ, Zheng H. Amyloid precursor protein regulates Cav1.2 L-type calcium channel levels and function to influence GABAergic short-term plasticity. J Neurosci. 2009;29(50):15660–8.PubMedPubMedCentralCrossRef Yang L, Wang Z, Wang B, Justice NJ, Zheng H. Amyloid precursor protein regulates Cav1.2 L-type calcium channel levels and function to influence GABAergic short-term plasticity. J Neurosci. 2009;29(50):15660–8.PubMedPubMedCentralCrossRef
69.
go back to reference Wang Y, Shi Y, Wei H. Calcium Dysregulation in Alzheimer's Disease: A Target for New Drug Development. J Alzheimers Dis Parkinsonism. 2017;7(5). Wang Y, Shi Y, Wei H. Calcium Dysregulation in Alzheimer's Disease: A Target for New Drug Development. J Alzheimers Dis Parkinsonism. 2017;7(5).
70.
go back to reference Samad N, Ishaq S, Bano S, Manzoor N. Calcium Regulation in Alzheimer's Disease: Mechanistic Understanding. J Coll Physicians Surg Pak. 2017;27(9):566–71.PubMed Samad N, Ishaq S, Bano S, Manzoor N. Calcium Regulation in Alzheimer's Disease: Mechanistic Understanding. J Coll Physicians Surg Pak. 2017;27(9):566–71.PubMed
71.
go back to reference Sciacca MFM, Monaco I, La Rosa C, Milardi D. The active role of Ca2+ ions in Aβ-mediated membrane damage. Chem Commun (Camb). 2018;54(29):3629–31.CrossRef Sciacca MFM, Monaco I, La Rosa C, Milardi D. The active role of Ca2+ ions in Aβ-mediated membrane damage. Chem Commun (Camb). 2018;54(29):3629–31.CrossRef
72.
go back to reference Lin L, Cao J, Yang SS, Fu ZQ, Zeng P, Chu J, et al. Endoplasmic reticulum stress induces spatial memory deficits by activating GSK-3. J Cell Mol Med. 2018;22(7):3489–502.PubMedPubMedCentralCrossRef Lin L, Cao J, Yang SS, Fu ZQ, Zeng P, Chu J, et al. Endoplasmic reticulum stress induces spatial memory deficits by activating GSK-3. J Cell Mol Med. 2018;22(7):3489–502.PubMedPubMedCentralCrossRef
73.
go back to reference Liu ZC, Chu J, Lin L, Song J, Ning LN, Luo HB, et al. SIL1 rescued bip elevation-related tau hyperphosphorylation in ER stress. Mol Neurobiol. 2016;53(2):983–94.PubMedCrossRef Liu ZC, Chu J, Lin L, Song J, Ning LN, Luo HB, et al. SIL1 rescued bip elevation-related tau hyperphosphorylation in ER stress. Mol Neurobiol. 2016;53(2):983–94.PubMedCrossRef
75.
go back to reference Samina S. Oxidative Stress and the Central Nervous System. J Pharmacol Exp Ther. 2017;360(1):201–5.CrossRef Samina S. Oxidative Stress and the Central Nervous System. J Pharmacol Exp Ther. 2017;360(1):201–5.CrossRef
77.
go back to reference García-Blanco A, Baquero M, Vento M, Gil E, Bataller L, Cháfer-Pericás C. Potential oxidative stress biomarkers of mild cognitive impairment due to Alzheimer disease. J Neurol Sci. 2017;373:295–302.PubMedCrossRef García-Blanco A, Baquero M, Vento M, Gil E, Bataller L, Cháfer-Pericás C. Potential oxidative stress biomarkers of mild cognitive impairment due to Alzheimer disease. J Neurol Sci. 2017;373:295–302.PubMedCrossRef
78.
go back to reference Breuer W, Ghoti H, Shattat A, Goldfarb A, Koren A, Levin C, et al. Non-transferrin bound iron in Thalassemia: differential detection of redox active forms in children and older patients. Am J Hematol. 2012;87(1):55–61.PubMedCrossRef Breuer W, Ghoti H, Shattat A, Goldfarb A, Koren A, Levin C, et al. Non-transferrin bound iron in Thalassemia: differential detection of redox active forms in children and older patients. Am J Hematol. 2012;87(1):55–61.PubMedCrossRef
79.
go back to reference Sayre LM, Perry G, Harris PL, Liu Y, Schubert KA, Smith MA. In situ oxidative catalysis by neurofibrillary tangles and senile plaques in Alzheimer' s disease:a central role forbound transition metals. J Neurochem. 2000;74(1):270–9.PubMedCrossRef Sayre LM, Perry G, Harris PL, Liu Y, Schubert KA, Smith MA. In situ oxidative catalysis by neurofibrillary tangles and senile plaques in Alzheimer' s disease:a central role forbound transition metals. J Neurochem. 2000;74(1):270–9.PubMedCrossRef
80.
go back to reference Smith MA, Zhu XW, Tabaton M, Liu G, McKeel DW Jr, Cohen ML, et al. Increased iron and free radical generation in preclinical Alzheimer disease and mild cognitive impairment. J Alzheimers Dis. 2010;19(1):363–72.PubMedPubMedCentralCrossRef Smith MA, Zhu XW, Tabaton M, Liu G, McKeel DW Jr, Cohen ML, et al. Increased iron and free radical generation in preclinical Alzheimer disease and mild cognitive impairment. J Alzheimers Dis. 2010;19(1):363–72.PubMedPubMedCentralCrossRef
81.
go back to reference Cheignon C, Tomas M, Bonnefont-Rousselot D, Faller P, Hureau C, Collin F. Oxidative stress and the amyloid beta peptide in Alzheimer's disease. Redox Biol. 2018;14:450–64.PubMedCrossRef Cheignon C, Tomas M, Bonnefont-Rousselot D, Faller P, Hureau C, Collin F. Oxidative stress and the amyloid beta peptide in Alzheimer's disease. Redox Biol. 2018;14:450–64.PubMedCrossRef
82.
go back to reference Rai RK, Chalana A, Karri R, Das R, Kumar B, et al. Role of hydrogen bonding by Thiones in protecting biomolecules from copper(I)-mediated oxidative damage. Inorg Chem. 2019;58(10):6628–38.PubMedCrossRef Rai RK, Chalana A, Karri R, Das R, Kumar B, et al. Role of hydrogen bonding by Thiones in protecting biomolecules from copper(I)-mediated oxidative damage. Inorg Chem. 2019;58(10):6628–38.PubMedCrossRef
83.
go back to reference Ahuja A, Dev K, Tanwar RS, Selwal KK, Tyagi PK. Copper mediated neurological disorder: visions into amyotrophic lateral sclerosis, Alzheimer and Menkes disease. J Trace Elem Med Biol. 2015;29:11–23.PubMedCrossRef Ahuja A, Dev K, Tanwar RS, Selwal KK, Tyagi PK. Copper mediated neurological disorder: visions into amyotrophic lateral sclerosis, Alzheimer and Menkes disease. J Trace Elem Med Biol. 2015;29:11–23.PubMedCrossRef
84.
go back to reference Myhre O, Utkilen H, Duale N, Brunborg G, Hofer T. Metal dyshomeostasis and inflammation in Alzheimer’s and Parkinson’s diseases: possible impact of environmental exposures. Oxidative Med Cell Longev. 2013;2013:726954.CrossRef Myhre O, Utkilen H, Duale N, Brunborg G, Hofer T. Metal dyshomeostasis and inflammation in Alzheimer’s and Parkinson’s diseases: possible impact of environmental exposures. Oxidative Med Cell Longev. 2013;2013:726954.CrossRef
85.
go back to reference Choo XY, Alukaidey L, White AR, Grubman A. Neuroinflammation and copper in Alzheimer’s disease. Int J Alzheimers Dis. 2013;2013:145345.PubMedPubMedCentral Choo XY, Alukaidey L, White AR, Grubman A. Neuroinflammation and copper in Alzheimer’s disease. Int J Alzheimers Dis. 2013;2013:145345.PubMedPubMedCentral
86.
go back to reference Vašák M, Meloni G. Mammalian metallothionein-3: New functional and structural insights. Int J Mol Sci. 2017;18(6). Vašák M, Meloni G. Mammalian metallothionein-3: New functional and structural insights. Int J Mol Sci. 2017;18(6).
87.
go back to reference Furuta T, Ohshima C, Matsumura M, Takebayashi N, Hirota E, Mawaribuchi T, et al. Oxidative stress upregulates zinc uptake activity via Zrt/Irt-like protein 1 (ZIP1) in cultured mouse astrocytes. Life Sci. 2016;151:305–12.PubMedCrossRef Furuta T, Ohshima C, Matsumura M, Takebayashi N, Hirota E, Mawaribuchi T, et al. Oxidative stress upregulates zinc uptake activity via Zrt/Irt-like protein 1 (ZIP1) in cultured mouse astrocytes. Life Sci. 2016;151:305–12.PubMedCrossRef
89.
go back to reference Maher P, van Leyen K, Dey PN, Honrath B, Dolga A, Methner A. The role of Ca2+ in cell death caused by oxidative glutamate toxicity and ferroptosis. Cell Calcium. 2018;70:47–55.PubMedCrossRef Maher P, van Leyen K, Dey PN, Honrath B, Dolga A, Methner A. The role of Ca2+ in cell death caused by oxidative glutamate toxicity and ferroptosis. Cell Calcium. 2018;70:47–55.PubMedCrossRef
91.
go back to reference Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozen A, et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 2016;12(1):1–222.PubMedPubMedCentralCrossRef Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozen A, et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 2016;12(1):1–222.PubMedPubMedCentralCrossRef
92.
go back to reference Boland B, Kumar A, Lee S, Platt FM, Wegiel J, Yu WH, et al. Autophagy induction and autophagosome clearance in neurons: relationship to autophagic pathology in Alzheimer’s disease. J Neurosci. 2008;28(27):6926–37.PubMedPubMedCentralCrossRef Boland B, Kumar A, Lee S, Platt FM, Wegiel J, Yu WH, et al. Autophagy induction and autophagosome clearance in neurons: relationship to autophagic pathology in Alzheimer’s disease. J Neurosci. 2008;28(27):6926–37.PubMedPubMedCentralCrossRef
93.
go back to reference Pi H, Li M, Tian L, Yang Z, Yu Z, Zhou Z. Enhancing lysosomal biogenesis and autophagic flux by activating the transcription factor EB protects against cadmium-induced neurotoxicity. Sci Rep. 2017;7:43466.PubMedPubMedCentralCrossRef Pi H, Li M, Tian L, Yang Z, Yu Z, Zhou Z. Enhancing lysosomal biogenesis and autophagic flux by activating the transcription factor EB protects against cadmium-induced neurotoxicity. Sci Rep. 2017;7:43466.PubMedPubMedCentralCrossRef
94.
go back to reference Atrián-Blasco E, Gonzalez P, Santoro A, Alies B, Faller P, Hureau C. Cu and Zn coordination to amyloid peptides: from fascinating chemistryto debated pathological relevance. Coord Chem Rev. 2018;375:38–55.PubMedPubMedCentralCrossRef Atrián-Blasco E, Gonzalez P, Santoro A, Alies B, Faller P, Hureau C. Cu and Zn coordination to amyloid peptides: from fascinating chemistryto debated pathological relevance. Coord Chem Rev. 2018;375:38–55.PubMedPubMedCentralCrossRef
95.
go back to reference De Biase D, Costagliola A, Pagano TB, Piegari G, Wojcik S, Dziewiątkowski J, et al. Amyloid precursor protein, lipofuscin accumulation and expression of autophagy markers in aged bovine brain. BMC Vet Re. 2017;13(1):102.CrossRef De Biase D, Costagliola A, Pagano TB, Piegari G, Wojcik S, Dziewiątkowski J, et al. Amyloid precursor protein, lipofuscin accumulation and expression of autophagy markers in aged bovine brain. BMC Vet Re. 2017;13(1):102.CrossRef
96.
go back to reference Belaidi AA, Bush AI. Iron neurochemistry in Alzheimer's disease and Parkinson's disease: targets for therapeutics. J Neurochem. 2016;139(Suppl 1):179–97.PubMedCrossRef Belaidi AA, Bush AI. Iron neurochemistry in Alzheimer's disease and Parkinson's disease: targets for therapeutics. J Neurochem. 2016;139(Suppl 1):179–97.PubMedCrossRef
97.
go back to reference Kwiatek-Majkusiak J, Dickson DW, Tacik P, Aoki N, Tomasiuk R, Koziorowski D, et al. Relationships between typical histopathological hallmarks and the ferritin in the hippocampus from patients with Alzheimer's disease. Acta Neurobiol Exp (Wars). 2015;75(4):391–8. Kwiatek-Majkusiak J, Dickson DW, Tacik P, Aoki N, Tomasiuk R, Koziorowski D, et al. Relationships between typical histopathological hallmarks and the ferritin in the hippocampus from patients with Alzheimer's disease. Acta Neurobiol Exp (Wars). 2015;75(4):391–8.
98.
go back to reference Figueiredo LS, de Freitas BS, Garcia VA, Dargél VA, Köbe LM, Kist LW, et al. Iron loading selectively increases hippocampal levels of Ubiquitinated proteins and impairs Hippocampus-dependent memory. Mol Neurobiol. 2016;53(9):6228–39.PubMedCrossRef Figueiredo LS, de Freitas BS, Garcia VA, Dargél VA, Köbe LM, Kist LW, et al. Iron loading selectively increases hippocampal levels of Ubiquitinated proteins and impairs Hippocampus-dependent memory. Mol Neurobiol. 2016;53(9):6228–39.PubMedCrossRef
99.
go back to reference Le W. Role of iron in UPS impairment model of Parkinson’s disease. Parkinsonism Relat Disord. 2014;20(Suppl 1):S158–61.PubMedCrossRef Le W. Role of iron in UPS impairment model of Parkinson’s disease. Parkinsonism Relat Disord. 2014;20(Suppl 1):S158–61.PubMedCrossRef
100.
go back to reference Bisceglie F, Alinovi R, Pinelli S, Galetti M, Pioli M, Tarasconi P, et al. Autophagy and apoptosis: studies on the effects of bisthiosemicarbazone copper (ii) complexes on p53 and p53-null tumour cell lines. Metallomics. 2016;8(12):1255–65.PubMedCrossRef Bisceglie F, Alinovi R, Pinelli S, Galetti M, Pioli M, Tarasconi P, et al. Autophagy and apoptosis: studies on the effects of bisthiosemicarbazone copper (ii) complexes on p53 and p53-null tumour cell lines. Metallomics. 2016;8(12):1255–65.PubMedCrossRef
101.
go back to reference Polishchuk EV, Polishchuk RS. The emerging role of lysosomes in copper homeostasis. Metallomics. 2016;8(9):853–62.PubMedCrossRef Polishchuk EV, Polishchuk RS. The emerging role of lysosomes in copper homeostasis. Metallomics. 2016;8(9):853–62.PubMedCrossRef
102.
go back to reference Sahni S, Bae DH, Jansson PJ, Richardson DR. The mechanistic role of chemically diverse metal ions in the induction of autophagy. Pharmacol Res. 2017;119:118–27.PubMedCrossRef Sahni S, Bae DH, Jansson PJ, Richardson DR. The mechanistic role of chemically diverse metal ions in the induction of autophagy. Pharmacol Res. 2017;119:118–27.PubMedCrossRef
103.
go back to reference Trejo-Solís C, Jimenez-Farfan D, Rodriguez-Enriquez S, Fernandez-Valverde F, Cruz-Salgado A, Ruiz-Azuara L, et al. Copper compound induces autophagy and apoptosis of glioma cells by reactive oxygen species and JNK activation. BMC Cancer. 2012;12:156.PubMedPubMedCentralCrossRef Trejo-Solís C, Jimenez-Farfan D, Rodriguez-Enriquez S, Fernandez-Valverde F, Cruz-Salgado A, Ruiz-Azuara L, et al. Copper compound induces autophagy and apoptosis of glioma cells by reactive oxygen species and JNK activation. BMC Cancer. 2012;12:156.PubMedPubMedCentralCrossRef
104.
go back to reference Amici M, Forti K, Nobili C, Lupidi G, Angeletti M, Fioretti E, et al. Effect of neurotoxic metal ions on the proteolytic activities of the 20S proteasome from bovine brain. J Biol InorgChem. 2002;7(7–8):750–6.CrossRef Amici M, Forti K, Nobili C, Lupidi G, Angeletti M, Fioretti E, et al. Effect of neurotoxic metal ions on the proteolytic activities of the 20S proteasome from bovine brain. J Biol InorgChem. 2002;7(7–8):750–6.CrossRef
105.
go back to reference Lanza V, Travaglia A, Malgieri G, Fattorusso R, Grasso G, Di Natale G, et al. Ubiquitin associates with the N-terminal domain of nerve growth factor: the role of copper (II) ions. Chemistry. 2016;22(49):17767–75.PubMedCrossRef Lanza V, Travaglia A, Malgieri G, Fattorusso R, Grasso G, Di Natale G, et al. Ubiquitin associates with the N-terminal domain of nerve growth factor: the role of copper (II) ions. Chemistry. 2016;22(49):17767–75.PubMedCrossRef
106.
go back to reference Zhao L, Liu Q, Ma S, Zhang Y, Liang P. TPEN attenuates neural autophagy induced by Synaptically-released zinc translocation and improves histological outcomes after traumatic brain injury in rats. Ann Clin Lab Sci. 2018;48(4):446–52.PubMed Zhao L, Liu Q, Ma S, Zhang Y, Liang P. TPEN attenuates neural autophagy induced by Synaptically-released zinc translocation and improves histological outcomes after traumatic brain injury in rats. Ann Clin Lab Sci. 2018;48(4):446–52.PubMed
107.
go back to reference Roy R, Singh SK, Chauhan LK, Das M, Tripathi A, Dwivedi PD. Zinc oxide nanoparticles induce apoptosis by enhancement of autophagy via PI3K/Akt/mTOR inhibition. Toxicol Lett. 2014;227(1):29–40.PubMedCrossRef Roy R, Singh SK, Chauhan LK, Das M, Tripathi A, Dwivedi PD. Zinc oxide nanoparticles induce apoptosis by enhancement of autophagy via PI3K/Akt/mTOR inhibition. Toxicol Lett. 2014;227(1):29–40.PubMedCrossRef
108.
go back to reference Song WJ, Jeong MS, Choi DM, Kim KN, Wie MB. Zinc Oxide Nanoparticles Induce Autophagy and Apoptosis via Oxidative Injury and Pro-Inflammatory Cytokines in Primary Astrocyte Cultures. Nanomaterials (Basel). 2019;9(7). Song WJ, Jeong MS, Choi DM, Kim KN, Wie MB. Zinc Oxide Nanoparticles Induce Autophagy and Apoptosis via Oxidative Injury and Pro-Inflammatory Cytokines in Primary Astrocyte Cultures. Nanomaterials (Basel). 2019;9(7).
109.
go back to reference Arena G, Fattorusso R, Grasso G, Grasso GI, Isernia C, Malgieri G, et al. Zinc (II) complexes of ubiquitin: speciation, Affinity and Binding Features. Chemistry. 2011;17(41):11596–603.PubMedCrossRef Arena G, Fattorusso R, Grasso G, Grasso GI, Isernia C, Malgieri G, et al. Zinc (II) complexes of ubiquitin: speciation, Affinity and Binding Features. Chemistry. 2011;17(41):11596–603.PubMedCrossRef
110.
go back to reference Kumar V, Singh D, Singh BK, Singh S, Mittra N, Jha RR, et al. Alpha-synuclein aggregation, ubiquitin proteasome system impairment, and L-Dopa response in zinc-induced parkinsonism: resemblance to sporadicParkinson's disease. Mol Cell Biochem. 2018;444(1–2):149–60.PubMedCrossRef Kumar V, Singh D, Singh BK, Singh S, Mittra N, Jha RR, et al. Alpha-synuclein aggregation, ubiquitin proteasome system impairment, and L-Dopa response in zinc-induced parkinsonism: resemblance to sporadicParkinson's disease. Mol Cell Biochem. 2018;444(1–2):149–60.PubMedCrossRef
111.
go back to reference Hung HH, Huang WP, Pan CY. Dopamine- and zinc-induced autophagosome formation facilitates PC12 cell survival. Cell Biol Toxicol. 2013;29(6):415–29.PubMedCrossRef Hung HH, Huang WP, Pan CY. Dopamine- and zinc-induced autophagosome formation facilitates PC12 cell survival. Cell Biol Toxicol. 2013;29(6):415–29.PubMedCrossRef
112.
go back to reference Portbury SD, Hare DJ, Sgambelloni C, Perronnes K, Portbury AJ, Finkelstein DI, et al. Trehalose improves cognition in the transgenic Tg2576 mouse model of Alzheimer's disease. J Alzheimers Dis. 2017;60(2):549–60.PubMedPubMedCentralCrossRef Portbury SD, Hare DJ, Sgambelloni C, Perronnes K, Portbury AJ, Finkelstein DI, et al. Trehalose improves cognition in the transgenic Tg2576 mouse model of Alzheimer's disease. J Alzheimers Dis. 2017;60(2):549–60.PubMedPubMedCentralCrossRef
113.
go back to reference Medina DL, Di Paola S, Peluso I, Armani A, De Stefani D, Venditti R, et al. Forrester, Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat Cell Biol. 2015;17(3):288–99.PubMedPubMedCentralCrossRef Medina DL, Di Paola S, Peluso I, Armani A, De Stefani D, Venditti R, et al. Forrester, Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat Cell Biol. 2015;17(3):288–99.PubMedPubMedCentralCrossRef
114.
go back to reference Zhang L, Fang Y, Cheng X, Lian Y, Xu H, Zeng Z, et al. TRPML1 participates in the progression of Alzheimer's disease by regulating the PPARγ/AMPK/Mtor Signalling pathway. Cell Physiol Biochem. 2017;43(6):2446–56.PubMedCrossRef Zhang L, Fang Y, Cheng X, Lian Y, Xu H, Zeng Z, et al. TRPML1 participates in the progression of Alzheimer's disease by regulating the PPARγ/AMPK/Mtor Signalling pathway. Cell Physiol Biochem. 2017;43(6):2446–56.PubMedCrossRef
115.
go back to reference Woods A, Dickerson K, Heath R, Hong SP, Momcilovic M, Johnstone SR, et al. Ca2+/calmodulin-dependent protein kinase kinase-beta acts upstream of AMP-activated protein kinase in mammalian cells. Cell Metab. 2005;2(1):21–33.PubMedCrossRef Woods A, Dickerson K, Heath R, Hong SP, Momcilovic M, Johnstone SR, et al. Ca2+/calmodulin-dependent protein kinase kinase-beta acts upstream of AMP-activated protein kinase in mammalian cells. Cell Metab. 2005;2(1):21–33.PubMedCrossRef
116.
go back to reference Jin Y, Bai Y, Ni H, Qiang L, Ye L, Shan Y, et al. Activation of autophagy through calcium-dependent AMPK/mTOR and PKCθ pathway causes activation of rat hepatic stellate cells under hypoxic stress. FEBS Lett. 2016;590(5):672–82.PubMedCrossRef Jin Y, Bai Y, Ni H, Qiang L, Ye L, Shan Y, et al. Activation of autophagy through calcium-dependent AMPK/mTOR and PKCθ pathway causes activation of rat hepatic stellate cells under hypoxic stress. FEBS Lett. 2016;590(5):672–82.PubMedCrossRef
117.
go back to reference Chang RY, Nouwens AS, Dodd PR, Etheridge N. The synaptic proteome in Alzheimer's disease. Alzheimers Dement. 2013;9(5):499–511.PubMedCrossRef Chang RY, Nouwens AS, Dodd PR, Etheridge N. The synaptic proteome in Alzheimer's disease. Alzheimers Dement. 2013;9(5):499–511.PubMedCrossRef
118.
go back to reference Rudisill SS, Martin BR, Mankowski KM, Tessier CR. Iron deficiency reduces synapse formation in the Drosophila clock circuit. Biol Trace Elem Res. 2019;189(1):241–50.PubMedCrossRef Rudisill SS, Martin BR, Mankowski KM, Tessier CR. Iron deficiency reduces synapse formation in the Drosophila clock circuit. Biol Trace Elem Res. 2019;189(1):241–50.PubMedCrossRef
119.
go back to reference Altamura S, Muckenthale MU. Iron toxicity in diseases of aging: Alzheimer’s disease, Parkinson’s disease and atherosclerosis. J Alzheimers Dis. 2009;16(4):879–95.PubMedCrossRef Altamura S, Muckenthale MU. Iron toxicity in diseases of aging: Alzheimer’s disease, Parkinson’s disease and atherosclerosis. J Alzheimers Dis. 2009;16(4):879–95.PubMedCrossRef
120.
go back to reference Wang X, Zhang J, Zhou L, Xu B, Ren X, He K, et al. Long-term iron exposure causes widespread molecular alterations associated with memory impairment in mice. Food Chem Toxicol. 2019;130:242–52.PubMedCrossRef Wang X, Zhang J, Zhou L, Xu B, Ren X, He K, et al. Long-term iron exposure causes widespread molecular alterations associated with memory impairment in mice. Food Chem Toxicol. 2019;130:242–52.PubMedCrossRef
121.
go back to reference Hung YH, Bush AI, Cherny RA. Copper in the brain and Alzheimer’s disease. J Biol Inorg Chem. 2010;15(1):61–76.PubMedCrossRef Hung YH, Bush AI, Cherny RA. Copper in the brain and Alzheimer’s disease. J Biol Inorg Chem. 2010;15(1):61–76.PubMedCrossRef
122.
go back to reference Kardos J, Kovacs I, Hajos F, Kalman M, Simonyi M. Nerve endings from rat brain tissue release copper upon depolarization. A possible role in regulating neuronal excitability. Neurosci Lett. 1989;103(2):139–44.PubMedCrossRef Kardos J, Kovacs I, Hajos F, Kalman M, Simonyi M. Nerve endings from rat brain tissue release copper upon depolarization. A possible role in regulating neuronal excitability. Neurosci Lett. 1989;103(2):139–44.PubMedCrossRef
123.
go back to reference Dodani SC, Firl A, Chan J, Nam CI, Aron AT, Onak CS, et al. Copper is an endogenous modulator of neural circuit spontaneous activity. Proc Natl Acad Sci U S A. 2014;111(46):16280–5.PubMedPubMedCentralCrossRef Dodani SC, Firl A, Chan J, Nam CI, Aron AT, Onak CS, et al. Copper is an endogenous modulator of neural circuit spontaneous activity. Proc Natl Acad Sci U S A. 2014;111(46):16280–5.PubMedPubMedCentralCrossRef
124.
go back to reference Yin HZ, Sensi SL, Ogoshi F, Weiss JH. Blockade of Ca2+−permeable AMPA/kainate channels decreases oxygen-glucose deprivation-induced Zn2+ accumulation and neuronal loss in hippocampal pyramidal neurons. J Neurosci. 2002;22(4):1273–9.PubMedPubMedCentralCrossRef Yin HZ, Sensi SL, Ogoshi F, Weiss JH. Blockade of Ca2+−permeable AMPA/kainate channels decreases oxygen-glucose deprivation-induced Zn2+ accumulation and neuronal loss in hippocampal pyramidal neurons. J Neurosci. 2002;22(4):1273–9.PubMedPubMedCentralCrossRef
125.
go back to reference Qiu M, Shentu YP, Zeng J, Wang XC, Yan X, Zhou XW, et al. Zinc mediates the neuronal activity- dependent anti-apoptotic effect. PLoS One. 2017;12(8):e0182150.PubMedPubMedCentralCrossRef Qiu M, Shentu YP, Zeng J, Wang XC, Yan X, Zhou XW, et al. Zinc mediates the neuronal activity- dependent anti-apoptotic effect. PLoS One. 2017;12(8):e0182150.PubMedPubMedCentralCrossRef
126.
go back to reference Leal G, Afonso PM, Salazar IL, Duarte CB. Regulation of hippocampal synaptic plasticity by brain-derived neurotrophic factor BDNF. Brain Res. 2015;1621:82–101.PubMedCrossRef Leal G, Afonso PM, Salazar IL, Duarte CB. Regulation of hippocampal synaptic plasticity by brain-derived neurotrophic factor BDNF. Brain Res. 2015;1621:82–101.PubMedCrossRef
127.
go back to reference Zagrebelsky M, Korte M. Form follows function: BDNF and its involvement in sculpting the function and structure of synapses. Neuropharmacology. 2014;76 Pt C:628–38.PubMedCrossRef Zagrebelsky M, Korte M. Form follows function: BDNF and its involvement in sculpting the function and structure of synapses. Neuropharmacology. 2014;76 Pt C:628–38.PubMedCrossRef
128.
go back to reference Hwang JJ, Park MH, Koh JY. Copper activates TrkB in cortical neurons in a metalloproteinase-dependent manner. J Neurosci Res. 2007;85(10):2160–6.PubMedCrossRef Hwang JJ, Park MH, Koh JY. Copper activates TrkB in cortical neurons in a metalloproteinase-dependent manner. J Neurosci Res. 2007;85(10):2160–6.PubMedCrossRef
129.
go back to reference Travaglia A, La Mendola D, Magrì A, Nicoletti VG, Pietropaolo A, et al. Copper, BDNF and Its N-terminal Domain: Inorganic Features and Biological Perspectives. Chemistry. 2012;18(49):15618–31.PubMedCrossRef Travaglia A, La Mendola D, Magrì A, Nicoletti VG, Pietropaolo A, et al. Copper, BDNF and Its N-terminal Domain: Inorganic Features and Biological Perspectives. Chemistry. 2012;18(49):15618–31.PubMedCrossRef
130.
go back to reference Frazzini V, Granzotto A, Bomba M, Massetti N, Castelli V, d'Aurora M, et al. The pharmacological perturbation of brain zinc impairs BDNF-related signaling and the cognitive performances of young mice. Sci Rep. 2018;8(1):976.CrossRef Frazzini V, Granzotto A, Bomba M, Massetti N, Castelli V, d'Aurora M, et al. The pharmacological perturbation of brain zinc impairs BDNF-related signaling and the cognitive performances of young mice. Sci Rep. 2018;8(1):976.CrossRef
131.
go back to reference Adlard PA, Parncutt JM, Finkelstein DI, Bush AI. Cognitive loss in ZincTransporter-3 Knock-out mice: a Phenocopy for the synaptic and memory, deficits of Alzheimer’s disease? J Neurosci. 2010;30(5):1631–6.PubMedPubMedCentralCrossRef Adlard PA, Parncutt JM, Finkelstein DI, Bush AI. Cognitive loss in ZincTransporter-3 Knock-out mice: a Phenocopy for the synaptic and memory, deficits of Alzheimer’s disease? J Neurosci. 2010;30(5):1631–6.PubMedPubMedCentralCrossRef
132.
go back to reference Cavallucci V, Berretta N, Nobili A, Nisticò R, Mercuri NB, D'Amelio M. Calcineurin inhibition rescues early synaptic plasticity deficits in a mouse model of Alzheimer’s disease. NeuroMolecular Med. 2013;15(3):541–8.PubMedCrossRef Cavallucci V, Berretta N, Nobili A, Nisticò R, Mercuri NB, D'Amelio M. Calcineurin inhibition rescues early synaptic plasticity deficits in a mouse model of Alzheimer’s disease. NeuroMolecular Med. 2013;15(3):541–8.PubMedCrossRef
133.
go back to reference Brown MR, Sullivan PG, Geddes JW. Synaptic mitochondria are more susceptible to Ca2+overload than nonsynaptic mitochondria. J Biol Chem. 2006;281(17):11658–68.PubMedCrossRef Brown MR, Sullivan PG, Geddes JW. Synaptic mitochondria are more susceptible to Ca2+overload than nonsynaptic mitochondria. J Biol Chem. 2006;281(17):11658–68.PubMedCrossRef
134.
go back to reference Goldstein JC, Waterhouse NJ, Juin P, Evan GI, Green DR. The coordinate release of cytochrome c during apoptosis is rapid, complete and kinetically invariant. Nat Cell Biol. 2000;2(3):156–62.PubMedCrossRef Goldstein JC, Waterhouse NJ, Juin P, Evan GI, Green DR. The coordinate release of cytochrome c during apoptosis is rapid, complete and kinetically invariant. Nat Cell Biol. 2000;2(3):156–62.PubMedCrossRef
135.
go back to reference Zervas M, Opitz T, Edelmann W, Wanier B, Kucherlapati R, Stanton PK. Impaired hippocampal long-term potentiation in microtubule-associated protein 1B-deficient mice. J Neurosci Res. 2005;82(1):83–92.PubMedCrossRef Zervas M, Opitz T, Edelmann W, Wanier B, Kucherlapati R, Stanton PK. Impaired hippocampal long-term potentiation in microtubule-associated protein 1B-deficient mice. J Neurosci Res. 2005;82(1):83–92.PubMedCrossRef
136.
go back to reference Fukunaga K, Muller D, Miyamoto E. CaM kinase II in long-term potentiation. Neurochem Int. 1996;28(4):343–58.PubMedCrossRef Fukunaga K, Muller D, Miyamoto E. CaM kinase II in long-term potentiation. Neurochem Int. 1996;28(4):343–58.PubMedCrossRef
137.
go back to reference Liu Y, Nguyen M, Robert A, Meunier B. Metal ions in Alzheimer’s disease: a key role or not? Acc ChemRes. 2019;52(7):2026–35.CrossRef Liu Y, Nguyen M, Robert A, Meunier B. Metal ions in Alzheimer’s disease: a key role or not? Acc ChemRes. 2019;52(7):2026–35.CrossRef
Metadata
Title
Current understanding of metal ions in the pathogenesis of Alzheimer’s disease
Authors
Lu Wang
Ya-Ling Yin
Xin-Zi Liu
Peng Shen
Yan-Ge Zheng
Xin-Rui Lan
Cheng-Biao Lu
Jian-Zhi Wang
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Translational Neurodegeneration / Issue 1/2020
Electronic ISSN: 2047-9158
DOI
https://doi.org/10.1186/s40035-020-00189-z

Other articles of this Issue 1/2020

Translational Neurodegeneration 1/2020 Go to the issue