Skip to main content
Top
Published in: Acta Neuropathologica Communications 1/2013

Open Access 01-12-2013 | Research

A central role for dityrosine crosslinking of Amyloid-β in Alzheimer’s disease

Authors: Youssra K Al-Hilaly, Thomas L Williams, Maris Stewart-Parker, Lenzie Ford, Eldhose Skaria, Michael Cole, William Grant Bucher, Kyle L Morris, Alaa Abdul Sada, Julian R Thorpe, Louise C Serpell

Published in: Acta Neuropathologica Communications | Issue 1/2013

Login to get access

Abstract

Background

Alzheimer’s disease (AD) is characterized by the deposition of insoluble amyloid plaques in the neuropil composed of highly stable, self-assembled Amyloid-beta (Aβ) fibrils. Copper has been implicated to play a role in Alzheimer’s disease. Dimers of Aβ have been isolated from AD brain and have been shown to be neurotoxic.

Results

We have investigated the formation of dityrosine cross-links in Aβ42 formed by covalent ortho-ortho coupling of two tyrosine residues under conditions of oxidative stress with elevated copper and shown that dityrosine can be formed in vitro in Aβ oligomers and fibrils and that these links further stabilize the fibrils. Dityrosine crosslinking was present in internalized Aβ in cell cultures treated with oligomeric Aβ42 using a specific antibody for dityrosine by immunogold labeling transmission electron microscopy. Results also revealed the prevalence of dityrosine crosslinks in amyloid plaques in brain tissue and in cerebrospinal fluid from AD patients.

Conclusions

Aβ dimers may be stabilized by dityrosine crosslinking. These results indicate that dityrosine cross-links may play an important role in the pathogenesis of Alzheimer’s disease and can be generated by reactive oxygen species catalyzed by Cu2+ ions. The observation of increased Aβ and dityrosine in CSF from AD patients suggests that this could be used as a potential biomarker of oxidative stress in AD.
Appendix
Available only for authorised users
Literature
1.
go back to reference Selkoe DJ: The molecular pathology of Alzheimer’s disease. Neuron 1991, 6: 487–498. 10.1016/0896-6273(91)90052-2CrossRefPubMed Selkoe DJ: The molecular pathology of Alzheimer’s disease. Neuron 1991, 6: 487–498. 10.1016/0896-6273(91)90052-2CrossRefPubMed
3.
go back to reference Hardy J, Selkoe DJ: The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 2002, 297: 353–356. 10.1126/science.1072994CrossRefPubMed Hardy J, Selkoe DJ: The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science 2002, 297: 353–356. 10.1126/science.1072994CrossRefPubMed
4.
go back to reference Luhrs T, Ritter C, Adrian M, Riek-Loher D, Bohrmann B, Dobeli H, Schubert D, Riek R: 3D structure of Alzheimer’s amyloid-beta(1–42) fibrils. Proc Natl Acad Sci USA 2005, 102: 17342–17347. 10.1073/pnas.0506723102PubMedCentralCrossRefPubMed Luhrs T, Ritter C, Adrian M, Riek-Loher D, Bohrmann B, Dobeli H, Schubert D, Riek R: 3D structure of Alzheimer’s amyloid-beta(1–42) fibrils. Proc Natl Acad Sci USA 2005, 102: 17342–17347. 10.1073/pnas.0506723102PubMedCentralCrossRefPubMed
5.
go back to reference Hensley K, Maidt ML, Yu Z, Sang H, Markesbery WR, Floyd RA: Electrochemical analysis of protein nitrotyrosine and dityrosine in the Alzheimer brain indicates region-specific accumulation. J Neurosci 1998, 18: 8126–8132.PubMed Hensley K, Maidt ML, Yu Z, Sang H, Markesbery WR, Floyd RA: Electrochemical analysis of protein nitrotyrosine and dityrosine in the Alzheimer brain indicates region-specific accumulation. J Neurosci 1998, 18: 8126–8132.PubMed
6.
go back to reference Bush AI, Curtain CC: Twenty years of metallo-neurobiology: where to now? Eur Biophys J 2008, 37: 241–245. 10.1007/s00249-007-0228-1CrossRefPubMed Bush AI, Curtain CC: Twenty years of metallo-neurobiology: where to now? Eur Biophys J 2008, 37: 241–245. 10.1007/s00249-007-0228-1CrossRefPubMed
7.
go back to reference Ali FE, Barnham KJ, Barrow CJ, Separovic F: Copper catalysed oxidation of amino acids and Alzheimer’s disease. Lett Pept Sci 2003, 10: 405–412. 10.1007/BF02442571CrossRef Ali FE, Barnham KJ, Barrow CJ, Separovic F: Copper catalysed oxidation of amino acids and Alzheimer’s disease. Lett Pept Sci 2003, 10: 405–412. 10.1007/BF02442571CrossRef
8.
go back to reference Sarell CJ, Wilkinson SR, Viles JH: Substoichiometric levels of Cu2+ ions accelerate the kinetics of fiber formation and promote cell toxicity of amyloid-Œ ≤ from Alzheimer disease. J Biol Chem 2010, 285: 41533–41540. 10.1074/jbc.M110.171355PubMedCentralCrossRefPubMed Sarell CJ, Wilkinson SR, Viles JH: Substoichiometric levels of Cu2+ ions accelerate the kinetics of fiber formation and promote cell toxicity of amyloid-Œ ≤ from Alzheimer disease. J Biol Chem 2010, 285: 41533–41540. 10.1074/jbc.M110.171355PubMedCentralCrossRefPubMed
9.
go back to reference Huang X, Moir RD, Tanzi RE, Bush AI, Rogers JT: Redox-active metals, oxidative stress, and Alzheimer’s disease pathology. Ann Ny Acad Sci 2004, 1012: 153–163. 10.1196/annals.1306.012CrossRefPubMed Huang X, Moir RD, Tanzi RE, Bush AI, Rogers JT: Redox-active metals, oxidative stress, and Alzheimer’s disease pathology. Ann Ny Acad Sci 2004, 1012: 153–163. 10.1196/annals.1306.012CrossRefPubMed
10.
go back to reference Bush AI: The metallobiology of Alzheimer's disease. Trends Neurosci 2003, 26: 207–214. 10.1016/S0166-2236(03)00067-5CrossRefPubMed Bush AI: The metallobiology of Alzheimer's disease. Trends Neurosci 2003, 26: 207–214. 10.1016/S0166-2236(03)00067-5CrossRefPubMed
11.
go back to reference Levine RL, Stadtman ER: Oxidative modification of proteins during aging. Exp Gerontol 2001, 36: 1495–1502. 10.1016/S0531-5565(01)00135-8CrossRefPubMed Levine RL, Stadtman ER: Oxidative modification of proteins during aging. Exp Gerontol 2001, 36: 1495–1502. 10.1016/S0531-5565(01)00135-8CrossRefPubMed
12.
go back to reference Lynch T, Cherny RA, Bush AI: Oxidative processes in Alzheimer’s disease: the role of Aβ-metal interactions. Exp Gerontol 2000, 35: 445–451. 10.1016/S0531-5565(00)00112-1CrossRefPubMed Lynch T, Cherny RA, Bush AI: Oxidative processes in Alzheimer’s disease: the role of Aβ-metal interactions. Exp Gerontol 2000, 35: 445–451. 10.1016/S0531-5565(00)00112-1CrossRefPubMed
13.
go back to reference Butterfield DA, Reed T, Newman SF, Sultana R: Roles of amyloid beta-peptide-associated oxidative stress and brain protein modifications in the pathogenesis of Alzheimer’s disease and mild cognitive impairment. Free Radic Biol Med 2007, 43: 658–677. 10.1016/j.freeradbiomed.2007.05.037PubMedCentralCrossRefPubMed Butterfield DA, Reed T, Newman SF, Sultana R: Roles of amyloid beta-peptide-associated oxidative stress and brain protein modifications in the pathogenesis of Alzheimer’s disease and mild cognitive impairment. Free Radic Biol Med 2007, 43: 658–677. 10.1016/j.freeradbiomed.2007.05.037PubMedCentralCrossRefPubMed
14.
go back to reference Double KL, Dedov VN, Fedorow H, Kettle E, Halliday GM, Garner B, Brunk UT: The comparative biology of neuromelanin and lipofuscin in the human brain. Cell Mol Life Sci 2008, 65: 1669–1682. 10.1007/s00018-008-7581-9CrossRefPubMed Double KL, Dedov VN, Fedorow H, Kettle E, Halliday GM, Garner B, Brunk UT: The comparative biology of neuromelanin and lipofuscin in the human brain. Cell Mol Life Sci 2008, 65: 1669–1682. 10.1007/s00018-008-7581-9CrossRefPubMed
15.
go back to reference Lovell MA, Robertson JD, Teesdale WJ, Campbell JL, Markesbery WR: Copper, iron and zinc in Alzheimer’s disease senile plaques. J Neurol Sci 1998, 158: 47–52. 10.1016/S0022-510X(98)00092-6CrossRefPubMed Lovell MA, Robertson JD, Teesdale WJ, Campbell JL, Markesbery WR: Copper, iron and zinc in Alzheimer’s disease senile plaques. J Neurol Sci 1998, 158: 47–52. 10.1016/S0022-510X(98)00092-6CrossRefPubMed
16.
go back to reference Huang X, Atwood CS, Hartshorn MA, Multhaup G, Goldstein LE, Scarpa RC, Cuajungco MP, Gray DN, Lim J, Moir RD, et al.: The A beta peptide of Alzheimer’s disease directly produces hydrogen peroxide through metal ion reduction. Biochemistry-Us 1999, 38: 7609–7616. 10.1021/bi990438fCrossRef Huang X, Atwood CS, Hartshorn MA, Multhaup G, Goldstein LE, Scarpa RC, Cuajungco MP, Gray DN, Lim J, Moir RD, et al.: The A beta peptide of Alzheimer’s disease directly produces hydrogen peroxide through metal ion reduction. Biochemistry-Us 1999, 38: 7609–7616. 10.1021/bi990438fCrossRef
17.
go back to reference Atwood CS, Moir RD, Huang X, Scarpa RC, Bacarra NME, Romano DM, Hartshorn MA, Tanzi RE, Bush AI: Dramatic aggregation of Alzheimer AŒ ≤ by Cu(II) is induced by conditions representing physiological acidosis. J Biol Chem 1998, 273: 12817–12826. 10.1074/jbc.273.21.12817CrossRefPubMed Atwood CS, Moir RD, Huang X, Scarpa RC, Bacarra NME, Romano DM, Hartshorn MA, Tanzi RE, Bush AI: Dramatic aggregation of Alzheimer AŒ ≤ by Cu(II) is induced by conditions representing physiological acidosis. J Biol Chem 1998, 273: 12817–12826. 10.1074/jbc.273.21.12817CrossRefPubMed
18.
go back to reference Souza JM, Giasson BI, Chen Q, Lee VM, Ischiropoulos H: Dityrosine cross-linking promotes formation of stable alpha -synuclein polymers. Implication of nitrative and oxidative stress in the pathogenesis of neurodegenerative synucleinopathies. J Biol Chem 2000, 275: 18344–18349. 10.1074/jbc.M000206200CrossRefPubMed Souza JM, Giasson BI, Chen Q, Lee VM, Ischiropoulos H: Dityrosine cross-linking promotes formation of stable alpha -synuclein polymers. Implication of nitrative and oxidative stress in the pathogenesis of neurodegenerative synucleinopathies. J Biol Chem 2000, 275: 18344–18349. 10.1074/jbc.M000206200CrossRefPubMed
19.
go back to reference Yoburn JC, Tian W, Brower JO, Nowick JS, Glabe CG, Van Vranken DL: Dityrosine cross-linked Abeta peptides: fibrillar beta-structure in Abeta(1–40) is conducive to formation of dityrosine cross-links but a dityrosine cross-link in Abeta(8–14) does not induce beta-structure. Chem Res Toxicol 2003, 16: 531–535. 10.1021/tx025666gCrossRefPubMed Yoburn JC, Tian W, Brower JO, Nowick JS, Glabe CG, Van Vranken DL: Dityrosine cross-linked Abeta peptides: fibrillar beta-structure in Abeta(1–40) is conducive to formation of dityrosine cross-links but a dityrosine cross-link in Abeta(8–14) does not induce beta-structure. Chem Res Toxicol 2003, 16: 531–535. 10.1021/tx025666gCrossRefPubMed
20.
go back to reference Atwood CS, Perry G, Zeng H, Kato Y, Jones WD, Ling KQ, Huang X, Moir RD, Wang D, Sayre LM, et al.: Copper mediates dityrosine cross-linking of Alzheimer’s amyloid-beta. Biochemistry-Us 2004, 43: 560–568. 10.1021/bi0358824CrossRef Atwood CS, Perry G, Zeng H, Kato Y, Jones WD, Ling KQ, Huang X, Moir RD, Wang D, Sayre LM, et al.: Copper mediates dityrosine cross-linking of Alzheimer’s amyloid-beta. Biochemistry-Us 2004, 43: 560–568. 10.1021/bi0358824CrossRef
21.
go back to reference Giulivi C, Traaseth NJ, Davies KJ: Tyrosine oxidation products: analysis and biological relevance. Amino Acids 2003, 25: 227–232. 10.1007/s00726-003-0013-0CrossRefPubMed Giulivi C, Traaseth NJ, Davies KJ: Tyrosine oxidation products: analysis and biological relevance. Amino Acids 2003, 25: 227–232. 10.1007/s00726-003-0013-0CrossRefPubMed
22.
go back to reference Smith DP, Smith DG, Curtain CC, Boas JF, Pilbrow JR, Ciccotosto GD, Lau TL, Tew DJ, Perez K, Wade JD, et al.: Copper-mediated amyloid-beta toxicity is associated with an intermolecular histidine bridge. J Biol Chem 2006, 281: 15145–15154. 10.1074/jbc.M600417200CrossRefPubMed Smith DP, Smith DG, Curtain CC, Boas JF, Pilbrow JR, Ciccotosto GD, Lau TL, Tew DJ, Perez K, Wade JD, et al.: Copper-mediated amyloid-beta toxicity is associated with an intermolecular histidine bridge. J Biol Chem 2006, 281: 15145–15154. 10.1074/jbc.M600417200CrossRefPubMed
23.
go back to reference DiMarco T, Giulivi C: Current analytical methods for the detection of dityrosine, a biomarker of oxidative stress, in biological samples. Mass Spectrom Rev 2007, 26: 108–120. 10.1002/mas.20109CrossRefPubMed DiMarco T, Giulivi C: Current analytical methods for the detection of dityrosine, a biomarker of oxidative stress, in biological samples. Mass Spectrom Rev 2007, 26: 108–120. 10.1002/mas.20109CrossRefPubMed
24.
go back to reference Amado R, Aeschbach R, Neukom H: Dityrosine - invitro production and characterization. Methods Enzymol 1984, 107: 377–388.CrossRefPubMed Amado R, Aeschbach R, Neukom H: Dityrosine - invitro production and characterization. Methods Enzymol 1984, 107: 377–388.CrossRefPubMed
25.
go back to reference Giulivi C, Davies KJ: Dityrosine: a marker for oxidatively modified proteins and selective proteolysis. Methods Enzymol 1994, 233: 363–371.CrossRefPubMed Giulivi C, Davies KJ: Dityrosine: a marker for oxidatively modified proteins and selective proteolysis. Methods Enzymol 1994, 233: 363–371.CrossRefPubMed
26.
go back to reference Guo ZW, Salamonczyk GM, Han K, Machiya K, Sih CJ: Enzymatic oxidative phenolic coupling. J Org Chem 1997, 62: 6700–6701. 10.1021/jo970995cCrossRef Guo ZW, Salamonczyk GM, Han K, Machiya K, Sih CJ: Enzymatic oxidative phenolic coupling. J Org Chem 1997, 62: 6700–6701. 10.1021/jo970995cCrossRef
27.
go back to reference Lee DI, Hwang S, Choi JY, Ahn IS, Lee CH: A convenient preparation of dityrosine via Mn(III)-mediated oxidation of tyrosine. Process Biochem 2008, 43: 999–1003. 10.1016/j.procbio.2008.04.020CrossRef Lee DI, Hwang S, Choi JY, Ahn IS, Lee CH: A convenient preparation of dityrosine via Mn(III)-mediated oxidation of tyrosine. Process Biochem 2008, 43: 999–1003. 10.1016/j.procbio.2008.04.020CrossRef
28.
go back to reference Skaff O, Jolliffe KA, Hutton CA: Synthesis of the side chain cross-linked tyrosine oligomers dityrosine, trityrosine, and pulcherosine. J Org Chem 2005, 70: 7353–7363. 10.1021/jo051076mCrossRefPubMed Skaff O, Jolliffe KA, Hutton CA: Synthesis of the side chain cross-linked tyrosine oligomers dityrosine, trityrosine, and pulcherosine. J Org Chem 2005, 70: 7353–7363. 10.1021/jo051076mCrossRefPubMed
29.
go back to reference Jacob JS, Cistola DP, Hsu FF, Muzaffar S, Mueller DM, Hazen SL, Heinecke JW: Human phagocytes employ the myeloperoxidase-hydrogen peroxide system to synthesize dityrosine, trityrosine, pulcherosine, and isodityrosine by a tyrosyl radical-dependent pathway. J Biol Chem 1996, 271: 19950–19956. 10.1074/jbc.271.33.19950CrossRefPubMed Jacob JS, Cistola DP, Hsu FF, Muzaffar S, Mueller DM, Hazen SL, Heinecke JW: Human phagocytes employ the myeloperoxidase-hydrogen peroxide system to synthesize dityrosine, trityrosine, pulcherosine, and isodityrosine by a tyrosyl radical-dependent pathway. J Biol Chem 1996, 271: 19950–19956. 10.1074/jbc.271.33.19950CrossRefPubMed
30.
go back to reference Williams TL, Day IJ, Serpell LC: The effect of Alzheimer’s Abeta aggregation state on the permeation of biomimetic lipid vesicles. Langmuir 2010, 26: 17260–17268. 10.1021/la101581gCrossRefPubMed Williams TL, Day IJ, Serpell LC: The effect of Alzheimer’s Abeta aggregation state on the permeation of biomimetic lipid vesicles. Langmuir 2010, 26: 17260–17268. 10.1021/la101581gCrossRefPubMed
31.
go back to reference Kato Y, Wu X, Naito M, Nomura H, Kitamoto N, Osawa T: Immunochemical detection of protein dityrosine in atherosclerotic lesion of apo-E-deficient mice using a novel monoclonal antibody. Biochem Biophys Res Commun 2000, 275: 11–15. 10.1006/bbrc.2000.3265CrossRefPubMed Kato Y, Wu X, Naito M, Nomura H, Kitamoto N, Osawa T: Immunochemical detection of protein dityrosine in atherosclerotic lesion of apo-E-deficient mice using a novel monoclonal antibody. Biochem Biophys Res Commun 2000, 275: 11–15. 10.1006/bbrc.2000.3265CrossRefPubMed
32.
go back to reference Agholme L, Hallbeck M, Benedikz E, Marcusson J, Kagedal K: Amyloid-beta secretion, generation, and lysosomal sequestration in response to proteasome inhibition: involvement of autophagy. J Alzheimers Dis 2012, 31: 343–358.PubMed Agholme L, Hallbeck M, Benedikz E, Marcusson J, Kagedal K: Amyloid-beta secretion, generation, and lysosomal sequestration in response to proteasome inhibition: involvement of autophagy. J Alzheimers Dis 2012, 31: 343–358.PubMed
33.
go back to reference Soura V, Stewart-Parker M, Williams TL, Ratnayaka A, Atherton J, Gorringe K, Tuffin J, Darwent E, Rambaran R, Klein W, et al.: Visualization of co-localization in Abeta42-administered neuroblastoma cells reveals lysosome damage and autophagosome accumulation related to cell death. Biochem J 2012, 441: 579–590. 10.1042/BJ20110749CrossRefPubMed Soura V, Stewart-Parker M, Williams TL, Ratnayaka A, Atherton J, Gorringe K, Tuffin J, Darwent E, Rambaran R, Klein W, et al.: Visualization of co-localization in Abeta42-administered neuroblastoma cells reveals lysosome damage and autophagosome accumulation related to cell death. Biochem J 2012, 441: 579–590. 10.1042/BJ20110749CrossRefPubMed
34.
go back to reference Thorpe JR, Morley SJ, Rulten SL: Utilizing the peptidyl-prolyl cis-trans isomerase Pin1 as a probe of its phosphorylated target proteins: Examples of binding to nuclear proteins in a human kidney cell line and to tau in Alzheimer’s diseased brain. J Histochem Cytochem 2001, 49: 97–107. 10.1177/002215540104900110CrossRefPubMed Thorpe JR, Morley SJ, Rulten SL: Utilizing the peptidyl-prolyl cis-trans isomerase Pin1 as a probe of its phosphorylated target proteins: Examples of binding to nuclear proteins in a human kidney cell line and to tau in Alzheimer’s diseased brain. J Histochem Cytochem 2001, 49: 97–107. 10.1177/002215540104900110CrossRefPubMed
35.
go back to reference Thorpe JR: The application of LR gold resin for immunogold labeling. Methods Mol Biol 1999, 117: 99–110.PubMed Thorpe JR: The application of LR gold resin for immunogold labeling. Methods Mol Biol 1999, 117: 99–110.PubMed
36.
go back to reference Malencik DA, Sprouse JF, Swanson CA, Anderson SR: Dityrosine: Preparation, isolation, and analysis. Anal Biochem 1996, 242: 202–213. 10.1006/abio.1996.0454CrossRefPubMed Malencik DA, Sprouse JF, Swanson CA, Anderson SR: Dityrosine: Preparation, isolation, and analysis. Anal Biochem 1996, 242: 202–213. 10.1006/abio.1996.0454CrossRefPubMed
37.
go back to reference Paravastu AK, Leapman RD, Yau WM, Tycko R: Molecular structural basis for polymorphism in Alzheimer’s beta-amyloid fibrils. Proc Natl Acad Sci USA 2008, 105: 18349–18354. 10.1073/pnas.0806270105PubMedCentralCrossRefPubMed Paravastu AK, Leapman RD, Yau WM, Tycko R: Molecular structural basis for polymorphism in Alzheimer’s beta-amyloid fibrils. Proc Natl Acad Sci USA 2008, 105: 18349–18354. 10.1073/pnas.0806270105PubMedCentralCrossRefPubMed
38.
39.
go back to reference Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ: Naturally secreted oligomers of amyloid Œ ≤ protein potently inhibit hippocampal long-term potentiation in vivo. Nature 2002, 416: 535–539. 10.1038/416535aCrossRefPubMed Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ: Naturally secreted oligomers of amyloid Œ ≤ protein potently inhibit hippocampal long-term potentiation in vivo. Nature 2002, 416: 535–539. 10.1038/416535aCrossRefPubMed
40.
go back to reference Kuo YM, Emmerling MR, Vigo-Pelfrey C, Kasunic TC, Kirkpatrick JB, Murdoch GH, Ball MJ, Roher AE: Water-soluble Abeta (N-40, N-42) oligomers in normal and Alzheimer disease brains. J Biol Chem 1996, 271: 4077–4081. 10.1074/jbc.271.8.4077CrossRefPubMed Kuo YM, Emmerling MR, Vigo-Pelfrey C, Kasunic TC, Kirkpatrick JB, Murdoch GH, Ball MJ, Roher AE: Water-soluble Abeta (N-40, N-42) oligomers in normal and Alzheimer disease brains. J Biol Chem 1996, 271: 4077–4081. 10.1074/jbc.271.8.4077CrossRefPubMed
41.
go back to reference Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, Brett FM, Farrell MA, Rowan MJ, Lemere CA, et al.: Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nature medicine 2008, 14: 837–842. 10.1038/nm1782PubMedCentralCrossRefPubMed Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, Brett FM, Farrell MA, Rowan MJ, Lemere CA, et al.: Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nature medicine 2008, 14: 837–842. 10.1038/nm1782PubMedCentralCrossRefPubMed
42.
go back to reference Kok WM, Cottam JM, Ciccotosto GD, Miles LA, Karas JA, Scanlon DB, Roberts BR, Parker MW, Cappai R, Barnham KJ, Hutton CA: Synthetic dityrosine-linked beta-amyloid dimers form stable, soluble neurotoxic oligomers. Chem Sci 2013, 4: 4449–4454. 10.1039/c3sc22295kCrossRef Kok WM, Cottam JM, Ciccotosto GD, Miles LA, Karas JA, Scanlon DB, Roberts BR, Parker MW, Cappai R, Barnham KJ, Hutton CA: Synthetic dityrosine-linked beta-amyloid dimers form stable, soluble neurotoxic oligomers. Chem Sci 2013, 4: 4449–4454. 10.1039/c3sc22295kCrossRef
43.
go back to reference Marshall KE, Serpell LC: Structural integrity of beta-sheet assembly. Biochem Soc Trans 2009, 37: 671–676. 10.1042/BST0370671CrossRefPubMed Marshall KE, Serpell LC: Structural integrity of beta-sheet assembly. Biochem Soc Trans 2009, 37: 671–676. 10.1042/BST0370671CrossRefPubMed
44.
go back to reference Bitan G, Kirkitadze MD, Lomakin A, Vollers SS, Benedek GB, Teplow DB: Amyloid beta -protein (Abeta) assembly: Abeta 40 and Abeta 42 oligomerize through distinct pathways. Proc Natl Acad Sci USA 2003, 100: 330–335. 10.1073/pnas.222681699PubMedCentralCrossRefPubMed Bitan G, Kirkitadze MD, Lomakin A, Vollers SS, Benedek GB, Teplow DB: Amyloid beta -protein (Abeta) assembly: Abeta 40 and Abeta 42 oligomerize through distinct pathways. Proc Natl Acad Sci USA 2003, 100: 330–335. 10.1073/pnas.222681699PubMedCentralCrossRefPubMed
45.
go back to reference Smith DP, Ciccotosto GD, Tew DJ, Fodero-Tavoletti MT, Johanssen T, Masters CL, Barnham KJ, Cappai R: Concentration dependent Cu2+ induced aggregation and dityrosine formation of the Alzheimer’s disease amyloid-beta peptide. Biochemistry-Us 2007, 46: 2881–2891. 10.1021/bi0620961CrossRef Smith DP, Ciccotosto GD, Tew DJ, Fodero-Tavoletti MT, Johanssen T, Masters CL, Barnham KJ, Cappai R: Concentration dependent Cu2+ induced aggregation and dityrosine formation of the Alzheimer’s disease amyloid-beta peptide. Biochemistry-Us 2007, 46: 2881–2891. 10.1021/bi0620961CrossRef
46.
go back to reference Huang X, Cuajungco MP, Atwood CS, Hartshorn MA, Tyndall JDA, Hanson GR, Stokes KC, Leopold M, Multhaup G, Goldstein LE, et al.: Cu(II) Potentiation of Alzheimer Aβ Neurotoxicity: CORRELATION WITH CELL-FREE HYDROGEN PEROXIDE PRODUCTION AND METAL REDUCTION. J Biol Chem 1999, 274: 37111–37116. 10.1074/jbc.274.52.37111CrossRefPubMed Huang X, Cuajungco MP, Atwood CS, Hartshorn MA, Tyndall JDA, Hanson GR, Stokes KC, Leopold M, Multhaup G, Goldstein LE, et al.: Cu(II) Potentiation of Alzheimer Aβ Neurotoxicity: CORRELATION WITH CELL-FREE HYDROGEN PEROXIDE PRODUCTION AND METAL REDUCTION. J Biol Chem 1999, 274: 37111–37116. 10.1074/jbc.274.52.37111CrossRefPubMed
47.
go back to reference Syme CD, Nadal RC, Rigby SE, Viles JH: Copper binding to the amyloid-beta (Abeta) peptide associated with Alzheimer’s disease: folding, coordination geometry, pH dependence, stoichiometry, and affinity of Abeta-(1–28): insights from a range of complementary spectroscopic techniques. J Biol Chem 2004, 279: 18169–18177. 10.1074/jbc.M313572200CrossRefPubMed Syme CD, Nadal RC, Rigby SE, Viles JH: Copper binding to the amyloid-beta (Abeta) peptide associated with Alzheimer’s disease: folding, coordination geometry, pH dependence, stoichiometry, and affinity of Abeta-(1–28): insights from a range of complementary spectroscopic techniques. J Biol Chem 2004, 279: 18169–18177. 10.1074/jbc.M313572200CrossRefPubMed
48.
go back to reference Atwood CS, Scarpa RC, Huang XD, Moir RD, Jones WD, Fairlie DP, Tanzi RE, Bush AI: Characterization of copper interactions with Alzheimer amyloid beta peptides: identification of an attomolar-affinity copper binding site on amyloid beta 1–42. J Neurochem 2000, 75: 1219–1233.CrossRefPubMed Atwood CS, Scarpa RC, Huang XD, Moir RD, Jones WD, Fairlie DP, Tanzi RE, Bush AI: Characterization of copper interactions with Alzheimer amyloid beta peptides: identification of an attomolar-affinity copper binding site on amyloid beta 1–42. J Neurochem 2000, 75: 1219–1233.CrossRefPubMed
49.
go back to reference Tickler AK, Smith DG, Ciccotosto GD, Tew DJ, Curtain CC, Carrington D, Masters CL, Bush AI, Cherny RA, Cappai R, et al.: Methylation of the imidazole side chains of the Alzheimer disease amyloid-beta peptide results in abolition of superoxide dismutase-like structures and inhibition of neurotoxicity. J Biol Chem 2005, 280: 13355–13363. 10.1074/jbc.M414178200CrossRefPubMed Tickler AK, Smith DG, Ciccotosto GD, Tew DJ, Curtain CC, Carrington D, Masters CL, Bush AI, Cherny RA, Cappai R, et al.: Methylation of the imidazole side chains of the Alzheimer disease amyloid-beta peptide results in abolition of superoxide dismutase-like structures and inhibition of neurotoxicity. J Biol Chem 2005, 280: 13355–13363. 10.1074/jbc.M414178200CrossRefPubMed
50.
go back to reference Curtain CC, Ali F, Volitakis I, Cherny RA, Norton RS, Beyreuther K, Barrow CJ, Masters CL, Bush AI, Barnham KJ: Alzheimer’s disease amyloid-beta binds copper and zinc to generate an allosterically ordered membrane-penetrating structure containing superoxide dismutase-like subunits. J Biol Chem 2001, 276: 20466–20473. 10.1074/jbc.M100175200CrossRefPubMed Curtain CC, Ali F, Volitakis I, Cherny RA, Norton RS, Beyreuther K, Barrow CJ, Masters CL, Bush AI, Barnham KJ: Alzheimer’s disease amyloid-beta binds copper and zinc to generate an allosterically ordered membrane-penetrating structure containing superoxide dismutase-like subunits. J Biol Chem 2001, 276: 20466–20473. 10.1074/jbc.M100175200CrossRefPubMed
51.
go back to reference Maiti NC, Jiang DL, Wain AJ, Patel S, Dinh KL, Zhou FM: Mechanistic studies of Cu(II) binding to amyloid-beta peptides and the fluorescence and redox behaviors of the resulting complexes. J Phys Chem B 2008, 112: 8406–8411. 10.1021/jp802038pCrossRefPubMed Maiti NC, Jiang DL, Wain AJ, Patel S, Dinh KL, Zhou FM: Mechanistic studies of Cu(II) binding to amyloid-beta peptides and the fluorescence and redox behaviors of the resulting complexes. J Phys Chem B 2008, 112: 8406–8411. 10.1021/jp802038pCrossRefPubMed
52.
go back to reference Marquez LA, Dunford HB: Kinetics of oxidation of tyrosine and dityrosine by myeloperoxidase compounds I and II. Implications for lipoprotein peroxidation studies. J Biol Chem 1995, 270: 30434–30440. 10.1074/jbc.270.51.30434CrossRefPubMed Marquez LA, Dunford HB: Kinetics of oxidation of tyrosine and dityrosine by myeloperoxidase compounds I and II. Implications for lipoprotein peroxidation studies. J Biol Chem 1995, 270: 30434–30440. 10.1074/jbc.270.51.30434CrossRefPubMed
53.
go back to reference Reynolds WF, Rhees J, Maciejewski D, Paladino T, Sieburg H, Maki RA, Masliah E: Myeloperoxidase polymorphism is associated with gender specific risk for Alzheimer’s disease. Exp Neurol 1999, 155: 31–41. 10.1006/exnr.1998.6977CrossRefPubMed Reynolds WF, Rhees J, Maciejewski D, Paladino T, Sieburg H, Maki RA, Masliah E: Myeloperoxidase polymorphism is associated with gender specific risk for Alzheimer’s disease. Exp Neurol 1999, 155: 31–41. 10.1006/exnr.1998.6977CrossRefPubMed
54.
go back to reference Zheng L, Roberg K, Jerhammar F, Marcusson J, Terman A: Autophagy of amyloid beta-protein in differentiated neuroblastoma cells exposed to oxidative stress. Neurosci Lett 2006, 394: 184–189. 10.1016/j.neulet.2005.10.035CrossRefPubMed Zheng L, Roberg K, Jerhammar F, Marcusson J, Terman A: Autophagy of amyloid beta-protein in differentiated neuroblastoma cells exposed to oxidative stress. Neurosci Lett 2006, 394: 184–189. 10.1016/j.neulet.2005.10.035CrossRefPubMed
55.
go back to reference Yu WH, Cuervo AM, Kumar A, Peterhoff CM, Schmidt SD, Lee JH, Mohan PS, Mercken M, Farmery MR, Tjernberg LO, et al.: Macroautophagy–a novel Beta-amyloid peptide-generating pathway activated in Alzheimer’s disease. J Cell Biol 2005, 171: 87–98. 10.1083/jcb.200505082PubMedCentralCrossRefPubMed Yu WH, Cuervo AM, Kumar A, Peterhoff CM, Schmidt SD, Lee JH, Mohan PS, Mercken M, Farmery MR, Tjernberg LO, et al.: Macroautophagy–a novel Beta-amyloid peptide-generating pathway activated in Alzheimer’s disease. J Cell Biol 2005, 171: 87–98. 10.1083/jcb.200505082PubMedCentralCrossRefPubMed
56.
go back to reference Yang AJ, Chandswangbhuvana D, Margol L, Glabe CG: Loss of endosomal/lysosomal membrane impermeability is an early event in amyloid A beta 1–42 pathogenesis. J Neurosci Res 1998, 52: 691–698. 10.1002/(SICI)1097-4547(19980615)52:6<691::AID-JNR8>3.0.CO;2-3CrossRefPubMed Yang AJ, Chandswangbhuvana D, Margol L, Glabe CG: Loss of endosomal/lysosomal membrane impermeability is an early event in amyloid A beta 1–42 pathogenesis. J Neurosci Res 1998, 52: 691–698. 10.1002/(SICI)1097-4547(19980615)52:6<691::AID-JNR8>3.0.CO;2-3CrossRefPubMed
57.
go back to reference Barnham KJ, Haeffner F, Ciccotosto GD, Curtain CC, Tew D, Mavros C, Beyreuther K, Carrington D, Masters CL, Cherny RA, et al.: Tyrosine gated electron transfer is key to the toxic mechanism of Alzheimer’s disease beta-amyloid. Faseb J 2004, 18: 1427–1429.PubMed Barnham KJ, Haeffner F, Ciccotosto GD, Curtain CC, Tew D, Mavros C, Beyreuther K, Carrington D, Masters CL, Cherny RA, et al.: Tyrosine gated electron transfer is key to the toxic mechanism of Alzheimer’s disease beta-amyloid. Faseb J 2004, 18: 1427–1429.PubMed
58.
go back to reference Chance B, Sies H, Boveris A: Hydroperoxide metabolism in mammalian organs. Physiol Rev 1979, 59: 527–605.PubMed Chance B, Sies H, Boveris A: Hydroperoxide metabolism in mammalian organs. Physiol Rev 1979, 59: 527–605.PubMed
59.
go back to reference Harman D: Aging: a theory based on free radical and radiation chemistry. Sci Aging Knowledge Environ 2002, 2002: cp14. Harman D: Aging: a theory based on free radical and radiation chemistry. Sci Aging Knowledge Environ 2002, 2002: cp14.
60.
go back to reference Murakami K, Shimizu T: Cytoplasmic superoxide radical: a possible contributing factor to intracellular Abeta oligomerization in Alzheimer disease. Comm Integ Biol 2012, 5: 255–258. 10.4161/cib.19548CrossRef Murakami K, Shimizu T: Cytoplasmic superoxide radical: a possible contributing factor to intracellular Abeta oligomerization in Alzheimer disease. Comm Integ Biol 2012, 5: 255–258. 10.4161/cib.19548CrossRef
61.
go back to reference Umeda T, Tomiyama T, Sakama N, Tanaka S, Lambert MP, Klein WL, Mori H: Intraneuronal amyloid beta oligomers cause cell death via endoplasmic reticulum stress, endosomal/lysosomal leakage, and mitochondrial dysfunction in vivo. J Neurosci Res 2011, 89: 1031–1042. 10.1002/jnr.22640CrossRefPubMed Umeda T, Tomiyama T, Sakama N, Tanaka S, Lambert MP, Klein WL, Mori H: Intraneuronal amyloid beta oligomers cause cell death via endoplasmic reticulum stress, endosomal/lysosomal leakage, and mitochondrial dysfunction in vivo. J Neurosci Res 2011, 89: 1031–1042. 10.1002/jnr.22640CrossRefPubMed
62.
go back to reference Misonou H, Morishima-Kawashima M, Ihara Y: Oxidative stress induces intracellular accumulation of amyloid Œ ≤ − protein (AŒ≤) in human neuroblastoma cells. Biochemistry-Us 2000, 39: 6951–6959. 10.1021/bi000169pCrossRef Misonou H, Morishima-Kawashima M, Ihara Y: Oxidative stress induces intracellular accumulation of amyloid Œ ≤ − protein (AŒ≤) in human neuroblastoma cells. Biochemistry-Us 2000, 39: 6951–6959. 10.1021/bi000169pCrossRef
63.
go back to reference Kurz T, Terman A, Gustafsson B, Brunk UT: Lysosomes and oxidative stress in aging and apoptosis. Biochim Biophys Acta Gen Subj 2008, 1780: 1291–1303. 10.1016/j.bbagen.2008.01.009CrossRef Kurz T, Terman A, Gustafsson B, Brunk UT: Lysosomes and oxidative stress in aging and apoptosis. Biochim Biophys Acta Gen Subj 2008, 1780: 1291–1303. 10.1016/j.bbagen.2008.01.009CrossRef
64.
go back to reference Friedrich RP, Tepper K, Ronicke R, Soom M, Westermann M, Reymann K, Kaether C, Fandrich M: Mechanism of amyloid plaque formation suggests an intracellular basis of Abeta pathogenicity. Proc Natl Acad Sci USA 2010, 107: 1942–1947. 10.1073/pnas.0904532106PubMedCentralCrossRefPubMed Friedrich RP, Tepper K, Ronicke R, Soom M, Westermann M, Reymann K, Kaether C, Fandrich M: Mechanism of amyloid plaque formation suggests an intracellular basis of Abeta pathogenicity. Proc Natl Acad Sci USA 2010, 107: 1942–1947. 10.1073/pnas.0904532106PubMedCentralCrossRefPubMed
65.
go back to reference Roher AE, O’Chaney M, Kuo Y, Webster SD, Stine WB, Haverkamp LJ, Woods AS, Cotter RJ, Tuohy JM, Krafft GA, et al.: Morphology and toxicity of Ab-(1–42) dimer derived from neuritic and vascular amyloid deposits of Alzheimer’s disease. J Biol Chem 1996, 271: 20631–20635. 10.1074/jbc.271.34.20631CrossRefPubMed Roher AE, O’Chaney M, Kuo Y, Webster SD, Stine WB, Haverkamp LJ, Woods AS, Cotter RJ, Tuohy JM, Krafft GA, et al.: Morphology and toxicity of Ab-(1–42) dimer derived from neuritic and vascular amyloid deposits of Alzheimer’s disease. J Biol Chem 1996, 271: 20631–20635. 10.1074/jbc.271.34.20631CrossRefPubMed
66.
go back to reference Cherny RA, Atwood CS, Xilinas ME, Gray DN, Jones WD, McLean CA, Barnham KJ, Volitakis I, Fraser FW, Kim YS, et al.: Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimer’s disease transgenic mice. Neuron 2001, 30: 665–676. 10.1016/S0896-6273(01)00317-8CrossRefPubMed Cherny RA, Atwood CS, Xilinas ME, Gray DN, Jones WD, McLean CA, Barnham KJ, Volitakis I, Fraser FW, Kim YS, et al.: Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimer’s disease transgenic mice. Neuron 2001, 30: 665–676. 10.1016/S0896-6273(01)00317-8CrossRefPubMed
67.
go back to reference Ritchie CW, Bush AI, Mackinnon A, Macfarlane S, Mastwyk M, MacGregor L, Kiers L, Cherny R, Li QX, Tammer A, et al.: Metal-protein attenuation with iodochlorhydroxyquin (clioquinol) targeting Abeta amyloid deposition and toxicity in Alzheimer disease: a pilot phase 2 clinical trial. Arch Neurol 2003, 60: 1685–1691. 10.1001/archneur.60.12.1685CrossRefPubMed Ritchie CW, Bush AI, Mackinnon A, Macfarlane S, Mastwyk M, MacGregor L, Kiers L, Cherny R, Li QX, Tammer A, et al.: Metal-protein attenuation with iodochlorhydroxyquin (clioquinol) targeting Abeta amyloid deposition and toxicity in Alzheimer disease: a pilot phase 2 clinical trial. Arch Neurol 2003, 60: 1685–1691. 10.1001/archneur.60.12.1685CrossRefPubMed
68.
go back to reference Ahmed N, Ahmed U, Thornalley PJ, Hager K, Fleischer G, Munch G: Protein glycation, oxidation and nitration adduct residues and free adducts of cerebrospinal fluid in Alzheimer’s disease and link to cognitive impairment. J Neurochem 2005, 92: 255–263. 10.1111/j.1471-4159.2004.02864.xCrossRefPubMed Ahmed N, Ahmed U, Thornalley PJ, Hager K, Fleischer G, Munch G: Protein glycation, oxidation and nitration adduct residues and free adducts of cerebrospinal fluid in Alzheimer’s disease and link to cognitive impairment. J Neurochem 2005, 92: 255–263. 10.1111/j.1471-4159.2004.02864.xCrossRefPubMed
69.
go back to reference Abdelrahim M, Morris E, Carver J, Facchina S, White A, Verma A: Liquid chromatographic assay of dityrosine in human cerebrospinal fluid. J Chromatogr B 1997, 696: 175–182. 10.1016/S0378-4347(97)00248-XCrossRef Abdelrahim M, Morris E, Carver J, Facchina S, White A, Verma A: Liquid chromatographic assay of dityrosine in human cerebrospinal fluid. J Chromatogr B 1997, 696: 175–182. 10.1016/S0378-4347(97)00248-XCrossRef
70.
go back to reference Maruyama M, Arai H, Sugita M, Tanji H, Higuchi M, Okamura N, Matsui T, Higuchi S, Matsushita S, Yoshida H, Sasaki H: Cerebrospinal fluid amyloid beta(1–42) levels in the mild cognitive impairment stage of Alzheimer’s disease. Exp Neurol 2001, 172: 433–436. 10.1006/exnr.2001.7814CrossRefPubMed Maruyama M, Arai H, Sugita M, Tanji H, Higuchi M, Okamura N, Matsui T, Higuchi S, Matsushita S, Yoshida H, Sasaki H: Cerebrospinal fluid amyloid beta(1–42) levels in the mild cognitive impairment stage of Alzheimer’s disease. Exp Neurol 2001, 172: 433–436. 10.1006/exnr.2001.7814CrossRefPubMed
Metadata
Title
A central role for dityrosine crosslinking of Amyloid-β in Alzheimer’s disease
Authors
Youssra K Al-Hilaly
Thomas L Williams
Maris Stewart-Parker
Lenzie Ford
Eldhose Skaria
Michael Cole
William Grant Bucher
Kyle L Morris
Alaa Abdul Sada
Julian R Thorpe
Louise C Serpell
Publication date
01-12-2013
Publisher
BioMed Central
Published in
Acta Neuropathologica Communications / Issue 1/2013
Electronic ISSN: 2051-5960
DOI
https://doi.org/10.1186/2051-5960-1-83

Other articles of this Issue 1/2013

Acta Neuropathologica Communications 1/2013 Go to the issue