You are on page 1of 4

View Article Online / Journal Homepage / Table of Contents for this issue

Metallomics Dynamic Article Links

Cite this: Metallomics, 2011, 3, 267–270

www.rsc.org/metallomics MINIREVIEW
Role of metal dyshomeostasis in Alzheimer’s diseasew
David J. Bonda,a Hyoung-gon Lee,a Jeffrey A. Blair,a Xiongwei Zhu,*a
George Perryab and Mark A. Smitha
Received 10th November 2010, Accepted 26th January 2011
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0MT00074D

DOI: 10.1039/c0mt00074d
Downloaded by North Carolina State University on 11 January 2013

Despite serving a crucial purpose in neurobiological function, transition metals play a sinister part
in the aging brain, where the abnormal accumulation and distribution of reactive iron, copper,
and zinc elicit oxidative stress and macromolecular damage that impedes cellular function.
Alzheimer’s disease (AD), an age-related neurodegenerative condition, presents marked
accumulations of oxidative stress-induced damage, and increasing evidence points to aberrant
transition metal homeostasis as a critical factor in its pathogenesis. Amyloid-b oligomerization
and fibrillation, considered by many to be the precipitating factor underlying AD onset and
development, is also induced by abnormal transition metal activity. We here elaborate on the
roles of iron, copper, and zinc in AD and describe the therapeutic implications they present.

A Introduction biomolecular cascades that aggregate to produce cognitive


decline, severe memory impairment, and ultimately death.1
Alzheimer’s disease (AD) is a progressive and fatal neuro- These cascades initially occur within a defined region of the
degenerative condition that affects 35 million people world- brain, namely the mediotemporal lobe, before spreading
wide.1 Though early-onset, familial AD may be attributed to cortically outward to the neocortex. The peculiar consistency
mutations in several known chromosomal regions, the much and predictability with which such anatomic dysfunction
more prevalent late-onset, sporadic AD is far less well progress is at present a mystery. Importantly, the patho-
characterized. Sporadic AD accounts for approximately physiological end-products of familial and sporadic AD are
95% of all AD cases and results from a complex array of quite similar despite their differing causations and ages of
onset (40–60 years old for the former, 65 and older for the
a
Department of Pathology, Case Western Reserve University, latter).1
Cleveland, Ohio, USA. E-mail: Xiongwei.zhu@case.edu;
Fax: +1 216-368-8964; Tel: +1 216-368-5903 AD is pathologically characterized by widespread oxidative
b
UTSA Neurosciences Institute and Department of Biology, stress, neuroinflammation, calcium dysregulation, mitochondrial
University of Texas at San Antonio, San Antonio, Texas, USA malformation and altered distribution, neurofibrillary tangle
w This article is published as part of a themed issue on Metals in
(NFT) formation, amyloid-b (Ab) oligomerization, synaptic
Neurodegenerative Diseases, Guest Edited by David Brown.
toxicity, and metal dyshomeostasis.1 Despite an extensive
understanding of each of these phenomena individually as
they occur within the cell, an adequate explanation for their
We are deeply saddened by the
origins, interactions, and evolution as they pertain to AD is
unexpected death of Mark A.
Smith, PhD, Professor of lacking. Oxidative stress undoubtedly plays a critical role, as
Pathology at Case Western evidence for its molecular impact exists very early in disease
Reserve University, on progression.2,3 Similarly, Ab, perhaps the most studied facet of
Sunday, December 19, 2010. AD pathology, confers striking neurotoxicity to neurons
He is survived by his wife, and is certainly an important aspect of the disease. Notably,
Gemma, his sons, his father, both phenomena (i.e., oxidative stress and Ab accumulation)
sisters, his lab family, as well share a common instigating factor—reactive transition metal
as numerous friends and aberration.
colleagues. Mark was a Transition metals, though crucial to many biochemical
devoted husband and father,
neuronal processes, are abnormally distributed in AD.4,5
beloved colleague, dedicated
mentor, and dear friend to so Imbalances in aluminium, silicon, lead, mercury, zinc, iron,
Mark A. Smith many. His passing is a tragic and copper have been reported in AD, and the latter three are
loss and he will remain in our known to be elevated in AD neuropil.6 Furthermore, in situ
hearts and serve as an inspiration in our pursuit to advance the iron detection has revealed a marked association between
health of humankind. redox-active iron and both NFTs and Ab-rich senile plaques.4

This journal is c The Royal Society of Chemistry 2011 Metallomics, 2011, 3, 267–270 267
View Article Online

Iron, copper, and zinc are also known to accumulate in high upregulation in response to increased iron stores.17 APP seems
concentrations within the core and peripheral areas of senile to facilitate iron export from cells, likely in an attempt to
plaques such that tissue exposure to metal-selective chelators minimize potential ROS generation by the presence of reactive
prevents lesion detection.7 The extensive role of metals in iron. APP has also been reported to prevent the release of
AD-type neurodegeneration is quite compelling and provides iron(II) from heme,18 thereby further reducing the toxic
a potential window for therapeutic intervention in AD. accumulation of redox-active iron, and exogenous iron load
reportedly promotes the a-secretase cleavage of APP, which
generates a soluble, potentially neuroprotective Ab peptide.
B Metals and oxidative stress production
Transition metal ions, however, also accelerate Ab40 and
Oxidative stress occurs when cellular antioxidant defense Ab42 aggregation in vitro and contribute to their toxicity.19,20
mechanisms become overwhelmed by reactive oxidative Specifically, aluminium, iron, and zinc, but not cobalt,
species (ROS) production such that macromolecular damage manganese, magnesium, calcium, sodium, or potassium, elicit
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0MT00074D

results. Stochastic modifications in DNA, RNA, membrane increased Ab aggregation. A mildly acidic environment with
proteins, and phospholipids by free radicals impose increased copper(II) and zinc(II)—common features of
deleterious consequences to cellular functioning and can inflammation and head trauma—also induce aggregation of
Downloaded by North Carolina State University on 11 January 2013

potentially initiate cascades of molecular aberration within Ab40.20,21 As stated above, iron, copper, and zinc are
the cell. Notably, ROS are inevitable byproducts of oxidative associated with Ab plaques in vitro, and reports on Ab-copper
phosphorylation,8 and oxidative stress plays a large role in binding have revealed a corresponding generation of hydrogen
normal aging as well as in neurodegenerative disease.9 AD is peroxide: a significant contributor to oxidative damage in the
marked by elevations in oxidatively damaged RNA, DNA, brain.22 Similarly, studies have shown that the coordination of
proteins, and phospholipids,10 and these damages temporally oxidized copper (Cu(II)) or iron (Fe(III)) to Ab result in the
precede the appearance of hallmark AD pathologies, such as reduction of these ions and thus their ability to engage in
NFT deposition and Ab aggregation.3,11 ROS-generating chemistry.23 Zinc(II), along with inducing Ab
Transition metals, such as iron and copper, can facilitate the aggregation, seems to simultaneously inhibit the ferroxidase
generation of free radicals in vivo.12 The Fenton reaction activity of APP mentioned above,17 thus further contributing
between reduced transition metals (typically iron(II) or to aberrant iron accumulation and ROS-generation. Taken
copper(II)) and hydrogen peroxide is particularly harmful, as together, the role of transition metal accumulation and
it yields the hydroxyl radical: a highly reactive oxidative Ab deposition invites scrutiny for therapeutic intervention.24
species with a limited diffusion distance.7 RNA-bound iron Several agents have indeed been identified, and testing is
poses a significant threat to neuronal viability in vulnerable underway.
neurons in AD. Its oxidation via the Fenton reaction produces
abnormalities in RNA, and in AD, ribosomal RNA is D Therapeutic considerations: metal chelation
particularly affected, yielding a great reduction in protein
therapy
synthesis.13
Copper exerts similar oxidative disturbances in neuronal The ubiquitous role of transition metals in AD-type neuro-
tissue. Its interactions with the regulatory protein cerulo- degeneration has received a great deal of attention in the field
plasmin, which converts redox-active iron(II) to a less reactive and has prompted the discovery of agents that prevent their
iron(III), suggests a role in oxidative damage in AD, as copper toxic actions in vivo. Metal-chelating compounds that
is increased in AD brains.13 Copper also interacts with selectively bind to and remove, or redox-silence, transition
amyloid-b protein precursor (APP) in an electron transfer metals are particularly attractive, and several candidates have
reaction that reduces copper(II) to copper(I), enhancing the reached clinical trials. Desferrioxamine (DFO), for instance,
production of a hydroxyl radical intermediate.13 Manganese, an FDA-approved drug for iron overload disease, has
zinc, and aluminium have also been implicated in free radical impeded the progression of AD in clinical trials, purportedly
generation in AD,14 and their altered concentrations and due to its chelation and clearance of iron(III) from the brain.25
distributions in AD brains15 suggests their importance in DFO also moderately binds aluminium, zinc, and copper,26,27
disease pathogenesis. which may contribute to the AD-attenuating effects of the
drug. The large molecular weight and hydrophilicity of DFO,
however, impedes its ability to cross the blood-brain barrier
C Metals and Ab
(BBB), and thus the positive clinical results are due to long,
Ab peptides are the product of the constitutive proteolytic subcutaneous administration of the compound.27,28 Likewise,
cleavage of APP by b- and g-secretases and may be 38, 40, or Deferiprone, or L1, is an iron and aluminium chelator that is
42 amino acids in length (Ab38, Ab40, Ab42, respectively). The approved for therapeutic use in Europe, but not in the United
biological functions of these cleavage products are unknown, States.29 It has moderate chelating effects, but its small size
however their deposition and aggregation in AD is extensively and lipophilicity, in contrast to DFO, renders it somewhat
documented. Interestingly, APP has been found to contain an toxic when administered orally. Specifically, the ease with
iron responsive element (IRE) on the 5 0 untranslated region of which L1 penetrates the BBB enables it to quickly remove
its mRNA with sequence homology to the IRE of the iron- iron from intracellular pools, thereby eliminating the supply of
storage ferritin protein.16,17 Studies on primary neuron iron needed for human ribonucleotide reductase, which is
cultures indicate a ferroxidase activity of APP, noting its required for DNA synthesis and cell proliferation.30 Even

268 Metallomics, 2011, 3, 267–270 This journal is c The Royal Society of Chemistry 2011
View Article Online

more, L1 and other small lipophilic metal chelators, are In vitro studies on nanoparticle delivery of metal chelators
thought to directly penetrate the ribonucleotide reductase has demonstrated an effective prevention of Ab aggregation
enzyme and physically remove the iron from within, thus and toxicity.35 Cells cultured with Ab plus nanoparticle
directly inhibiting enzyme functioning and DNA synthesis.31 chelator conjugates were salvaged from Ab-induced cell death,
These hindering factors have ultimately led to the development and cells cultured with only nanoparticle compounds did not
of nanaoparticle delivery systems of metal chelators that suffer any toxic side effects. Although more research is
enable a safe and effective administration of chelating necessary before any conclusions are made, it seems that
compounds for AD. nanoparticle delivery of metal chelators is a viable candidate
Nanoparticle delivery involves the binding of polymeric for AD therapy.
particles (ranging in size from 10–1000 nm) to drug
compounds such that the latter may be delivered to physio-
logical regions typically prohibitive of their entry.25 DFO, for Conclusions
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0MT00074D

instance, a large, hydrophilic molecule incapable of crossing A delicate balance of transition metal activity is necessary for
the BBB, may be bound to a nanoparticle that is able to breach prolonged neuronal functioning. As with any physical system,
the lipid barrier and may thus enter the brain regions that errors inevitably occur, and within the brain, transition metals
Downloaded by North Carolina State University on 11 January 2013

necessitate metal chelation. Nanoparticle delivery offers gradually become disproportionately positioned such that
several benefits to chelation therapy that could otherwise not impaired cellular functioning results. AD presents a strong
be achieved: (1) the chelator needs not be lipophilic to cross correlation with age and is certainly the result of such
the BBB and enter the brain if it is bound to a nanoparticle, aberration in metal homeostasis, at least partly. Transition
thereby eliminating the use of small chelators with toxic side metal chelation therefore provides a method of therapeutic
effects; (2) the nanoparticle system enables large, lipophobic intervention that targets early-occurring disturbances in
molecules like DFO to enter the BBB without prolonged, transition metal activity. Such sequestration would ultimately
invasive means of administration; and (3) the mobility of the help prevent the generation and accumulation of ROS that
nanoparticle through the BBB facilitates its removal from the incur oxidative damage to neuronal tissue and would simulta-
brain following chelation. This latter phenomenon is crucial to neously prevent the aggregation and deposition of Ab peptide.
the effectiveness of any chelation system, as a redox-silenced Furthermore, the nanoparticle delivery system provides a
transition metal must be removed from its site of action before method of chelator administration that ensures an effective
it becomes reactivated.25 Interestingly, even the small, bioactivity with little or no adverse consequences. There is
lipophilic chelators do not possess the ability to exit the substantial need for an efficient and early preventative measure
BBB once they have complexed with a metal substrate due for AD in the rapidly aging population, and targeting
to their altered lipophilicity following transition metal transition metals in the brain, particularly with nanoparticle
binding.25,32 conjugates, is an increasingly attractive means of doing so.
The method of BBB penetration of nanoparticle conjugates
is not fully understood. Several scenarios have been identified,
all of which likely act in combination to yield the desired Acknowledgements
effect. These include: (1) increased retention of nanoparticles
Work in the authors’ laboratories is supported by the National
in blood-brain capillaries combined with absorption into
Institutes of Health (AG0248028) and the Alzheimer’s
capillary walls, creating a higher concentration gradient and
Association.
enhancing their transport across endothelial cell layers into the
brain; (2) enhanced drug permeability due to a surfactant
effect; (3) tight junction opening between endothelial cells; Notes and references
(4) transcytosis of nanoparticle compound through the
1 R. J. Castellani, R. K. Rolston and M. A. Smith, Alzheimer
endothelial cell layer; (5) an inhibition of the efflux system; disease, Dis. Mon., 2010, 56, 484–546.
and (6) endocytosis of the nanoparticle-chelator conjugate.33 2 H. G. Lee, G. Casadesus, X. Zhu, A. Takeda, G. Perry and
Notably, evidence for the endocytosis of the conjugate is the M. A. Smith, Challenging the amyloid cascade hypothesis: senile
most compelling. When coated with polysorbate-80, nano- plaques and amyloid-beta as protective adaptations to Alzheimer
disease, Ann. N. Y. Acad. Sci., 2004, 1019, 1–4.
particles absorb the trafficking protein apolipoprotein E 3 R. J. Castellani, H. G. Lee, S. L. Siedlak, A. Nunomura,
(ApoE) onto their surface in a manner that coincides with T. Hayashi, M. Nakamura, X. Zhu, G. Perry and M. A. Smith,
the BBB penetration of the conjugate.34 ApoE absorption Reexamining Alzheimer’s disease: evidence for a protective role for
amyloid-beta protein precursor and amyloid-beta, J. Alzheimers
facilitates the endocytosis of low-density lipoprotein (LDL)
Dis., 2009, 18, 447–452.
across the BBB via LDL receptor-mediated endocytosis,33 4 M. A. Smith, P. L. Harris, L. M. Sayre and G. Perry, Iron
and thus polysorbate-80-coated nanoparticles may mimic accumulation in Alzheimer disease is a source of redox-generated
LDL via ApoE absorption and utilize LDL receptor- free radicals, Proc. Natl. Acad. Sci. U. S. A., 1997, 94, 9866–9868.
5 M. A. Smith, K. Wehr, P. L. Harris, S. L. Siedlak, J. R. Connor
mediated endocytosis to penetrate the BBB. This ApoE and G. Perry, Abnormal localization of iron regulatory protein in
mimetic system may also be responsible for the ability of the Alzheimer’s disease, Brain Res., 1998, 788, 232–236.
nanoparticle-chelator-metal conjugate to exit the brain as 6 M. A. Lovell, J. D. Robertson, W. J. Teesdale, J. L. Campbell and
needed. In any case, the nanoparticle delivery system seems W. R. Markesbery, Copper, iron and zinc in Alzheimer’s disease
senile plaques, J. Neurol. Sci., 1998, 158, 47–52.
to offer a safe and effective method of transition metal 7 L. M. Sayre, G. Perry, P. L. Harris, Y. Liu, K. A. Schubert and
chelation. M. A. Smith, In situ oxidative catalysis by neurofibrillary tangles

This journal is c The Royal Society of Chemistry 2011 Metallomics, 2011, 3, 267–270 269
View Article Online

and senile plaques in Alzheimer’s disease: a central role for bound model of Alzheimer’s disease, J. Neurochem., 2010, 114,
transition metals, J. Neurochem., 2000, 74, 270–279. 1630–1638, DOI: 10.1111/j.1471-4159.2010.06888.x.
8 G. Perry, L. M. Sayre, C. S. Atwood, R. J. Castellani, A. D. Cash, 22 D. J. Bonda, X. Wang, G. Perry, A. Nunomura, M. Tabaton,
C. A. Rottkamp and M. A. Smith, The role of iron and copper in X. Zhu and M. A. Smith, Oxidative stress in Alzheimer disease: a
the aetiology of neurodegenerative disorders: therapeutic implica- possibility for prevention, Neuropharmacology, 2010, 59, 290–294.
tions, CNS Drugs, 2002, 16, 339–352. 23 C. Hureau and P. Faller, Abeta-mediated ROS production by Cu
9 P. M. Doraiswamy and A. E. Finefrock, Metals in our minds: ions: structural insights, mechanisms and relevance to Alzheimer’s
therapeutic implications for neurodegenerative disorders, Lancet disease, Biochimie, 2009, 91, 1212–1217, DOI: 10.1016/
Neurol., 2004, 3, 431–434. j.biochi.2009.03.013.
10 X. Zhu, R. J. Castellani, A. Takeda, A. Nunomura, C. S. Atwood, 24 J. A. Duce and A. I. Bush, Biological metals and Alzheimer’s
G. Perry and M. A. Smith, Differential activation of neuronal disease: implications for therapeutics and diagnostics, Prog.
ERK, JNK/SAPK and p38 in Alzheimer disease: the ’two hit’ Neurobiol., 2010, 92, 1–18, DOI: 10.1016/j.pneurobio.2010.04.003.
hypothesis, Mech. Ageing Dev., 2001, 123, 39–46; X. Zhu, 25 G. Liu, P. Men, G. Perry and M. A. Smith, Nanoparticle and iron
H. G. Lee, G. Perry and M. A. Smith, Alzheimer disease, the chelators as a potential novel Alzheimer therapy, Methods Mol.
two-hit hypothesis: an update, Biochim. Biophys. Acta, 2007, 1772, Biol., 2010, 610, 123–144.
Published on 07 February 2011 on http://pubs.rsc.org | doi:10.1039/C0MT00074D

494–502; X. Zhu, A. K. Raina, G. Perry and M. A. Smith, 26 H. Keberle, The Biochemistry Of Desferrioxamine And Its
Alzheimer’s disease: the two-hit hypothesis, Lancet Neurol., 2004, Relation To Iron Metabolism, Ann. N. Y. Acad. Sci., 1964, 119,
3, 219–226. 758–768; D. R. McLachlan, T. P. Kruck, W. J. Lukiw and
11 H. G. Lee, X. Zhu, R. J. Castellani, A. Nunomura, G. Perry and S. S. Krishnan, Would decreased aluminium ingestion reduce the
Downloaded by North Carolina State University on 11 January 2013

M. A. Smith, Amyloid-beta in Alzheimer disease: the null versus the incidence of Alzheimer’s disease?, CMAJ, 1991, 145, 793–804;
alternate hypotheses, J. Pharmacol. Exp. Ther., 2007, 321, 823–829. M. P. Cuajungco, K. Y. Faget, X. Huang, R. E. Tanzi and
12 C. W. Olanow, An introduction to the free radical hypothesis in A. I. Bush, Metal chelation as a potential therapy for Alzheimer’s
Parkinson’s disease, Ann. Neurol., 1992, 32(Suppl.), S2–9; disease, Ann. N. Y. Acad. Sci., 2000, 920, 292–304.
B. Halliwell and J. M. C. Gutteridge, Free Radicals in Biology 27 D. R. Richardson and P. Ponka, Development of iron chelators to
and Medicine, Oxford University Press, New York, 1999. treat iron overload disease and their use as experimental tools to
13 X. Zhu, B. Su, X. Wang, M. A. Smith and G. Perry, Causes of probe intracellular iron metabolism, Am. J. Hematol., 1998, 58,
oxidative stress in Alzheimer disease, Cell. Mol. Life Sci., 2007, 64, 299–305.
2202–2210. 28 N. F. Olivieri and G. M. Brittenham, Iron-chelating therapy and
14 X. Huang, R. D. Moir, R. E. Tanzi, A. I. Bush and J. T. Rogers, the treatment of thalassemia, Blood, 1997, 89, 739–761.
Redox-active metals, oxidative stress, and Alzheimer’s disease 29 G. J. Kontoghiorghes, New concepts of iron and aluminium
pathology, Ann. N. Y. Acad. Sci., 2004, 1012, 153–163. chelation therapy with oral L1 (deferiprone) and other chelators
15 B. F. Gh Popescu and H. Nichol, Mapping brain metals to A review, Analyst, 1995, 120, 845–851.
evaluate therapies forneurodegenerative disease, CNS neuroscience 30 R. C. Hider, J. B. Porter and S. Singh, in The development of iron
& therapeutics, 2010, DOI: 10.1111/j.1755-5949.2010.00149.x. chelators for clinical use, ed. R. J. Bergeron and G. M. Brittenham,
16 J. T. Rogers, J. D. Randall, C. M. Cahill, P. S. Eder, X. Huang, CRC, Boca Raton, 1994, pp. 353–371; P. Reichard, Interactions
H. Gunshin, L. Leiter, J. McPhee, S. S. Sarang, T. Utsuki, between deoxyribonucleotide and DNA synthesis, Annu. Rev.
N. H. Greig, D. K. Lahiri, R. E. Tanzi, A. I. Bush, T. Giordano Biochem., 1988, 57, 349–374; J. Stubbe, Ribonucleotide reductases:
and S. R. Gullans, An iron-responsive element type II in the amazing and confusing, J. Biol. Chem., 1990, 265, 5329–5332;
5 0 -untranslated region of the Alzheimer’s amyloid precursor K. P. Hoyes, R. C. Hider and J. B. Porter, Cell cycle synchronization
protein transcript, J. Biol. Chem., 2002, 277, 45518–45528. and growth inhibition by 3-hydroxypyridin-4-one iron chelators in
17 J. A. Duce, A. Tsatsanis, M. A. Cater, S. A. James, E. Robb, leukemia cell lines, Cancer Res., 1992, 52, 4591–4599; C. E. Cooper,
K. Wikhe, S. L. Leong, K. Perez, T. Johanssen, M. A. Greenough, G. R. Lynagh, K. P. Hoyes, R. C. Hider, R. Cammack and
H. H. Cho, D. Galatis, R. D. Moir, C. L. Masters, C. McLean, J. B. Porter, The relationship of intracellular iron chelation to the
R. E. Tanzi, R. Cappai, K. J. Barnham, G. D. Ciccotosto, inhibition and regeneration of human ribonucleotide reductase,
J. T. Rogers and A. I. Bush, Iron-export ferroxidase activity of J. Biol. Chem., 1996, 271, 20291–20299.
beta-amyloid precursor protein is inhibited by zinc in Alzheimer’s 31 C. Gerez and M. Fontecave, Reduction of the small subunit of
disease, Cell, 2010, 142, 857–867. Escherichia coli ribonucleotide reductase by hydrazines and hydroxyl-
18 M. Takahashi, S. Dore, C. D. Ferris, T. Tomita, A. Sawa, amines, Biochemistry, 1992, 31, 780–786.
H. Wolosker, D. R. Borchelt, T. Iwatsubo, S. H. Kim, 32 J. B. Porter, M. Gyparaki, L. C. Burke, E. R. Huehns, P. Sarpong,
G. Thinakaran, S. S. Sisodia and S. H. Snyder, Amyloid precursor V. Saez and R. C. Hider, Iron mobilization from hepatocyte
proteins inhibit heme oxygenase activity and augment neuro- monolayer cultures by chelators: the importance of membrane
toxicity in Alzheimer’s disease, Neuron, 2000, 28, 461–473. permeability and the iron-binding constant, Blood, 1988, 72,
19 P. W. Mantyh, J. R. Ghilardi, S. Rogers, E. DeMaster, C. J. Allen, 1497–1503.
E. R. Stimson and J. E. Maggio, Aluminium, iron, and zinc ions 33 J. Kreuter, Nanoparticulate systems for brain delivery of drugs,
promote aggregation of physiological concentrations of beta- Adv. Drug Delivery Rev., 2001, 47, 65–81.
amyloid peptide, J. Neurochem., 1993, 61, 1171–1174; C. J. Lin, 34 J. Kreuter, D. Shamenkov, V. Petrov, P. Ramge, K. Cychutek,
H. C. Huang and Z. F. Jiang, Cu(II) interaction with amyloid-beta C. Koch-Brandt and R. Alyautdin, Apolipoprotein-mediated
peptide: a review of neuroactive mechanisms in AD brains, Brain transport of nanoparticle-bound drugs across the blood-brain
Res. Bull., 2010, 82, 235–242, DOI: 10.1016/j.brainres- barrier, J. Drug Targeting, 2002, 10, 317–325; R. N. Alyaudtin,
bull.2010.06.003; Y. Miller, B. Ma and R. Nussinov, Zinc ions A. Reichel, R. Lobenberg, P. Ramge, J. Kreuter and D. J. Begley,
promote Alzheimer Abeta aggregation via population shift of Interaction of poly(butylcyanoacrylate) nanoparticles with the
polymorphic states, Proc. Natl. Acad. Sci. U. S. A., 2010, 107, blood-brain barrier in vivo and in vitro, J. Drug Targeting, 2001,
9490–9495, DOI: 10.1073/pnas.0913114107. 9, 209–221; J. Kreuter, R. N. Alyautdin, D. A. Kharkevich and
20 T. Marino, N. Russo, M. Toscano and M. Pavelka, On the metal A. A. Ivanov, Passage of peptides through the blood-brain barrier
ion (Zn(2+), Cu(2+)) coordination with beta-amyloid peptide: with colloidal polymer particles (nanoparticles), Brain Res., 1995,
DFT computational study, Interdiscip. Sci., 2010, 2, 57–69, DOI: 674, 171–174; P. Ramge, R. E. Unger, J. B. Oltrogge, D. Zenker,
10.1007/s12539-010-0086-x. D. Begley, J. Kreuter and H. Von Briesen, Polysorbate-80 coating
21 C. S. Atwood, R. D. Moir, X. Huang, R. C. Scarpa, enhances uptake of polybutylcyanoacrylate (PBCA)-nanoparticles
N. M. Bacarra, D. M. Romano, M. A. Hartshorn, R. E. Tanzi by human and bovine primary brain capillary endothelial cells,
and A. I. Bush, Dramatic aggregation of Alzheimer abeta by Cu(II) Eur. J. Neurosci., 2000, 12, 1931–1940.
is induced by conditions representing physiological acidosis, 35 G. Liu, P. Men, G. Perry and M. A. Smith, Chapter 5 - Develop-
J. Biol. Chem., 1998, 273, 12817–12826; Z. Zheng, C. White, ment of iron chelator-nanoparticle conjugates as potential
J. Lee, T. S. Peterson, A. I. Bush, G. Y. Sun, G. A. Weisman therapeutic agents for Alzheimer disease, Prog. Brain Res., 2009,
and M. J. Petris, Altered microglial copper homeostasis in a mouse 180, 97–108.

270 Metallomics, 2011, 3, 267–270 This journal is c The Royal Society of Chemistry 2011

You might also like