G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Journal of Trace Elements in Medicine and Biology

jou rnal homepage: www.elsevier.de/jtemb

10th NTES Symposium

Review

The of , and in Parkinson’s and

Wilson’s diseases

a,b,∗ c,d e f

Petr Dusek , Per M. Roos , Tomasz Litwin , Susanne A. Schneider ,

g h

Trond Peder Flaten , Jan Aaseth

a

Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague,

1st Faculty of Medicine and General University Hospital in Prague, Czech Republic

b

Institute of Neuroradiology, University Medicine Göttingen, Göttingen, Germany

c

Department of Neurology, Division of Clinical Neurophysiology, Oslo University Hospital, Oslo, Norway

d

Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden

e

2nd Department of Neurology, Institute of Psychiatry and Neurology, Warsaw, Poland

f

Neurology Department, University of Kiel, Kiel, Germany

g

Department of Chemistry, Norwegian University of Science and Technology, Trondheim, Norway

h

Department of Medicine, Innlandet Hospital Trust, Kongsvinger Hospital Division, Kongsvinger, Norway

a r t i c l e i n f o a b s t r a c t

Article history: Impaired cellular homeostasis of metals, particularly of Cu, Fe and Mn may trigger

Received 31 January 2014

through various mechanisms, notably induction of , promotion of ␣-synuclein aggrega-

Accepted 22 May 2014

tion and fibril formation, activation of microglial cells leading to inflammation and impaired production

of . In this article we review available studies concerning Fe, Cu and Mn in Parkinson’s

Keywords:

disease and Wilson’s disease. In Parkinson’s disease local dysregulation of iron in the subs-

Iron

tantia nigra (SN) seems to be related to neurodegeneration with an increase in SN iron concentration,

Copper

accompanied by decreased SN Cu and ceruloplasmin concentrations and increased free Cu concentrations

Manganese

and decreased ferroxidase activity in the cerebrospinal fluid. Available data in Wilson’s disease suggest

Parkinson’s disease

that substantial increases in CNS Cu concentrations persist for a long time during chelating treatment

Wilson’s disease

Chelating agents and that local accumulation of Fe in certain brain nuclei may occur during the course of the disease.

Consequences for chelating treatment strategies are discussed.

© 2014 Elsevier GmbH. All rights reserved.

Contents

Introduction ...... 00

Parkinson’s disease (PD) ...... 00

Environmental exposure...... 00

Animal models ...... 00

Dysregulation of brain metal homeostasis ...... 00

Wilson’s disease (WD) ...... 00

Local dysregulation of metal homeostasis...... 00

Animal models ...... 00

Concluding remarks ...... 00

Conflict of interest ...... 00

Acknowledgments ...... 00

References ...... 00

Corresponding author at: Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University

Hospital in Prague, Katerinska 30, 120 00 Praha 2, Czech Republic. Tel.: +420 224 965 528.

E-mail addresses: [email protected], [email protected] (P. Dusek).

http://dx.doi.org/10.1016/j.jtemb.2014.05.007

0946-672X/© 2014 Elsevier GmbH. All rights reserved.

G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

2 P. Dusek et al. / Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx

Introduction Parkinson’s disease (PD)

Impaired cellular homeostasis of metals may initiate neurode- Accumulation of several metals, including Fe, Cu and Mn in subs-

generation through various mechanisms. Of these, oxidative stress tantia nigra (SN) has been suggested to play a causative role in the

induced by the formation of free radicals is the best established [1]. pathophysiology of PD [34]. Metal related PD hypotheses are based

Other possible mechanisms are impaired production of metallopro- on:

teins [2,3], activation of microglial cells leading to inflammation [4]

and promotion of aggregation and fibril formation of ␣-synuclein, (1) Epidemiological studies assessing the relationship between

a highly conserved protein with unknown function. This protein environmental exposure to metals and the risk of PD.

is the major constituent of Lewy bodies found as intracytoplas- (2) Clinical observations that disorders with brain metal accu-

matic inclusions in Parkinson’s disease (PD) neurons [5]. Recent mulation such as WD, manganism or the syndromes of

in vitro studies have shown that mutant ␣-synuclein interacts with neurodegeneration with brain iron accumulation (NBIA) may

metals and that iron (Fe) and copper (Cu) seem to aggravate the manifest with parkinsonism.

toxicity caused by the ␣-synuclein [6,7]. It has been suggested that (3) Animal studies examining exposure to high levels of metals and

-synuclein is a Cu binding protein acting as a cellular ferrireduc- the effects of treatment.

tase [8,9]. Lewy bodies contain reactive iron along with aggregated (4) Measurements of metal concentrations and metal regulatory

proteins and oxidized lipid material [10]. protein expression in post-mortem brain tissue samples.

Our knowledge regarding cellular metal homeostasis has (5) Measurements of metal concentrations and metal regulatory

improved substantially over the last years. Several proteins proteins in the CSF.

involved in cellular import, export, trafficking and storage of Fe, Cu

and manganese (Mn) have been identified [11–13]. Among those, Several of these empirical observations are discussed in the fol-

two novel Mn carriers involved in intracellular Mn transport into lowing sections.

lysosomes and Golgi, ATP13A2 and SLC30A10 respectively, have

been found. Other recent findings include roles of the copper trans- Environmental exposure

porter protein 1 (CTR1) in Cu uptake, of Cu chaperones in synthesis

of specific cuproproteins, and of ceruloplasmin in cellular Fe export Effects of Fe and Cu exposure have been sparsely studied and

[14]. Mechanisms of metal transport across the blood–brain bar- a large environmental survey lent little support for an association

rier (BBB) and the blood–cerebrospinal fluid barrier (BCB) have between environmental exposure to Fe or Cu and PD [35]. How-

been thoroughly investigated [11,15,16]. While the brain seems ever, combined exposure to Cu and Fe [36] or Mn and Fe [37]

to be protected against high plasma Fe concentrations as docu- lasting for two to three decades have been reported to significantly

mented in hereditary hemochromatosis [17], this is apparently not increase the risk of PD. Also, toxic effects of Mn have been exten-

true for Mn and Cu. Thus, increased plasma Cu and Mn concen- sively studied [38,39] and excessive Mn intake to a specific

trations may to brain deposits and CNS damage [18]. It was condition with motor, cognitive and psychiatric symptoms known

recently suggested that Mn enters the CNS predominantly through as manganism, with many similarities to PD. Manganism has been

the BCB and that high Mn concentration impairs the integrity of described largely in workers in mining, welding and smelting

this barrier [19]. There is ongoing research on the roles of Mn industries and in battery factories, but has also been reported in

species and their transporter molecules in brain Mn influx; Mn- communities using drinking water with high Mn concentrations.

citrate was suggested as the species with the highest influx rate Manganism may also arise from intravenous Mn intake, as in long-

[20], and is presumably also the major Mn species in the CSF [21,22]. term parenteral nutrition or in methcathinon (ephedron) abusers

This is in contrast to Fe and Cu which are present in CSF mainly as who use potassium permanganate for the synthesis of the drug [12].

high molecular weight species [18]. Mn and Fe share the trans- Similar clinical symptoms as in manganism have been described

port pathway using the transferrin receptor (TfR) [23,24] and it in end-stage liver diseases and due to mutations in the Mn trans-

has been suggested that Fe deficit may increase brain accumu- porter gene SLC30A10 associated with insufficient biliary excretion

lation of Mn [25,26]. Some authors argue that Fe deficiency may of Mn [40]. Clinical symptoms, neuroimaging findings, pathological

lead to divalent metal transporter 1 (DMT1) upregulation leading examination [41] and lack of response to dopaminergic drugs [42]

to increased Mn uptake [27,28]. There is however controversy to suggest that manganism is not caused by dysfunction of dopami-

what extent the DMT1 is involved in Mn transport [16,29]. Interest- nergic neurons in the SN and that it is pathophysiologically different

ingly, DMT1 is supposedly implicated in Mn uptake by the olfactory from idiopathic PD. It has been suggested that chronic Mn expo-

tract which is another possible route for brain Mn accumulation sure in doses insufficient to induce manganism may be toxic to

[30]. In addition to Fe and Mn interdependencies, interactions the SN rather than the globus pallidus (GP) and can increase the

between Fe and Cu [31] as well as between Fe and calcium [32] risk of developing idiopathic PD [43]. Several epidemiological stud-

metabolism have been reported. Comprehensive discussions of Mn, ies have found an association between Mn exposure and risk of

Fe and Cu transport across BBB and BCB can be found in recent idiopathic PD [36,37,44–49], however other studies do not sup-

reviews [11,15,16,33]. port such an association [35,50,51]. Some reviews [52,53] and one

Despite these advances, it is still unclear to what extent impaired meta-analysis [54] concluded that available data argue against a

metal metabolism affects the pathophysiology of particular neu- relationship between Mn exposure and PD. The latter analysis used

rodegenerative disorders. The aim of this article is to review current strict inclusion criteria leading to exclusion of all studies favor-

literature covering association between metal dyshomeostasis and ing the role of Mn in PD pathophysiology and the topic is still

neurodegeneration, particularly studies measuring concentrations subject to debate. Efforts to establish a firm conclusion are partly

of Fe, Cu and Mn in the cerebrospinal fluid (CSF) or brain tis- hampered by difficulties in differentiating between idiopathic PD

sues of patients with PD and Wilson’s disease (WD) as well as and manganism-related atypical parkinsonism in large scale epi-

studies examining the pathophysiology of metal abnormalities. PD demiological studies [55]. Several factors may increase individual

is a common neurodegenerative disorder with largely unknown susceptibility to PD-related Mn toxicity, e.g. age, ATP13A2 muta-

pathophysiology, while WD is a rare disorder with well described tions, liver dysfunction or iron deficiency [12]. Epidemiological

pathophysiology involving Cu toxicosis where established che- studies of the relationship between Mn exposure and PD should

lating therapy exists. take these factors into account [43].

G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

P. Dusek et al. / Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx 3

Animal models The interval between death and tissue freezing or fixation is also

an important factor influencing the extent of lytic changes that

A selective dopaminergic loss has been produced by intranigral might also alter tissue metal composition. Since procedures used

Fe infusion in healthy mice [56,57], and also by repeated intraperi- to avoid contamination [95,96] are either not reported or not con-

toneal injection of 10 mg iron dextran three times per week during sidered in the majority of studies, it cannot be excluded that errors

four weeks in young Wistar rats [58]. In the 1-methyl-4-phenyl- during sample processing contribute to disparate results [91]. It

1,2,3,6-tetrahydropyridine (MPTP) PD mouse model it was shown should be also noted that routinely used metal analytical methods

that unilateral MPTP treatment can induce 100% increase in Fe con- such as AAS or ICP-MS alone cannot distinguish between different

centration in the ipsilateral SN after 3–6 months [59,60] along with metal species or oxidation states. Also, differences in total con-

decreased ceruloplasmin ferroxidase activity [61]. While exam- centration of a particular metal in a tissue may reflect differences

ining the timeline of consequences of MPTP treatment, it was in the content of metallo-proteins and enzymes (e.g. ceruloplas-

observed that Fe deposits in SN appeared 4.5 months after treat- min in the case of serum Cu [97] or glutamine synthetase in the

ment. Simultaneously a nigral cell loss of 40% had occurred [62] case of brain Mn [98]). Thus, finding of abnormal total tissue metal

indicating that Fe increase in the SN does not precede but may concentrations does not automatically implicate abnormal metal

follow the onset of neurodegenerative changes. Nigral cell dam- stores or abnormal labile (free) metal concentrations. Hyphen-

age in the 6-hydroxydopamine (6-OHDA) animal model of PD is ated techniques, notably high performance liquid chromatography

also accompanied by increased Fe levels in the SN [63]. (HPLC) and capillary zone electrophoresis (CZE) online with ICP-

Chelator pretreatment reduces degeneration of SN neurons in MS, have been used for studying Mn species in body compartments

MPTP, 6-OHDA and lactacystin-induced PD animal models [63–69]. of healthy subjects [99,100]. These studies documented that most

DMT1 upregulation has been documented in animal PD models. of the Mn-containing compounds found in liver were Mn-enzymes

Mice with dysfunctional DMT1 were partially protected against (for review see Ref. [101]). In order to better understand the role of

PD-inducing toxins, stressing the role of this protein in Fe medi- altered metal content in neurodegenerative diseases, similar spe-

ated degeneration [70]. Ceruloplasmin null mice develop increased ciation studies need to be performed in brain tissue and CSF of

nigral Fe levels accompanied by nigral cell loss, both of which were patients.

prevented by administration of the Fe binding agent deferiprone Using microscopic in situ imaging techniques with resolution at

[61]. the micro- or nanometer scale it was shown that Fe deposits in the

Intrastriatal injection of Mn has been shown to cause loss of SN of PD patients are predominantly associated with neuromelanin

dopaminergic cells [71]. Long-term exposure to Mn in developing in dopaminergic neurons [86,102] and there is evidence that Fe in

rats leads to impaired motor coordination and increased signs of these deposits is not strongly bound and thus potentially redox

oxidative stress in the striatum [72]. Contrary to results in humans, active. Suboptimal concentrations of the ferritin protein have been

Mn intoxication in rodents leads to a 20–75% reduction in the num- reported in the SN of PD patients and also in cases of incidental

ber of SN dopaminergic cells [73,74]. Lewy body disease [78,85,103–105]. Normal ferritin upregulation

Taken together, published data support the notion of Fe and Mn associated with increased Fe content during aging was absent in

toxicity directed toward nigral dopaminergic cells and protective PD patients due to sustained activity of iron regulatory protein 1

effects of Fe chelating agents such as deferiprone in animal models (IRP1) [106]. Ferritin is the main Fe storage compound in glial cells

of PD. and its decreased levels does not necessarily affect Fe metabolism in

neurons [107]. Interestingly, ferritin was shown to be a component

Dysregulation of brain metal homeostasis of neuromelanin granules suggesting its possible iron-regulatory

function also in neurons [108]. Moreover, neuromelanin granules

The majority of autopsy studies have reported 30–100% increase overloaded with Fe [10,81,83] exhibit higher level of redox activity

of Fe content in the SN of PD patients compared to age-matched in PD [109]. Taken together, these studies suggest that disrup-

controls but several studies did not confirm these findings. Possible tion of the ferritin complex along with other disturbances of Fe

reasons include different analytical methods [75] and pathophysi- regulation may eventually overwhelm the storage capacity of the

ological heterogeneity in PD [76]. One study indicated that the Fe neuromelanin system, which may lead to an increased labile Fe

content in the SN was not elevated in mildly affected but only in pool promoting the neurodegenerative process [110]. Increased

3+ 2+

advanced PD patients [77], suggesting that Fe increase may not Fe /Fe ratio in PD was also noted in two studies from the same

be involved in the early disease development. The majority of group, using spectrophotometric measurements on samples from

studies using inductively coupled plasma mass spectrometry (ICP- fresh frozen brains pretreated with hydrochloric acid and pepsin

MS) or atomic absorption spectroscopy (AAS) showed increased where ascorbic acid was used to reduce Fe. They concluded that an

3+ 2+

Fe concentration in PD [78–88] (Table 1). It has been argued that increased Fe /Fe ratio is a result of ongoing oxidative processes

negative results yielded by Mössbauer spectroscopy [89,90] are a [77,87]. This interpretation is speculative since conditions known

consequence of its relatively poor sensitivity compared to other to increase oxidative stress such as ischemic insult or Mn overload

2+ 3+

analytical techniques [91]. Coulometry performed using Ferrochem cause the opposite effect, that is an increased Fe /Fe ratio [111].

II, which was employed by Loeffler et al. [92], is known for inter- Moreover, this finding was disputed by other groups who did not

2+

ference with various substances [93] and it cannot be excluded find any detectable Fe iron in SN using Mössbauer spectroscopy

that interference with some compound in PD SN contributed to [89] and micro-XANES technique [112]. The abnormally high con-

2+

the negative results. centration of Fe might have been an artifact of the measurement

Measurement of metal concentrations in tissues is prone to technique used by Sofic et al. [91,113]. This is in line with the his-

3+ 2+

various biases such as instability of metal species during sam- tochemical distribution of Fe and Fe [114] and the finding that

ple preparation and analysis, sampling contamination, and metal the major Fe storage molecules in SN, ferritin and neuromelanin,

3+

leaching during tissue storage. In post-mortem studies tissue metal store Fe in the ferric (Fe ) form [115].

composition may differ from the living condition. Agonal changes Iron accumulation in PD is confined to the SN, while its lev-

including ischemia, BBB disruption and cessation of cell respira- els in the striatum and cerebral cortex are comparable to those of

tion might alter not only the redox state and labile metal pool controls or even decreased [116]. Results in the GP are inconclu-

proportion but also total tissue metal concentration as has been sive since both normal [77,87], increased [82,92] and decreased

documented for Fe during transient brain ischemia in rats [94]. [78,117] concentrations have all been reported. Also CSF studies

G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

4 P. Dusek et al. / Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx

Table 1

Concentration of metals in post-mortem basal ganglia and cortical tissue in PD patients.

Reference Method N Main findings (Fe, Cu, Mn content) Comments

Ayton [61] AAS 10 ↑Fe in SN +42% Ferroxidase activity in SN

↓Cu in SN −51%

=Fe, Cu in frontal GM

Davies [145] SRXFMPIXE 5 ↓Cu in SN (neuromelanin) -50%, LC =Zn in SN, LC, occipital GM

(neuromelanin) −57% ↓Ctr1 in SN neurons

↑Fe in SN (neuromelanin) +37% ↑SOD1 in SN

=Fe, Cu in occipital GM

* ↑

Visanji [105] AAS 3 ↑Fe in SN +100% Ferroportin expression in SN

=Fe in Put, pons ↑Ferritin in Put

↓TfR in Put

Szczerbowska- SRXFM 4 ↑Fe in SN (neurons) +60% Ca, Zn in SN (neurons)

Boruchowska (2012, =Cu in SN (neurons) ↑Rb, Zn in SN (non-neuronal tissue)

2004) [208], [209] =Fe, Cu in SN (non-neuronal tissue)

Barapatre (2010) [210] PIXE 6 =Fe in SN (neuromelanin/non-neuronal tissue) ↓Ca in SN (neuromelanin/non-neuronal tissue)

Wypijewska (2010) MS AAS 17 =Fe, Cu in SN ↑labile Fe in SN

[90] =Labile Cu in SN

3+

Only Fe detected in SN

Popescu [117] XFM 1 ↑Fe in SN +40% Zn in SN, Gp, Put, CN, GM

↓Fe in GP, Put, CN, cGM

↓Cu in SN

Oakley [86] WDXMA 16 ↑Fe in SN (neurons) +100%

↑Fe in SN (neuropil) +70%

* ↑

Reinert (2007) [211] PIXE 1 =Fe in SN (neuromelanin) Ca in SN (neuromelanin)

*

↓Cu in SN (neuromelanin) −190% ↓Ni in SN (neuromelanin)

Morawski (2005) [212] PIXE 6 ↑Fe in SN (neuromelanin) +100%

=Fe (non-significant increase) in SN

(cytoplasm, glia)

Griffiths [82,88] AAS EXAFS 6 Fe in SN +100%, GP +43% Ferritin loading in SN

= Fe in Put, CN, cGM

3+

Galazka-Friedman [89] MS 6 =Fe in SN Only Fe detected in SN

* 3+

Kienzl (1995) [213] EDXMA 3 ↑Fe in SN (neuromelanin) Only Fe detected in SN

*

Jellinger [84] =Fe in SN (Lewy bodies, cytoplasm)

Loeffler [92] COL 14 =Fe in SN, Put, CN, frontal GM =Tf in SN, GP, Put, CN, frontal GM

*

↑Fe in GP

Mann [85] ICP-MS 18 ↑Fe in SN +56% =Zn in SN

=ferritin in SN

*

Good [81] LAMMA 3 ↑Fe in SN (neuromelanin) +45% ↑Al in SN (neuromelanin)

*

=Fe in SN (cytoplasm/neuropil)

Dexter (1989) ICP-MS 27 ↑Fe in SN +35% Zn in SN, Put, CN

[214,78,79] ↓Fe in GP −29% =Pb in SN, cGM

↓Cu in SN −34%

=Mn in SN, Put, CN, cGM

=Cu in GP, Put, CN, cGM

=Fe in Put, CN, cGM

Hirsch [83] WDXMA 5 ↑Fe in SN (non-neuromelanin) +240% =Zn in SN

↑Fe in SN (Lewy bodies) +400% ↑Al in SN (Lewy bodies)

↓Fe in SN (neuromelanin) -35%

Riederer [77] AAS 13 ↑Fe in SN (only in severe degeneration) +35% ↑Zn in RN

↑Cu in RN =Ca, Mg, Zn in SN, GP, Put, CN, RN, cGM

=Fe in GP, Put, CN, RN, cGM ↑Ferritin in SN

3+ 2+

=Cu in SN, GP, Put, CN, cGM ↑Fe /Fe ratio

Uitti [146] AAS, AES 9 ↓Cu in SN Mg in CN

=Fe in SN, CN = Ag, Al, As, B, Be, Ca, Cd, Co, Cr, K, Pb, Mn, Mo,

=Mn in SN, CN Na, Ni, P, Se, Ti, V, W, Zn in SN, CD

↑ 3+ 2+

Sofic [87] SPF 8 ↑Fe in SN +76% Fe /Fe ratio

=Fe in Put, GP, cGM

Earle [80] XFS 11 ↑Fe in SN, BG +100% =Zn in SN, BG

=Mn in SN, BG

AAS – atomic absorption spectroscopy, AES – atomic emission spectroscopy, ICP-AES – inductively coupled plasma-atomic emission spectrometry, ICP-MS – inductively

coupled plasma-mass spectrometry, SPF -spectrophotometry, MS – Mössbauer spectroscopy, (SR)XFM – (synchrotron radiation) X-ray fluorescence microscopy, PIXE –

particle induced X-ray emission, WDXMA – wavelength dispersive X-ray microanalysis, XFS – X-ray fluorescence spectroscopy, LAMMA – Laser Microprobe Mass Analysis,

EXAFS – Extended X-ray absorption fine structure, EDXMA – energy dispersive X-ray microanalysis, SN – substantia nigra, LC – locus coeruleus, Put – putamen, CN – caudate

nucleus, RN – red nucleus, BG – basal ganglia, (c)GM – (cortical) gray matter, Tf(R) – transferrin (receptor).

N = number of patients.

*

Not statistically significant or statistical analysis not performed.

have reported both normal [118–121] and decreased [122–124] Fe The cause of metal regulatory protein dysfunction in PD SN is

concentration in PD suggesting no overall Fe increase in the CNS unknown. Several genes involved in the causation of PD are related

(Table 2). Here it is important to note that speciation analysis was to the metabolism of metals [126] and mutations in DMT1, cerulo-

not performed in these CSF studies and neither abnormal cellular Fe plasmin and transferrin were found to be weakly associated with

2+ 3+

handling, Fe /Fe dysbalance nor increased proportion of labile the risk of developing PD [127–129]. Several studies have docu-

Fe can be excluded. Alteration in Fe balance can be suspected since mented abnormalities in ceruloplasmin function in PD. Specifically,

decreased CSF ferroxidase activity (see below) has been found in decreased ferroxidase activity was found in plasma [130,131],

PD [125]. CSF [125,132] and post-mortem SN tissue [61]. Dysfunction of

G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

P. Dusek et al. / Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx 5

Table 2

Concentration of metals in the CSF of PD patients.

Reference Method N Main findings Comments

Hozumi [119] ICP-MS 20 ↑Cu +80%, Mn +70% ↑Zn +174%

=Fe =Mg

Boll [132] AAS 22 ↑Cu +230% (free) Free Cu correlated with motor impairment

Alimonti [122] ICP-AESSF-ICP-MS 42 ↓Fe −20% Co, Cr, Pb, Si, Sn

=Cu, Mn =Al, Ba, Be, Bi, Ca, Cd, Hg, Li, Mg, Mo, Ni, Sr, Tl, V, W, Zn, Zr

Bocca [123] ICP-AESSF-ICP-MS 91 ↓Fe −37%, =Cu, Mn ↓Si

* ↓

Forte [124] ICP-AESSF-ICP-MS 26 ↓Fe −55% Si

=Cu, Mn =Al, Ca, Mg, Zn

Boll [125] AAS 49 =Cu (total) Ferroxidase activity/total Cu inversely related to PD stage

Jimenez-Jimenez [121] AAS 37 =Fe, Mn, Cu ↓Zn

Gazzaniga [118] AAS 11 =Fe, Mn, Cu

Pall [120] AAS 31 =Fe, Mn ↑Cu (free) confirmed by

↑Cu phenantroline-copper assay

AAS – atomic absorption spectroscopy, AES – atomic emission spectroscopy, ICP-AES – inductively coupled plasma-atomic emission spectrometry, ICP-MS – inductively

coupled plasma-mass spectrometry.

N = number of patients.

*

Not statistically significant or statistical analysis not performed.

ceruloplasmin may be a consequence of oxidative changes in dysfunction of the blood–CSF barrier similar to the situation in

this enzyme [133]. Decreased ferroxidase activity was found WD [147].

to be associated with earlier disease onset [134,135] and with Abnormal concentrations of Mn have not been reported in

markers of iron deposition in the SN of PD patients [129,136,137]. PD brains [78–80,146], but all published studies are rather old.

Other studies have reported abnormal expression of Fe regulatory The majority of studies also found normal Mn levels in the CSF

proteins. Nigral cells behave as if they were Fe deficient in PD; [118,120–124].

expression of proteins involved in iron uptake as DMT1, TfR2

and lactotransferrin receptor (LTFR) is upregulated [138,139],

Wilson’s disease (WD)

whereas expression of proteins involved in iron efflux such as

FPN1 is downregulated [140]. SN areas with the most severe

WD is a genetic disorder caused by mutation of the ATP7B

cell degeneration showed an increased LTFR expression [139]. At

protein which is mostly expressed in the liver [148]. Dysfunc-

the cellular level, regulation of Fe metabolism is dependent on

tion of ATP7B leads to disturbed Cu metabolism with a gradual

mitochondria where Fe is incorporated into iron-sulfur (Fe S)

accumulation of Cu in the body and reduced synthesis of various

clusters by the action of a complex of multiple enzymes. Fe S

cuproproteins, in particular ceruloplasmin. The traditional view on

clusters are prosthetic groups necessary for proper function of

the pathophysiology of neurologic symptoms in WD holds that

several proteins such as Complexes I–III of oxidative phosphory-

overwhelming of the liver storage capacity for Cu leads to its

lation, ubiquinone and aconitase. The latter is also known as iron

spillage into plasma and CSF, leading to cellular uptake and oxida-

responsive element binding protein 1 (IREB1) since it is involved

tive damage in the CNS [149], although alternative theories have

in the regulation of the transcription of Fe transport proteins.

been proposed [150]. However, ATP7B is expressed also in the brain,

Low level of Fe S cluster production leads to low cytoplasmic

namely in the choroid plexus, basal ganglia, cerebellum and cor-

aconitase activity and increased transcription of TfR and DMT1

tex [145] and its dysfunction may lead to neurological symptoms

[141]. Upregulation of Fe uptake may be related to mitochondrial

through other mechanisms than just overflow of hepatic Cu. ATP7B

dysfunction associated with decreased Fe S cluster synthesis,

dysfunction in astrocytes may hypothetically lead to impaired

which was documented in nigral cells in PD [142]. Alternatively, it

production of ceruloplasmin with the possible consequence of

has been show that iron regulatory protein 2 (IRP2) is degraded by

impaired Fe efflux, similar to the situation in aceruloplasminemia

the ubiquitin–proteasome system and its dysfunction may lead to

[151]. It has been shown that ATP7B is expressed in the choroid

high IRP2 levels which in turn stimulate production of DMT1 [143].

plexus and its dysfunction may hamper the elimination of excess

Concerning Cu, greater variation in Cu metabolic profiles has

65 Cu across the blood–CSF barrier [152].

been shown in PD compared to healthy controls using Cu

The combined evidence of animal, neuroimaging, CSF and post-

isotope tracer. Most importantly, higher peak plasma Cu val-

mortem metal studies (see below) suggests that metals other than

ues were documented suggesting more efficient absorption in

Cu may accumulate in WD.

the gut [144]. Increased concentration of redox available Cu has

been reported in PD CSF [120] and its concentration was corre-

lated with motor impairment [132]. The SN contains high levels Local dysregulation of metal homeostasis

of Cu in healthy subjects [145]. The decrease in total Cu con-

centrations reported in the SN in the majority of studies in In patients with the neuropsychiatric form of WD, measure-

PD patients [61,78,102,146] may be a reflection of loss of the ments of Cu in CSF and post-mortem brain tissue clearly show that

cuproproteins 1 (SOD1) or ceruloplasmin. Cu concentrations are diffusely increased in the CNS up to 850% of

In post-mortem PD SN tissue normal SOD1 activity was found normal levels [153] (Table 3). With chelating treatment it takes

[102]. Moreover, decreased Cu levels are not accompanied by sim- up to 4 years for CSF Cu concentrations to normalize [154,155]

ilar decreases in (Zn) levels in the majority of post-mortem and high levels are a marker of non-compliance [156]. Profound

studies (Table 1) which would be expected in the case of SOD1 Cu accumulation is still persistent in all brain regions after 10

loss. Currently, it seems more likely that decreased SN Cu lev- months of chelating treatment as shown by post-mortem studies

els is a consequence of reduced ceruloplasmin content which [157–159]. Another post-portem study showed abnormal brain Cu

is in line with the low ceruloplasmin ferroxidase activity found deposits even after several years of chelating treatment, and also

in SN [61]. Increased labile Cu in the CSF of PD patients may that the severity of neuropathological findings was correlated with

be caused by its impaired incorporation into cuproproteins or cerebral copper content [160].

G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

6 P. Dusek et al. / Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx

Table 3

Concentration of metals in the CFS and brain tissue in WD.

Reference Method N Type of tissue Main findings Comments

Kodama [153] AAS 4 (2 neu, 2 hep) CSF ↑Cu in neuro WD +280% =Zn

=Cu in hepato WD ↓Cp concentration

Cu decreases with therapy

Stuerenburg [155] AAS 4 (neu) CSF ↑Cu WN Cu decreases with therapy

Weisner [154] AAS 5 (neu) CSF ↑Cu +330% ↓Cp concentration

Cu decreases with therapy

*

Cumings (1948) [158] COL 3 Brain ↑Cu in CN(+160%), Put(+600%), thal(+380%), GP(+60%),

WM(290%), GM(+320%)

*

↑Fe in CN(+100%), Put(+20%), thal(+160%), WM(+38%),

GM(+140%)

=Fe in GP

** ↓

Faa [159] ICP-AES 1 Brain ↑Cu in all brain regions Mg, Zn, Ca in all brain

**

↓Fe in all brain regions regions

Litwin [157] ICP-MS 12 (10 neu, 2 hep) Brain Cu in Put(+540%), DN(+590%), pons(+540%), frontal =Zn in Put, DN, pons, frontal

GM(+850%) GM

↑Fe in ND(+74%)

=Fe in Put, pons, frontal GM

=Mn in Put, DN, pons, frontal GM

AAS – atomic absorption spectroscopy, COL – colorimetry, ICP-AES – inductively coupled plasma-atomic emission spectrometry, ICP-MS – inductively coupled plasma-mass

spectrometry.

SN – substantia nigra, DN – dentate nucleus, Put – putamen, CN – caudate nucleus, GM – gray matter, WM – white matter.

Neu – neuropsychiatric form, hep – hepatic form, N = number of patients.

*

No statistical analysis performed.

**

No control group, compared to published reference values.

Increased Fe content has been reported in the striatum, gray because it decreases the availability of Cu for ceruloplasmin

matter, white matter [158] and dentate nucleus of the cerebellum production. Zinc treatment can have opposite effects [174]. No

[157] in post-mortem studies. The latter study reported normal difference in brain Fe concentration was however found between

brain Mn concentrations, but Mn was not measured in the GP which patients treated with Zn and penicillamine [157]. Ceruloplasmin

is the structure with the greatest predisposition toward accumu- also interacts with Mn and its decreased activity can alter the neu-

lation of Mn and other divalent metals [26]. In fact, MRI studies rotoxicity of Mn [175].

indicate that elements such as Fe and Mn may also accumulate in Altogether, these data suggest that profoundly increased Cu con-

the brain over the course of WD. T1 hyperintensities in GP are sug- centrations in the CNS of WD patients persist for a long time during

gestive of Mn deposits in patients with the hepatic form of WD chelating treatment and that local accumulation of Fe in certain

[161,162]. Several MRI studies documented T2 hypointensities in brain nuclei may occur during the course of WD.

GP and other brains regions in WD patients suggestive of deposits

with paramagnetic properties [163–167]. It has been suggested that Animal models

2+

the GP changes might reflect Cu deposits [168], since Cu species

are paramagnetic. However, little is known of which form Cu is Established animal models for WD are the Long Evans Cinnamon

deposited in the WD CNS, and another possibility is that these T2 (LEC) rat [176], the toxic milk mouse [177] and the Atp7b(−/−)

hypointensities correspond to the paramagnetic effect of ferritin- mouse [178]. In LEC rats markedly increased concentrations of

iron. It has been documented that the area of T2 hypointensities loosely bound Cu were found in liver and plasma when total Cu con-

increases during chelating treatment but it seems unlikely that centration exceeded the capacity of liver metallothionein synthesis

this is due to increased brain Cu concentrations [165]. Indeed, a [179–181]. Increased brain Fe levels have been demonstrated in

PET study with radioactively labeled Fe in human subjects showed LEC rats [182] and low Fe diet as well as Fe removal by phlebotomy

increased brain Fe uptake in WD patients compared to healthy was beneficial in terms of decreased oxidative stress markers in

volunteers [169]. the liver [183,184]. Treatment with Zn not only reduced the liver

Iron accumulation in WD may be a consequence of low cerulo- damage through induction of metallothionein expression but also

plasmin levels. However, it has been reported that as little as 10% of led to decreased liver Cu and Fe concentrations in LEC rats [185].

the normal plasma level of ceruloplasmin was sufficient for normal In the toxic milk mouse, increased Cu stores were found in all tis-

Fe metabolism in a pig model [170] and in liver preparations [171]. sues, but elevated Fe was present only in the kidneys, possibly as a

This finding was confirmed in WD patients in whom defective Fe result of hemolysis [186]. The brain Fe content was even decreased

metabolism was observed only in those who had ceruloplasmin in the hippocampus [187]. Penicillamine treatment in these mice

levels less than 5% of the normal range [172]. The fact that most increased labile Cu in plasma and brain tissue as a result of its

WD patients have more than 10% of the normal level of cerulo- mobilization from the large tissue deposits and enhanced oxida-

plasmin also argues against gross changes in Fe metabolism in this tive stress, which may explain the initial worsening seen in human

disease. Although some authors reported elevated liver Fe concen- patients [188].

tration in WD patients [151], results of the largest published study In the Atp7b(−/−) mouse imaging of Cu metabolism using

64

argue against a clinically relevant elevation of Fe in the liver in the ingested Cu showed Cu accumulation in liver but not in brain

majority of patients [173]. This, however, pertains to peripheral Fe [189]. In the same WD model decreased ferroxidase activity of

metabolism and CNS may be more susceptible to oxidative damage ceruloplasmin was related to decreased levels of serum Fe and

caused by decreased ceruloplasmin function. Another factor con- transferrin saturation [190]. In contrast, Cu intoxication itself does

tributing to the risk of Fe accumulation may be mode of treatment. not cause Fe accumulation in liver or brain of rats [191]. This find-

Standard penicillamine chelation does not remove Fe from the ing suggests indirectly that decreased ceruloplasmin, rather than

organism; conversely it may lead to higher tissue Fe accumulation Cu toxicity, affects impaired Fe homeostasis in WD.

G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

P. Dusek et al. / Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx 7

Concluding remarks larger groups of WD patients and studies validating the use of brain

MRI in determining the regional distribution of specific metals.

Current data from studies measuring metal concentrations in

brain tissue and CSF do not support the idea of a general metal Conflict of interest

accumulation in the entire CNS of PD patients. Local dysregulation

The authors have no conflict of interest.

of Fe metabolism seems to occur in the SN, at least in a sub-

group of PD patients. The pathophysiological relation of iron to

Acknowledgments

neurodegeneration and why it selectively affects the SN remains

unanswered. It has been suggested that dopamine producing cells

This work was supported by the Czech Ministry of Education,

are particularly vulnerable to metallotoxic effects [192]. One of the

research project PRVOUKP26/LF1/4 and by the Czech-Norwegian

vulnerable enzymes in the dopamine synthesis pathway may be

Research Programme CZ09.

tyrosine hydroxylase, which requires appropriate amounts of iron

and molecular oxygen as cofactors. Animal data as well as follow-

References

up of patients with manganism gives some support to the idea of

exposure to Mn or Cu as aggravating factors in PD [193,194].

[1] Halliwell B, Gutteridge JM. Oxygen toxicity, oxygen radicals, transition metals

Abundant evidence suggests that decreased activity of cerulo-

and disease. Biochem J 1984;219(1):1–14.

plasmin in CSF and SN is contributing to Fe accumulation in PD and [2] Morello M, Zatta P, Zambenedetti P, Martorana A, D‘Angelo V, Melchiorri

G, et al. Manganese intoxication decreases the expression of manganopro-

may be relevant to its pathophysiology. It should not be forgot-

teins in the rat basal ganglia: an immunohistochemical study. Brain Res Bull

ten that in contrast to Fe deposition, neurodegenerative changes

2007;74(6):406–15.

in PD are not restricted to dopaminergic neurons in the SN [195]. [3] Lill R, Hoffmann B, Molik S, Pierik AJ, Rietzschel N, Stehling O, et al. The role of

This argues against the hypothesis that Fe accumulation alone is mitochondria in cellular iron-sulfur protein biogenesis and iron metabolism.

Biochim Biophys Acta 2012;1823(9):1491–508.

the trigger of neurodegeneration in PD. Nevertheless, based on

[4] Zhang W, Phillips K, Wielgus AR, Liu J, Albertini A, Zucca FA, et al. Neu-

the likely presence of labile Fe in the SN, labile Cu in the CSF and

romelanin activates microglia and induces degeneration of dopaminergic

results from animal studies, we suggest along with other authors neurons: implications for progression of Parkinson’s disease. Neurotox Res

2011;19(1):63–72.

[196] that a therapeutic trial with metal chelating agents is jus-

[5] Uversky VN, Li J, Fink AL. Metal-triggered structural transformations, aggre-

tified in PD. Several points, however, need to be considered. The

gation, and fibrillation of human alpha-synuclein. A possible molecular

dose of the chelating drug should be rather low, aiming at targeted NK between Parkinson’s disease and heavy metal exposure. J Biol Chem

2001;276(47):44284–96.

metal detoxification and redistribution of iron to tissues capable

[6] Chew KC, Ang ET, Tai YK, Tsang F, Lo SQ, Ong E, et al. Enhanced autophagy from

of reutilizing it [197]. Due to the risk of increased concentrations

chronic toxicity of iron and mutant A53T alpha-synuclein: implications for

of labile Cu, a drug with Cu chelating capabilities may be pre- neuronal cell death in Parkinson disease. J Biol Chem 2011;286(38):33380–9.

[7] Wang X, Moualla D, Wright JA, Brown DR. Copper binding regulates intra-

ferred in future studies. The Fe chelator deferiprone is currently

cellular alpha-synuclein localisation, aggregation and toxicity. J Neurochem

being tested in early stage PD (Clinicaltrials.gov NCT01539837 and

2010;113(3):704–14.

NCT00943748). Results from the latter study document that a mod- [8] Davies P, Moualla D, Brown DR. Alpha-synuclein is a cellular ferrireductase.

PLoS ONE 2011;6(1):e15814.

erate dose of deferiprone (30 mg/kg/day) given for 12 months is

[9] Brown DR. alpha-Synuclein as a ferrireductase. Biochem Soc Trans

safe and efficient in Fe removal from the SN. Moreover, using the

2013;41(6):1513–7.

delayed-start paradigm this study indicated a potential disease- [10] Castellani RJ, Siedlak SL, Perry G, Smith MA. Sequestration of iron by Lewy

modifying effect of deferiprone in PD [69]. Since oxidative stress bodies in Parkinson’s disease. Acta Neuropathol 2000;100(2):111–4.

[11] Zheng W, Monnot AD. Regulation of brain iron and copper homeostasis by

and depletion apparently accompany the progression

brain barrier systems: implication in neurodegenerative diseases. Pharmacol

of PD treatments with have been examined. Although

Ther 2012;133(2):177–88.

controversial results of such treatments have been reported, e.g. [12] Tuschl K, Mills PB, Clayton PT. Manganese and the brain. Int Rev Neurobiol

2013;110:277–312.

using vitamin E or vitamin C [198–200], the glutathione preser-

[13] Madsen E, Gitlin JD. Copper and iron disorders of the brain. Annu Rev Neurosci

ving agent lipoic acid appears to possess a therapeutic potential 2007;30:317–37.

according to an animal study [201]. [14] Kristinsson J, Snaedal J, Torsdottir G, Johannesson T. Ceruloplasmin and iron

in Alzheimer’s disease and Parkinson’s disease: a synopsis of recent studies.

Regarding WD, little is known about accumulation of other

Neuropsychiatr Dis Treat 2012;8:515–21.

metals beside Cu, but dysfunction of ceruloplasmin may lead to Fe

[15] Zheng W, Aschner M, Ghersi-Egea JF. Brain barrier systems: a new

accumulation while liver dysfunction may lead to Mn accumula- frontier in metal neurotoxicological research. Toxicol Appl Pharmacol

2003;192(1):1–11.

tion in the CNS. Both metals may contribute to neurodegeneration,

[16] Yokel RA. Manganese flux across the blood–brain barrier. Neuromol Med

especially while acting synergistically with Cu. The rationale of

2009;11(4):297–310.

studying synergistic metal toxicity is further strengthened by [17] Russo N, Edwards M, Andrews T, O‘Brien M, Bhatia KP. Hereditary haemochro-

matosis is unlikely to cause movement disorders – a critical review. J Neurol

recent studies of multiple metals with neurotoxic properties in the

2004;251(7):849–52.

CSF in other neurodegenerative disorders [202,203]. More stud-

[18] Nischwitz V, Berthele A, Michalke B. Speciation analysis of selected metals

ies of toxic interactions among metals are needed, because such and determination of their total contents in paired serum and cerebrospinal

studies may produce invaluable information for the optimal choice fluid samples: an approach to investigate the permeability of the human

blood–cerebrospinal fluid-barrier. Anal Chim Acta 2008;627(2):258–69.

of therapeutic metal chelating agents. Disodium calcium edetate

[19] Bornhorst J, Wehe CA, Huwel S, Karst U, Galla HJ, Schwerdtle T. Impact of

(CaNa –EDTA) has been successfully used to decrease tissue man-

2 manganese on and transfer across blood–brain and blood–cerebrospinal fluid

ganese levels and improve neurological symptoms in manganism barrier in vitro. J Biol Chem 2012;287(21):17140–51.

[20] Yokel RA, Crossgrove JS. Manganese toxicokinetics at the blood–brain barrier.

[204,205]. EDTA chelation has often been administered together

Res Rep Health Eff Inst 2004;119:7–58 [discussion 59–73].

with high doses of vitamin C in order to decrease oxidative stress

[21] Michalke B, Berthele A, Mistriotis P, Ochsenkuhn-Petropoulou M, Halbach S.

in several disorders. This approach, however, does not seem to Manganese species from human serum, cerebrospinal fluid analyzed by size

exclusion chromatography-, capillary electrophoresis coupled to inductively

be substantiated by recent clinical observations, and paradoxically

coupled plasma mass spectrometry. J Trace Elem Med Biol 2007;21(Suppl.

vitamin C may enhance oxidative stress [206]. Unlike penicillamine, 1):4–9.

trientine not only chelates Cu but also Fe and Mn, although the sta- [22] Michalke B, Lucio M, Berthele A, Kanawati B. Manganese speciation in paired

serum and CSF samples using SEC-DRC-ICP-MS and CE-ICP-DRC-MS. Anal

bility constants of these complexes are lower than that with Cu

Bioanal Chem 2013;405(7):2301–9.

[207]. Thus, trientine may be useful in patients with signs of Fe and

[23] Aschner M, Gannon M. Manganese (Mn) transport across the rat blood–brain

Mn accumulation in combination with copper. For this reason, there barrier: saturable and transferrin-dependent transport mechanisms. Brain

Res Bull 1994;33(3):345–9.

is a need for further studies examining free metal concentrations in

G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

8 P. Dusek et al. / Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx

[24] Fitsanakis VA, Zhang N, Garcia S, Aschner M. Manganese (Mn) and [53] Jankovic J. Searching for a relationship between manganese and welding and

iron (Fe): interdependency of transport and regulation. Neurotox Res Parkinson’s disease. Neurology 2005;64(12):2021–8.

2010;18(2):124–31. [54] Mortimer JA, Borenstein AR, Nelson LM. Associations of welding and

[25] Mena I, Horiuchi K, Burke K, Cotzias GC. Chronic manganese poisoning. Indi- manganese exposure with Parkinson disease: review and meta-analysis. Neu-

vidual susceptibility and absorption of iron. Neurology 1969;19(10):1000–6. rology 2012;79(11):1174–80.

[26] Erikson KM, Syversen T, Steinnes E, Aschner M. Globus pallidus: a target [55] Park RM. Neurobehavioral deficits and parkinsonism in occupations with

brain region for divalent metal accumulation associated with dietary iron manganese exposure: a review of methodological issues in the epidemio-

deficiency. J Nutr Biochem 2004;15(6):335–41. logical literature. Saf Health Work 2013;4(3):123–35.

[27] Erikson KM, Syversen T, Aschner JL, Aschner M. Interactions between exces- [56] Wesemann W, Blaschke S, Solbach M, Grote C, Clement HW, Riederer P.

sive manganese exposures and dietary iron-deficiency in neurodegeneration. Intranigral injected iron progressively reduces striatal dopamine metabolism.

Environ Toxicol Pharmacol 2005;19(3):415–21. J Neural Transm Park Dis Dement Sect 1994;8(3):209–14.

[28] Wang X, Li GJ, Zheng W. Upregulation of DMT1 expression in choroidal epithe- [57] Sengstock GJ, Olanow CW, Menzies RA, Dunn AJ, Arendash GW. Infusion of

lia of the blood–CSF barrier following manganese exposure in vitro. Brain Res iron into the rat substantia nigra: nigral pathology and dose-dependent loss

2006;1097(1):1–10. of striatal dopaminergic markers. J Neurosci Res 1993;35(1):67–82.

[29] Crossgrove JS, Yokel RA. Manganese distribution across the blood–brain [58] Jiang H, Song N, Wang J, Ren LY, Xie JX. Peripheral iron dextran induced

barrier III. The divalent metal transporter-1 is not the major mechanism degeneration of dopaminergic neurons in rat substantia nigra. Neurochem

mediating brain manganese uptake. Neurotoxicology 2004;25(3):451–60. Int 2007;51(1):32–6.

[30] Thompson K, Molina RM, Donaghey T, Schwob JE, Brain JD, Wessling-Resnick [59] Mochizuki H, Imai H, Endo K, Yokomizo K, Murata Y, Hattori N, et al.

M. Olfactory uptake of manganese requires DMT1 and is enhanced by anemia. Iron accumulation in the substantia nigra of 1-methyl-4-phenyl-1,2,3,6-

FASEB J 2007;21(1):223–30. tetrahydropyridine (MPTP)-induced hemiparkinsonian monkeys. Neurosci

[31] Monnot AD, Zheng G, Zheng W. Mechanism of copper transport at the Lett 1994;168(1–2):251–3.

blood–cerebrospinal fluid barrier: influence of iron deficiency in an in vitro [60] Temlett JA, Landsberg JP, Watt F, Grime GW. Increased iron in the substan-

model. Exp Biol Med (Maywood) 2012;237(3):327–33. tia nigra compacta of the MPTP-lesioned hemiparkinsonian African green

[32] Gaasch JA, Geldenhuys WJ, Lockman PR, Allen DD, Van der Schyf CJ. Voltage- monkey: evidence from proton microprobe elemental microanalysis. J Neu-

gated calcium channels provide an alternate route for iron uptake in neuronal rochem 1994;62(1):134–46.

cell cultures. Neurochem Res 2007;32(10):1686–93. [61] Ayton S, Lei P, Duce JA, Wong BX, Sedjahtera A, Adlard PA, et al. Ceruloplas-

[33] Michalke B, Fernsebner K. New insights into manganese toxicity and specia- min dysfunction and therapeutic potential for Parkinson disease. Ann Neurol

tion. J Trace Elem Med Biol 2014;28(2):106–16. 2013;73(4):554–9.

[34] Graham DG. Catecholamine toxicity: a proposal for the molecular pathogen- [62] He Y, Thong PS, Lee T, Leong SK, Mao BY, Dong F, et al. Dopaminergic cell

esis of manganese neurotoxicity and Parkinson’s disease. Neurotoxicology death precedes iron elevation in MPTP-injected monkeys. Free Radic Biol Med

1984;5(1):83–95. 2003;35(5):540–7.

[35] Dick FD, De Palma G, Ahmadi A, Scott NW, Prescott GJ, Bennett J, et al. [63] Shachar DB, Kahana N, Kampel V, Warshawsky A, Youdim MB. Neu-

Environmental risk factors for Parkinson’s disease and parkinsonism: the roprotection by a novel brain permeable iron chelator, VK-28, against

Geoparkinson study. Occup Environ Med 2007;64(10):666–72. 6-hydroxydopamine lession in rats. Neuropharmacology 2004;46(2):254–63.

[36] Gorell JM, Johnson CC, Rybicki BA, Peterson EL, Kortsha GX, Brown GG, et al. [64] Berg D, Hochstrasser H. Iron metabolism in Parkinsonian syndromes. Mov

Occupational exposures to metals as risk factors for Parkinson’s disease. Neu- Disord 2006;21(9):1299–310.

rology 1997;48(3):650–8. [65] Ben-Shachar D, Eshel G, Finberg JP, Youdim MB. The iron chelator

[37] Zayed J, Ducic S, Campanella G, Panisset JC, Andre P, Masson H, et al. Envi- desferrioxamine (Desferal) retards 6-hydroxydopamine-induced degen-

ronmental factors in the etiology of Parkinson’s disease. Can J Neurol Sci eration of nigrostriatal dopamine neurons. J Neurochem 1991;56(4):

1990;17(3):286–91. 1441–4.

[38] Racette BA, Aschner M, Guilarte TR, Dydak U, Criswell SR, Zheng W. [66] Youdim MB, Stephenson G, Ben Shachar D. Ironing iron out in Parkinson’s

Pathophysiology of manganese-associated neurotoxicity. Neurotoxicology disease and other neurodegenerative diseases with iron chelators: a lesson

2012;33(4):881–6. from 6-hydroxydopamine and iron chelators, desferal and VK-28. Ann N Y

[39] Aschner M, Erikson KM, Herrero Hernandez E, Tjalkens R. Manganese and its Acad Sci 2004;1012:306–25.

role in Parkinson’s disease: from transport to neuropathology. Neuromol Med [67] Zhu W, Xie W, Pan T, Xu P, Fridkin M, Zheng H, et al. Prevention and restoration

2009;11(4):252–66. of lactacystin-induced nigrostriatal dopamine neuron degeneration by novel

[40] Maeda H, Sato M, Yoshikawa A, Kimura M, Sonomura T, Terada M, et al. brain-permeable iron chelators. FASEB J 2007;21(14):3835–44.

Brain MR imaging in patients with hepatic cirrhosis: relationship between [68] Kaur D, Yantiri F, Rajagopalan S, Kumar J, Mo JQ, Boonplueang R, et al.

high intensity signal in basal ganglia on T1-weighted images and elemental Genetic or pharmacological iron chelation prevents MPTP-induced neuro-

concentrations in brain. Neuroradiology 1997;39(8):546–50. toxicity in vivo: a novel therapy for Parkinson’s disease. Neuron 2003;37(6):

[41] Perl DP, Olanow CW. The neuropathology of manganese-induced Parkinso- 899–909.

nism. J Neuropathol Exp Neurol 2007;66(8):675–82. [69] Devos D, Moreau C, Devedjian JC, Kluza J, Petrault M, Laloux C, et al. Target-

[42] Koller WC, Lyons KE, Truly W. Effect of levodopa treatment for parkinsonism ing chelatable iron as a therapeutic modality in Parkinson’s disease. Antioxid

in welders: a double-blind study. Neurology 2004;62(5):730–3. Redox Signal 2014, http://dx.doi.org/10.1089/ars.2013.5593, ahead of print.

[43] Lucchini RG, Martin CJ, Doney BC. From manganism to manganese-induced

[70] Salazar J, Mena N, Hunot S, Prigent A, Alvarez-Fischer D, Arredondo M,

parkinsonism: a conceptual model based on the evolution of exposure. Neu-

et al. Divalent metal transporter 1 (DMT1) contributes to neurodegener-

romol Med 2009;11(4):311–21.

ation in animal models of Parkinson’s disease. Proc Natl Acad Sci U S A

[44] Finkelstein MM, Jerrett M. A study of the relationships between Parkinson’s 2008;105(47):18578–83.

disease and markers of traffic-derived and environmental manganese air pol-

[71] Brouillet EP, Shinobu L, McGarvey U, Hochberg F, Beal MF. Manganese injec-

lution in two Canadian cities. Environ Res 2007;104(3):420–32.

tion into the rat striatum produces excitotoxic lesions by impairing energy

[45] Willis AW, Evanoff BA, Lian M, Galarza A, Wegrzyn A, Schootman M, et al.

metabolism. Exp Neurol 1993;120(1):89–94.

Metal emissions and urban incident Parkinson disease: a community health

[72] Cordova FM, Aguiar Jr AS, Peres TV, Lopes MW, Goncalves FM, Pedro DZ, et al.

study of Medicare beneficiaries by using geographic information systems. Am

Manganese-exposed developing rats display motor deficits and striatal oxida-

J Epidemiol 2010;172(12):1357–63.

tive stress that are reversed by Trolox. Arch Toxicol 2013;87(7):1231–44.

[46] Gorell JM, Rybicki BA, Cole Johnson C, Peterson EL. Occupational

[73] Stanwood GD, Leitch DB, Savchenko V, Wu J, Fitsanakis VA, Anderson DJ, et al.

metal exposures and the risk of Parkinson’s disease. Neuroepidemiology

Manganese exposure is cytotoxic and alters dopaminergic and GABAergic

1999;18(6):303–8.

neurons within the basal ganglia. J Neurochem 2009;110(1):378–89.

[47] Hudnell HK. Effects from environmental Mn exposures: a review of

[74] Sanchez-Betancourt J, Anaya-Martinez V, Gutierrez-Valdez AL, Ordonez-

the evidence from non-occupational exposure studies. Neurotoxicology

Librado JL, Montiel-Flores E, Espinosa-Villanueva J, et al. Manganese mixture

1999;20(2–3):379–97.

inhalation is a reliable Parkinson disease model in rats. Neurotoxicology

[48] Lucchini RG, Albini E, Benedetti L, Borghesi S, Coccaglio R, Malara EC, et al. 2012;33(5):1346–55.

High prevalence of Parkinsonian disorders associated to manganese expo-

[75] Friedman A, Galazka-Friedman J, Koziorowski D. Iron as a cause of Parkinson

sure in the vicinities of ferroalloy industries. Am J Ind Med 2007;50(11):

disease – a myth or a well established hypothesis? Parkinsonism Relat Disord 788–800.

2009;15(Suppl. 3):S212–4.

[49] Racette BA, McGee-Minnich L, Moerlein SM, Mink JW, Videen TO, Perlmutter

[76] Sian-Hulsmann J, Mandel S, Youdim MB, Riederer P. The relevance of iron in

JS. Welding-related parkinsonism: clinical features, treatment, and patho-

the pathogenesis of Parkinson’s disease. J Neurochem 2011;118(6):939–57.

physiology. Neurology 2001;56(1):8–13.

[77] Riederer P, Sofic E, Rausch WD, Schmidt B, Reynolds GP, Jellinger K, et al. Tran-

[50] Fored CM, Fryzek JP, Brandt L, Nise G, Sjogren B, McLaughlin JK, et al.

sition metals, ferritin, glutathione, and ascorbic acid in parkinsonian brains. J

Parkinson’s disease and other basal ganglia or movement disorders in

Neurochem 1989;52(2):515–20.

a large nationwide cohort of Swedish welders. Occup Environ Med

[78] Dexter DT, Carayon A, Javoy-Agid F, Agid Y, Wells FR, Daniel SE, et al. Alter-

2006;63(2):135–40.

ations in the levels of iron, ferritin and other trace metals in Parkinson’s

[51] Kenborg L, Lassen CF, Hansen J, Olsen JH. Parkinson’s disease and other neu-

disease and other neurodegenerative diseases affecting the basal ganglia.

rodegenerative disorders among welders: a Danish cohort study. Mov Disord

Brain 1991;114(Pt 4):1953–75.

2012;27(10):1283–9.

[79] Dexter DT, Wells FR, Agid F, Agid Y, Lees AJ, Jenner P, et al. Increased

[52] Guilarte TR. Manganese and Parkinson’s disease: a critical review and new

nigral iron content in postmortem parkinsonian brain. Lancet 1987;2(8569):

findings. Environ Health Perspect 2010;118(8):1071–80. 1219–20.

G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

P. Dusek et al. / Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx 9

[80] Earle KM. Studies on Parkinson’s disease including X-ray fluorescent [109] Faucheux BA, Martin ME, Beaumont C, Hauw JJ, Agid Y, Hirsch EC. Neu-

spectroscopy of formalin fixed brain tissue. J Neuropathol Exp Neurol romelanin associated redox-active iron is increased in the substantia nigra

1968;27(1):1–14. of patients with Parkinson’s disease. J Neurochem 2003;86(5):1142–8.

[81] Good PF, Olanow CW, Perl DP. Neuromelanin-containing neurons of the subs- [110] Zecca L, Wilms H, Geick S, Claasen JH, Brandenburg LO, Holzknecht C, et al.

tantia nigra accumulate iron and aluminum in Parkinson’s disease: a LAMMA Human neuromelanin induces neuroinflammation and neurodegeneration

study. Brain Res 1992;593(2):343–6. in the rat substantia nigra: implications for Parkinson’s disease. Acta Neu-

[82] Griffiths PD, Crossman AR. Distribution of iron in the basal ganglia and neo- ropathol 2008;116(1):47–55.

cortex in postmortem tissue in Parkinson’s disease and Alzheimer’s disease. [111] Fernsebner K, Zorn J, Kanawati B, Walker A, Michalke B. Manganese leads to

Dementia 1993;4(2):61–5. an increase in markers of oxidative stress as well as to a shift in the ratio of

[83] Hirsch EC, Brandel JP, Galle P, Javoy-Agid F, Agid Y. Iron and aluminum Fe(II)/(III) in rat brain tissue. Metallomics 2014;6(4):921–31.

increase in the substantia nigra of patients with Parkinson’s disease: an X-ray [112] Chwiej J, Adamek D, Szczerbowska-Boruchowska M, Krygowska-Wajs A,

microanalysis. J Neurochem 1991;56(2):446–51. Wojcik S, Falkenberg G, et al. Investigations of differences in iron oxida-

[84] Jellinger K, Kienzl E, Rumpelmair G, Riederer P, Stachelberger H, Ben-Shachar tion state inside single neurons from substantia nigra of Parkinson’s disease

D, et al. Iron-melanin complex in substantia nigra of parkinsonian brains: an and control patients using the micro-XANES technique. J Biol Inorg Chem

X-ray microanalysis. J Neurochem 1992;59(3):1168–71. 2007;12(2):204–11.

[85] Mann VM, Cooper JM, Daniel SE, Srai K, Jenner P, Marsden CD, et al. Com- [113] Friedman A, Galazka-Friedman J. The history of the research of iron in parkin-

plex I, iron, and ferritin in Parkinson’s disease substantia nigra. Ann Neurol sonian substantia nigra. J Neural Transm 2012;119(12):1507–10.

1994;36(6):876–81. [114] Morris CM, Candy JM, Oakley AE, Bloxham CA, Edwardson JA. Histochem-

[86] Oakley AE, Collingwood JF, Dobson J, Love G, Perrott HR, Edwardson JA, et al. ical distribution of non-haem iron in the human brain. Acta Anat (Basel)

Individual dopaminergic neurons show raised iron levels in Parkinson dis- 1992;144(3):235–57.

ease. Neurology 2007;68(21):1820–5. [115] Zecca L, Youdim MB, Riederer P, Connor JR, Crichton RR. Iron, brain ageing

[87] Sofic E, Riederer P, Heinsen H, Beckmann H, Reynolds GP, Hebenstreit G, et al. and neurodegenerative disorders. Nat Rev Neurosci 2004;5(11):863–73.

Increased iron (III) and total iron content in post mortem substantia nigra of [116] Yu X, Du T, Song N, He Q, Shen Y, Jiang H, et al. Decreased iron levels in

parkinsonian brain. J Neural Transm 1988;74(3):199–205. the temporal cortex in postmortem human brains with Parkinson disease.

[88] Griffiths PD, Dobson BR, Jones GR, Clarke DT. Iron in the basal ganglia Neurology 2013;80(5):492–5.

in Parkinson’s disease. An in vitro study using extended X-ray absorp- [117] Popescu BF, George MJ, Bergmann U, Garachtchenko AV, Kelly ME, McCrea RP,

tion fine structure and cryo-electron microscopy. Brain 1999;122(Pt 4): et al. Mapping metals in Parkinson’s and normal brain using rapid-scanning

667–73. X-ray fluorescence. Phys Med Biol 2009;54(3):651–63.

[89] Galazka-Friedman J, Bauminger ER, Friedman A, Barcikowska M, Hechel D, [118] Gazzaniga GC, Ferraro B, Camerlingo M, Casto L, Viscardi M, Mamoli A. A case

Nowik I. Iron in parkinsonian and control substantia nigra – a Mossbauer control study of CSF copper, iron and manganese in Parkinson disease. Ital J

spectroscopy study. Mov Disord 1996;11(1):8–16. Neurol Sci 1992;13(3):239–43.

[90] Wypijewska A, Galazka-Friedman J, Bauminger ER, Wszolek ZK, Schweitzer [119] Hozumi I, Hasegawa T, Honda A, Ozawa K, Hayashi Y, Hashimoto K, et al.

KJ, Dickson DW, et al. Iron and reactive oxygen species activity in parkinsonian Patterns of levels of biological metals in CSF differ among neurodegenerative

substantia nigra. Parkinsonism Relat Disord 2010;16(5):329–33. diseases. J Neurol Sci 2011;303(1–2):95–9.

[91] Hare DJ, Gerlach M, Riederer P. Considerations for measuring iron in [120] Pall HS, Williams AC, Blake DR, Lunec J, Gutteridge JM, Hall M, et al. Raised

post-mortem tissue of Parkinson’s disease patients. J Neural Transm cerebrospinal-fluid copper concentration in Parkinson’s disease. Lancet

2012;119(12):1515–21. 1987;2(8553):238–41.

[92] Loeffler DA, Connor JR, Juneau PL, Snyder BS, Kanaley L, DeMaggio AJ, et al. [121] Jimenez-Jimenez FJ, Molina JA, Aguilar MV, Meseguer I, Mateos-Vega CJ,

Transferrin and iron in normal, Alzheimer’s disease, and Parkinson’s disease Gonzalez-Munoz MJ, et al. Cerebrospinal fluid levels of transition metals in

brain regions. J Neurochem 1995;65(2):710–6. patients with Parkinson’s disease. J Neural Transm 1998;105(4–5):497–505.

[93] Palmer JH, Sapienza TJ, Zink EW. Pyrazinamide interference in the Ferrochem [122] Alimonti A, Bocca B, Pino A, Ruggieri F, Forte G, Sancesario G. Elemental profile

II determination of iron. Clin Chem 1988;34(7):1510–1. of cerebrospinal fluid in patients with Parkinson’s disease. J Trace Elem Med

[94] Danielisova V, Gottlieb M, Burda J. Iron deposition after transient forebrain Biol 2007;21(4):234–41.

ischemia in rat brain. Neurochem Res 2002;27(3):237–42. [123] Bocca B, Alimonti A, Senofonte O, Pino A, Violante N, Petrucci F, et al. Metal

[95] Versieck J, Barbier F, Cornelis R, Hoste J. Sample contamination as a source of changes in CSF and peripheral compartments of parkinsonian patients. J Neu-

error in trace-element analysis of biological samples. Talanta 1982;29(11 Pt rol Sci 2006;248(1–2):23–30.

2):973–84. [124] Forte G, Bocca B, Senofonte O, Petrucci F, Brusa L, Stanzione P, et al. Trace

[96] Versieck J, Vanballenberghe L, De Kesel A, Van Renterghem D. Accuracy of and major elements in whole blood, serum, cerebrospinal fluid and urine of

biological trace-element determinations. Biol Trace Elem Res 1987;12(1): patients with Parkinson’s disease. J Neural Transm 2004;111(8):1031–40.

45–54. [125] Boll MC, Sotelo J, Otero E, Alcaraz-Zubeldia M, Rios C. Reduced ferroxidase

[97] Twomey PJ, Viljoen A, House IM, Reynolds TM, Wierzbicki AS. Relationship activity in the cerebrospinal fluid from patients with Parkinson’s disease.

between serum copper, ceruloplasmin, and non-ceruloplasmin-bound cop- Neurosci Lett 1999;265(3):155–8.

per in routine clinical practice. Clin Chem 2005;51(8):1558–9. [126] Funke C, Schneider SA, Berg D, Kell DB. Genetics and iron in the systems

[98] Wedler FC, Denman RB. Glutamine synthetase: the major Mn(II) enzyme in biology of Parkinson’s disease and some related disorders. Neurochem Int

mammalian brain. Curr Top Cell Regul 1984;24:153–69. 2013;62(5):637–52.

[99] Michalke B. Manganese speciation using capillary electrophoresis-ICP-mass [127] He Q, Du T, Yu X, Xie A, Song N, Kang Q, et al. DMT1 polymorphism and risk

spectrometry. J Chromatogr A 2004;1050(1):69–76. of Parkinson’s disease. Neurosci Lett 2011;501(3):128–31.

[100] Quintana M, Klouda AD, Gondikas A, Ochsenkuhn-Petropoulou M, Michalke [128] Borie C, Gasparini F, Verpillat P, Bonnet AM, Agid Y, Hetet G, et al. French

B. Analysis of size characterized manganese species from liver extracts Parkinson’s disease genetic study g. Association study between iron-related

using capillary zone electrophoresis coupled to inductively coupled genes polymorphisms and Parkinson’s disease. J Neurol 2002;249(7):801–4.

plasma mass spectrometry (CZE-ICP-MS). Anal Chim Acta 2006;573–574: [129] Hochstrasser H, Bauer P, Walter U, Behnke S, Spiegel J, Csoti I, et al. Cerulo-

172–80. plasmin gene variations and substantia nigra hyperechogenicity in Parkinson

[101] Michalke B, Halbach S, Nischwitz V. Speciation and toxicological relevance of disease. Neurology 2004;63(10):1912–7.

manganese in humans. J Environ Monit 2007;9(7):650–6. [130] Torsdottir G, Sveinbjornsdottir S, Kristinsson J, Snaedal J, Johannesson T.

[102] Davies KM, Bohic S, Carmona A, Ortega R, Cottam V, Hare DJ, et al. Copper Ceruloplasmin and superoxide dismutase (SOD1) in Parkinson’s disease: a

pathology in vulnerable brain regions in Parkinson’s disease. Neurobiol Aging follow-up study. J Neurol Sci 2006;241(1–2):53–8.

2014;35(4):858–66. [131] Torsdottir G, Kristinsson J, Sveinbjornsdottir S, Snaedal J, Johannesson T. Cop-

[103] Friedman A, Arosio P, Finazzi D, Koziorowski D, Galazka-Friedman J. Fer- per, ceruloplasmin, superoxide dismutase and iron parameters in Parkinson’s

ritin as an important player in neurodegeneration. Parkinsonism Relat Disord disease. Pharmacol Toxicol 1999;85(5):239–43.

2011;17(6):423–30. [132] Boll MC, Alcaraz-Zubeldia M, Montes S, Rios C. Free copper, ferroxidase

[104] Dexter DT, Carayon A, Vidailhet M, Ruberg M, Agid F, Agid Y, et al. Decreased and SOD1 activities, lipid peroxidation and NO(x) content in the CSF. A dif-

ferritin levels in brain in Parkinson’s disease. J Neurochem 1990;55(1):16–20. ferent marker profile in four neurodegenerative diseases. Neurochem Res

[105] Visanji NP, Collingwood JF, Finnegan ME, Tandon A, House E, Hazrati LN. Iron 2008;33(9):1717–23.

deficiency in parkinsonism: region-specific iron dysregulation in Parkinson’s [133] Olivieri S, Conti A, Iannaccone S, Cannistraci CV, Campanella A, Barbariga

disease and multiple system atrophy. J Parkinsons Dis 2013;3(4):523–37. M, et al. Ceruloplasmin oxidation, a feature of Parkinson’s disease CSF,

[106] Faucheux BA, Martin ME, Beaumont C, Hunot S, Hauw JJ, Agid Y, et al. Lack of inhibits ferroxidase activity and promotes cellular iron retention. J Neurosci

up-regulation of ferritin is associated with sustained iron regulatory protein- 2011;31(50):18568–77.

1 binding activity in the substantia nigra of patients with Parkinson’s disease. [134] Bharucha KJ, Friedman JK, Vincent AS, Ross ED. Lower serum ceruloplasmin

J Neurochem 2002;83(2):320–30. levels correlate with younger age of onset in Parkinson’s disease. J Neurol

[107] Zecca L, Gallorini M, Schunemann V, Trautwein AX, Gerlach M, Riederer P, et al. 2008;255(12):1957–62.

Iron, neuromelanin and ferritin content in the substantia nigra of normal sub- [135] Jin L, Wang J, Jin H, Fei G, Zhang Y, Chen W, et al. Nigral iron deposi-

jects at different ages: consequences for iron storage and neurodegenerative tion occurs across motor phenotypes of Parkinson’s disease. Eur J Neurol

processes. J Neurochem 2001;76(6):1766–73. 2012;19(7):969–76.

[108] Tribl F, Asan E, Arzberger T, Tatschner T, Langenfeld E, Meyer HE, et al. Identi- [136] Jin L, Wang J, Zhao L, Jin H, Fei G, Zhang Y, et al. Decreased serum ceruloplas-

fication of L-ferritin in neuromelanin granules of the human substantia nigra: min levels characteristically aggravate nigral iron deposition in Parkinson’s

a targeted proteomics approach. Mol Cell Proteomics 2009;8(8):1832–8. disease. Brain 2011;134(Pt 1):50–8.

G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

10 P. Dusek et al. / Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx

[137] Martinez-Hernandez R, Montes S, Higuera-Calleja J, Yescas P, Boll MC, Diaz- [167] Mironov A. Decreased signal intensity of the putamen and the caudate nucleus

Ruiz A, et al. Plasma ceruloplasmin ferroxidase activity correlates with the in Wilson disease of the brain. Neuroradiology 1993;35(2):166.

nigral sonographic area in Parkinson’s disease patients: a pilot study. Neu- [168] Fritzsch D, Reiss-Zimmermann M, Trampel R, Turner R, Hoffmann KT, Schafer

rochem Res 2011;36(11):2111–5. A. Seven-Tesla magnetic resonance imaging in Wilson disease using quantita-

[138] Ke Y, Qian ZM. Brain iron metabolism: neurobiology and neurochemistry. tive susceptibility mapping for measurement of copper accumulation. Invest

Prog Neurobiol 2007;83(3):149–73. Radiol 2014;49(5):299–306.

[139] Faucheux BA, Nillesse N, Damier P, Spik G, Mouatt-Prigent A, Pierce A, et al. [169] Bruehlmeier M, Leenders KL, Vontobel P, Calonder C, Antonini A, Weindl A.

52

Expression of lactoferrin receptors is increased in the mesencephalon of Increased cerebral iron uptake in Wilson’s disease: a Fe-citrate PET study. J

patients with Parkinson disease. Proc Natl Acad Sci U S A 1995;92(21):9603–7. Nucl Med 2000;41(5):781–7.

[140] Wang J, Jiang H, Xie JX. Ferroportin1 and hephaestin are involved in [170] Ragan HA, Nacht S, Lee GR, Bishop CR, Cartwright GE. Effect of ceruloplasmin

the nigral iron accumulation of 6-OHDA-lesioned rats. Eur J Neurosci on plasma iron in copper-deficient swine. Am J Physiol 1969;217(5):1320–3.

2007;25(9):2766–72. [171] Osaki S, Johnson DA, Frieden E. The mobilization of iron from the perfused

[141] Crichton RR, Dexter DT, Ward RJ. Brain iron metabolism and its perturbation mammalian liver by a serum copper enzyme, ferroxidase I. J Biol Chem

in neurological diseases. J Neural Transm 2011;118(3):301–14. 1971;246(9):3018–23.

[142] Gille G, Reichmann H. Iron-dependent functions of mitochondria-relation to [172] Roeser HP, Lee GR, Nacht S, Cartwright GE. The role of ceruloplasmin in iron

neurodegeneration. J Neural Transm 2011;118(3):349–59. metabolism. J Clin Invest 1970;49(12):2408–17.

[143] Li XP, Xie WJ, Zhang Z, Kansara S, Jankovic J, Le WD. A mechanistic study [173] Pfeiffenberger J, Gotthardt DN, Herrmann T, Seessle J, Merle U, Schirmacher

of proteasome inhibition-induced iron misregulation in dopamine neuron P, et al. Iron metabolism and the role of HFE gene polymorphisms in Wilson

degeneration. Neurosignals 2012;20(4):223–36. disease. Liver Int 2012;32(1):165–70.

[144] Larner F, Sampson B, Rehkamper M, Weiss DJ, Dainty JR, O‘Riordan S, et al. High [174] Medici V, Di Leo V, Lamboglia F, Bowlus CL, Tseng SC, D‘Inca R, et al. Effect

precision isotope measurements reveal poor control of copper metabolism in of penicillamine and zinc on iron metabolism in Wilson’s disease. Scand J

parkinsonism. Metallomics 2013;5(2):125–32. Gastroenterol 2007;42(12):1495–500.

[145] Davies KM, Hare DJ, Cottam V, Chen N, Hilgers L, Halliday G, et al. Local- [175] Jursa T, Smith DR. Ceruloplasmin alters the tissue disposition and neu-

ization of copper and copper transporters in the human brain. Metallomics rotoxicity of manganese, but not its loading onto transferrin. Toxicol Sci

2013;5(1):43–51. 2009;107(1):182–93.

[146] Uitti RJ, Rajput AH, Rozdilsky B, Bickis M, Wollin T, Yuen WK. Regional metal [176] Wu J, Forbes JR, Chen HS, Cox DW. The LEC rat has a deletion in the copper

concentrations in Parkinson’s disease, other chronic neurological diseases, transporting ATPase gene homologous to the Wilson disease gene. Nat Genet

and control brains. Can J Neurol Sci 1989;16(3):310–4. 1994;7(4):541–5.

[147] Choi BS, Zheng W. Copper transport to the brain by the blood–brain barrier [177] Theophilos MB, Cox DW, Mercer JF. The toxic milk mouse is a murine model

and blood–CSF barrier. Brain Res 2009;1248:14–21. of Wilson disease. Hum Mol Genet 1996;5(10):1619–24.

[148] Tanzi RE, Petrukhin K, Chernov I, Pellequer JL, Wasco W, Ross B, et al. The [178] Buiakova OI, Xu J, Lutsenko S, Zeitlin S, Das K, Das S, et al. Null mutation

Wilson disease gene is a copper transporting ATPase with homology to the of the murine ATP7B (Wilson disease) gene results in intracellular copper

Menkes disease gene. Nat Genet 1993;5(4):344–50. accumulation and late-onset hepatic nodular transformation. Hum Mol Genet

[149] Telianidis J, Hung YH, Materia S, Fontaine SL. Role of the P-Type ATPases, 1999;8(9):1665–71.

ATP7A and ATP7B in brain copper homeostasis. Front Aging Neurosci [179] Suzuki KT. Disordered copper metabolism in LEC rats, an animal model of

2013;5:44. Wilson disease: roles of metallothionein. Res Commun Mol Pathol Pharmacol

[150] Zischka H, Lichtmannegger J, Schmitt S, Jagemann N, Schulz S, Wartini D, et al. 1995;89(2):221–40.

Liver mitochondrial membrane crosslinking and destruction in a rat model of [180] Koizumi M, Fujii J, Suzuki K, Inoue T, Inoue T, Gutteridge JM, et al. A marked

Wilson disease. J Clin Invest 2011;121(4):1508–18. increase in free copper levels in the plasma and liver of LEC rats: an ani-

[151] Hayashi H, Yano M, Fujita Y, Wakusawa S. Compound overload of copper mal model for Wilson disease and liver cancer. Free Radic Res 1998;28(5):

and iron in patients with Wilson’s disease. Med Mol Morphol 2006;39(3): 441–50.

121–6. [181] Suzuki KT, Kanno S, Misawa S, Sumi Y. Changes in hepatic copper distribu-

[152] Barnes N, Tsivkovskii R, Tsivkovskaia N, Lutsenko S. The copper-transporting tion leading to hepatitis in LEC rats. Res Commun Chem Pathol Pharmacol

ATPases, Menkes and Wilson disease proteins, have distinct roles in adult and 1993;82(2):217–24.

developing cerebellum. J Biol Chem 2005;280(10):9640–5. [182] Kim JM, Ko SB, Kwon SJ, Kim HJ, Han MK, Kim DW, et al. Ferrous and ferric

[153] Kodama H, Okabe I, Yanagisawa M, Nomiyama H, Nomiyama K, Nose O, et al. iron accumulates in the brain of aged Long-Evans Cinnamon rats, an animal

Does CSF copper level in Wilson disease reflect copper accumulation in the model of Wilson’s disease. Neurosci Lett 2005;382(1–2):143–7.

brain? Pediatr Neurol 1988;4(1):35–7. [183] Sugawara N, Ohta T, Lai YR, Sugawara C, Yuasa M, Nakamura M, et al. Iron

[154] Weisner B, Hartard C, Dieu C. CSF copper concentration: a new parameter for depletion prevents adenine nucleotide decomposition and an increase of xan-

diagnosis and monitoring therapy of Wilson’s disease with cerebral manifes- thine oxidase activity in the liver of the Long Evans Cinnamon (LEC) rat, an

tation. J Neurol Sci 1987;79(1–2):229–37. animal model of Wilson’s disease. Life Sci 1999;65(13):1423–31.

[155] Stuerenburg HJ, copper concentrations CSF. blood–brain barrier function, and [184] Hayashi H, Wakusawa S, Yano M. Iron removal by phlebotomy for the pro-

coeruloplasmin synthesis during the treatment of Wilson’s disease. J Neural phylaxis of fulminant hepatitis in a Wilson disease model of Long-Evans

Transm 2000;107(3):321–9. Cinnamon Rats. Hepatol Res 2006;35(4):276–80.

[156] Stuerenburg HJ, Eggers C. Early detection of non-compliance in Wilson’s dis- [185] Santon A, Irato P, Medici V, D‘Inca R, Albergoni V, Sturniolo GC. Effect and

ease by consecutive copper determination in cerebrospinal fluid. J Neurol possible role of Zn treatment in LEC rats, an animal model of Wilson’s disease.

Neurosurg Psychiatry 2000;69(5):701–2. Biochim Biophys Acta 2003;1637(1):91–7.

[157] Litwin T, Gromadzka G, Szpak GM, Jablonka-Salach K, Bulska E, [186] Allen KJ, Buck NE, Cheah DM, Gazeas S, Bhathal P, Mercer JF. Chronological

Czlonkowska A. Brain metal accumulation in Wilson’s disease. J Neurol Sci changes in tissue copper, zinc and iron in the toxic milk mouse and effects of

2013;329(1–2):55–8. copper loading. Biometals 2006;19(5):555–64.

[158] Cumings JN. The copper and iron content of brain and liver in the normal and [187] Przybylkowski A, Gromadzka G, Wawer A, Bulska E, Jablonka-Salach K,

in hepato-lenticular degeneration. Brain 1948;71(Pt. 4):410–5. Grygorowicz T, et al. Neurochemical and behavioral characteristics of

[159] Faa G, Lisci M, Caria MP, Ambu R, Sciot R, Nurchi VM, et al. Brain copper, iron, toxic milk mice: an animal model of Wilson’s disease. Neurochem Res

magnesium, zinc, calcium, sulfur and phosphorus storage in Wilson’s disease. 2013;38(10):2037–45.

J Trace Elem Med Biol 2001;15(2–3):155–60. [188] Chen DB, Feng L, Lin XP, Zhang W, Li FR, Liang XL, et al. Penicillamine increases

[160] Horoupian DS, Sternlieb I, Scheinberg IH. Neuropathological findings in free copper and enhances oxidative stress in the brain of toxic milk mice. PLoS

penicillamine-treated patients with Wilson’s disease. Clin Neuropathol ONE 2012;7(5):e37709.

1988;7(2):62–7. [189] Peng F, Lutsenko S, Sun X, Muzik O. Imaging copper metabolism imbalance

− −

[161] Sinha S, Taly AB, Ravishankar S, Prashanth LK, Venugopal KS, Arunodaya GR, in Atp7b( / ) knockout mouse model of Wilson’s disease with PET-CT and

64

et al. Wilson’s disease: cranial MRI observations and clinical correlation. Neu- orally administered CuCl2. Mol Imaging Biol 2012;14(5):600–7.

roradiology 2006;48(9):613–21. [190] Merle U, Tuma S, Herrmann T, Muntean V, Volkmann M, Gehrke SG,

[162] Sinha S, Taly AB, Prashanth LK, Ravishankar S, Arunodaya GR, Vasudev MK. et al. Evidence for a critical role of ceruloplasmin oxidase activity in iron

Sequential MRI changes in Wilson’s disease with de-coppering therapy: a metabolism of Wilson disease gene knockout mice. J Gastroenterol Hepatol

study of 50 patients. Br J Radiol 2007;80(957):744–9. 2010;25(6):1144–50.

[163] Skowronska M, Litwin T, Dziezyc K, Wierzchowska A, Czlonkowska A. Does [191] Pal A, Vasishta R, Prasad R. Hepatic and hippocampus iron status is not altered

brain degeneration in Wilson disease involve not only copper but also iron in response to increased serum ceruloplasmin and serum “free” copper in

accumulation? Neurol Neurochir Pol 2013;47(6):542–6. Wistar rat model for non-Wilsonian brain copper toxicosis. Biol Trace Elem

[164] Lee JH, Yang TI, Cho M, Yoon KT, Baik SK, Han YH. Widespread cerebral cor- Res 2013;154(3):403–11.

tical mineralization in Wilson’s disease detected by susceptibility-weighted [192] Chakraborty S, Aschner M. Altered manganese homeostasis: implications for

imaging. J Neurol Sci 2012;313(1–2):54–6. BLI-3-dependent dopaminergic neurodegeneration and SKN-1 protection in

[165] Engelbrecht V, Schlaug G, Hefter H, Kahn T, Modder U. MRI of the brain C. elegans. J Trace Elem Med Biol 2012;26(2–3):183–7.

in Wilson disease: T2 signal loss under therapy. J Comput Assist Tomogr [193] Kim Y. High signal intensities on T1-weighted MRI as a biomarker of exposure

1995;19(4):635–8. to manganese. Ind Health 2004;42(2):111–5.

[166] Brugieres P, Combes C, Ricolfi F, Degos JD, Poirier J, Gaston A. Atypical MR pre- [194] Sikk K, Haldre S, Aquilonius SM, Asser A, Paris M, Roose A, et al. Manganese-

sentation of Wilson disease: a possible consequence of paramagnetic effect induced parkinsonism in methcathinone abusers: bio-markers of exposure

of copper? Neuroradiology 1992;34(3):222–4. and follow-up. Eur J Neurol 2013;20(6):915–20.

G Model

JTEMB-25537; No. of Pages 11 ARTICLE IN PRESS

P. Dusek et al. / Journal of Trace Elements in Medicine and Biology xxx (2014) xxx–xxx 11

[195] Jellinger KA. Neuropathology of sporadic Parkinson’s disease: evaluation and [206] Hininger I, Waters R, Osman M, Garrel C, Fernholz K, Roussel AM, et al. Acute

changes of concepts. Mov Disord 2012;27(1):8–30. prooxidant effects of vitamin C in EDTA and long-term

[196] Ayton S, Lei P. Nigral iron elevation is an invariable feature of Parkin- benefits of therapy. Free Radic Biol Med 2005;38(12):1565–70.

son’s disease and is a sufficient cause of neurodegeneration. Biomed Res Int [207] Crisponi G, Nurchi VM, Fanni D, Gerosa C, Nemolato S, Faa G. Copper-

2014;2014:581256. related diseases: from chemistry to molecular pathology. Coord Chem Rev

[197] Cabantchik ZI, Munnich A, Youdim MB, Devos D. Regional siderosis: a new 2010;254(7–8):876–89.

challenge for iron chelation therapy. Front Pharmacol 2013;4:167. [208] Szczerbowska-Boruchowska M, Krygowska-Wajs A, Adamek D. Elemen-

[198] Beal MF, Oakes D, Shoulson I, Henchcliffe C, Galpern WR, Haas R, et al. A tal micro-imaging and quantification of human substantia nigra using

randomized clinical trial of high-dosage coenzyme Q10 in early Parkinson synchrotron radiation based x-ray fluorescence–in relation to Parkinson’s

disease: no evidence of benefit. JAMA Neurol 2014;71(5):543–52. disease. J Phys Condens Matter 2012;24(24):244104.

[199] Persson T, Popescu BO, Cedazo-Minguez A. Oxidative stress in Alzheimer’s [209] Szczerbowska-Boruchowska M, Lankosz M, Ostachowicz J, Adamek D,

disease: why did antioxidant therapy fail? Oxid Med Cell Longev Krygowska-Wajs A, Tomik B, Szczudlik A, Simionovici A, Bohic S. Topographic

2014;2014:427318. and quantitative microanalysis of human central tissue using

[200] Shoulson I. DATATOP: a decade of neuroprotective inquiry, Parkinson Study synchrotron radiation. X-Ray Spectrometry 2004;33(1):3–11.

Group. Deprenyl and tocopherol antioxidative therapy of parkinsonism. Ann [210] Barapatre N, Morawski M, Butz T, Reinert T. Trace element mapping in Parkin-

Neurol 1998;44(3 Suppl. 1):S160–6. sonian brain by quantitative ion beam microscopy. Nuclear Instruments &

[201] Zaitone SA, Abo-Elmatty DM, Shaalan AA. Acetyl-l-carnitine and alpha-lipoic Methods in Physics Research Section B-Beam Interactions with Materials and

acid affect rotenone-induced damage in nigral dopaminergic neurons of rat Atoms 2010;268(11–12): 2156–2159.

brain, implication for Parkinson’s disease therapy. Pharmacol Biochem Behav [211] Reinert T, Fiedler A, Morawski M, Arendt T. High resolution quantitative ele-

2012;100(3):347–60. ment mapping of neuromelanin-containing neurons. Nuclear Instruments &

[202] Squitti R. Metals in Alzheimer’s disease: a systemic perspective. Front Biosci Methods in Physics Research Section B-Beam Interactions with Materials and

(Landmark Ed) 2012;17:451–72. Atoms 2007;260(1):227–30.

[203] Roos PM, Vesterberg O, Syversen T, Flaten TP, Nordberg M. Metal concentra- [212] Morawski M, Meinecke C, Reinert T, Dorffel AC, Riederer P, Arendt T, Butz

tions in cerebrospinal fluid and blood plasma from patients with amyotrophic T. Determination of trace elements in the human substantia nigra. Nuclear

lateral sclerosis. Biol Trace Elem Res 2013;151(2):159–70. Instruments & Methods in Physics Research Section B-Beam Interactions with

[204] Stamelou M, Tuschl K, Chong WK, Burroughs AK, Mills PB, Bhatia Materials and Atoms 2005;231 (1–4): 224–228.

KP, et al. Dystonia with brain manganese accumulation resulting from [213] Kienzl E, Puchinger L, Jellinger K, Linert W, Stachelberger H, Jameson RF. 1995.

SLC30A10 mutations: a new treatable disorder. Mov Disord 2012;27(10): The role of transition metals in the pathogenesis of Parkinson’s disease. J

1317–22. Neurol Sci 1995: 134 (Suppl) 69–78.

[205] Herrero Hernandez E, Discalzi G, Valentini C, Venturi F, Chio A, Carmellino [214] Dexter DT, Wells FR, Lees AJ, Agid F, Agid Y, Jenner P, Marsden CD. Increased

C, et al. Follow-up of patients affected by manganese-induced Parkinsonism nigral iron content and alterations in other metal ions occurring in brain in

after treatment with CaNa2EDTA. Neurotoxicology 2006;27(3):333–9. Parkinson’s disease. J Neurochem 1989;52(6):1830–6.