Hostname: page-component-7c8c6479df-995ml Total loading time: 0 Render date: 2024-03-27T10:46:44.667Z Has data issue: false hasContentIssue false

Copper and iron in Alzheimer's disease: a systematic review and its dietary implications

Published online by Cambridge University Press:  18 July 2011

Martin Loef
Affiliation:
European University Viadrina, Institute of Transcultural Health Studies, Große Scharrnstraße 59, 15230Frankfurt (Oder), Germany Samueli Institute, European Office, Frankfurt (Oder), Germany
Harald Walach*
Affiliation:
European University Viadrina, Institute of Transcultural Health Studies, Große Scharrnstraße 59, 15230Frankfurt (Oder), Germany Samueli Institute, European Office, Frankfurt (Oder), Germany
*
*Corresponding author: H. Walach, fax +49 335 5534 2348, email walach@europa-uni.de
Rights & Permissions [Opens in a new window]

Abstract

Fe and Cu could represent dietary risk factors for Alzheimer's disease (AD), which has become a global health concern. To establish the relationship between diets high in Cu and Fe and cognitive decline or AD, we have conducted a systematic review of the literature (up to January 2011). We identified two meta-analyses, two systematic reviews, eleven placebo-controlled trials, five observational studies, forty-five case–control studies, thirty autopsy and five uncontrolled studies, and one case report. There were eleven interventional trials that tried to either supplement or deplete Fe and Cu, but none of them provided clear evidence of a beneficial effect on cognitive performance in patients with AD. The prospective studies revealed an association between a diet simultaneously high in SFA and Cu and cognitive decline. Case–control and autopsy studies showed elevated Fe levels in the brains of AD patients, whereas the evidence was less consistent for Cu. In most of the studies, Cu concentrations were unchanged in the cerebrospinal fluid and the brain but increased in the serum. In conclusion, the existing data suggest that diets excessive in Fe or Cu, together with a high intake of SFA, should be avoided in the elderly who are not at risk of anaemia. Basic studies and, building on this, clinical investigations are needed to further elucidate in which dietary patterns and in which patient groups an Fe- and Cu-rich diet might foster the risk of developing AD.

Type
Systematic Review
Copyright
Copyright © The Authors 2011

Fe and Cu are essential to human life(Reference Mertz1). Their chemical properties as transition metals make them crucial for a plethora of important biological processes such as oxidative metabolism, electron transport in mitochondria and cellular immune response(Reference Muñoz, Villar and García-Erce2, Reference Linder and Hazegh-Azam3). On the other hand, both metals catalyse the Fenton and the Haber–Weiss reactions, producing reactive oxygen species(Reference Kehrer4), which can foster the pathological paths of neurodegenerative disorders(Reference Rayman5) and have been implicated in age-associated diseases(Reference Brewer6Reference Weinberg and Miklossy9). In the brain, they are found at high concentrations(Reference Gerlach, Ben-Shachar and Riederer10, Reference Gaggelli, Kozlowski and Valensin11) that increase with age(Reference Hallgren and Sourander12Reference Speziali, Orvini and Zatta15), and increasing evidence suggests that the neuronal homeostases of Cu, Fe and Zn are altered in Alzheimer's disease (AD)(Reference Bush16). Although Zn has multiple physiological roles in AD including the aggregation and degradation of the amyloid β protein (Aβ)(Reference Watt, Whitehouse and Hooper17), it is redox silent and distinct from Cu and Fe in its chemical properties, its age- and tissue-specific concentration dynamics and its nutritional status in the elderly. Therefore, the present review focuses on Cu and Fe only.

AD is a progressive brain disease that symptomatically leads to an impairment of memory and diverse cognitive functions(18). It accounts for up to 75 % of the 35·6 million dementia cases, which are estimated to have occurred globally in 2010(19). Its prevalence is forecasted to quadruple by 2050, when it is anticipated that one in eighty-five persons will be living with AD(Reference Brookmeyer, Johnson and Ziegler-Graham20).

Whereas familial AD is inherited in an autosomal dominant manner, a variety of risk factors influence the sporadic late-onset AD that accounts for the vast majority of all cases(Reference Campion, Dumanchin and Hannequin21). Thus, a number of genetic risk factors have been identified, such as mutations in genes of the apoE, the amyloid β precursor protein (APP) or the presenilin 1 and 2, which participate in the cleavage of APP(Reference Brouwers, Sleegers and Van Broeckhoven22). In addition, there is increasing evidence of the important roles of metals in the molecular biology of AD(Reference Bush16) and lifestyle parameters – in particular diet – as protective factors(Reference Butterfield, Castegna and Pocernich23Reference Solfrizzi, Panza and Capurso25).

The characteristic neuropathological features of AD are intracellular neurofibrillary tangles (brain regions: entorhinal cortex, hippocampus, amygdala, limbic system and isocortex) and extracellular plaques laden with the Aβ (brain region: isocortex)(Reference Braak and Braak26). The amyloid hypothesis suggests that the amyloid precursor protein is processed in neurons into Aβ, which in turn triggers a cascade of events inducing oxidative stress, neuronal dysfunction, impaired plasticity and neurogenesis, and finally leading to apoptosis(Reference Mattson27). In this process, it has been hypothesised that Aβ passes through the membrane and aggregates to amyloid plaques(Reference Bush and Tanzi28) in the presence of high concentrations of Cu, Fe and Zn(Reference Lovell, Robertson and Teesdale29). Fe and Cu may also mediate the toxicity of Aβ by hypermetallating the peptide, leading to increased oxidative stress. Despite the involvement of both metals in the molecular pathology of AD, their homeostatic deviances in patients with AD are still a matter of debate.

In the light of the importance of Cu and Fe for the development of AD(Reference Hung, Bush and Cherny30, Reference Zecca, Youdim and Riederer31), we conducted a systematic review to address the clinical relationship between Cu and Fe and AD and to discuss the dietary implications.

Method

We searched for studies dealing with Fe and/or Cu and AD. We used terms related to AD (e.g. dementia, cognitive decline and cognitive impairment) and Fe or Cu. We searched the following databases from their start date to January 2011: Medline; Embase; Cochrane Central Register of Controlled Trials; Cochrane Database of Systematic Reviews; Biosis; Science Citation Index; Publisher Database of Kluwer, Karger, Springer, Thieme, Krause & Paschernegg; Toxibo; Clinicaltrials.gov and the ALOIS register by the Cochrane Dementia and Cognitive Improvement Group. We also searched key authors' names and reference lists in the most recent and most cited published research and review articles.

No selection criteria were applied concerning the research design or the type of work; thus all papers written in English that dealt with Fe/Cu and AD in humans were integrated.

Ethics statement

The studies included in the present review were conducted according to the guidelines laid down in the Declaration of Helsinki, and all procedures involving human subjects/patients were approved by the respective ethics committee (e.g. institutional review board of Rush University Medical Center(Reference Morris, Evans and Tangney32)). Written (or verbal) informed consent was obtained from all subjects/patients. For animal studies, institutional and national guidelines for the care and use of animals were followed, and all experimental procedures involving animals were approved by the respective ethics committee.

Results

In total, we integrated 101 studies in the present review. There are two meta-analyses(Reference Bucossi, Ventriglia and Panetta33, Reference Ellervik, Birgens and Tybjærg Hansen34), two systematic reviews(Reference Jenagaratnam and McShane35, Reference Sampson, Jenagaratnam and McShane36), eleven randomised controlled trials(Reference Kessler, Bayer and Bach37Reference McLachlan, Kruck and Kalow49), two prospective studies(Reference Morris, Evans and Tangney32, Reference Squitti, Bressi and Pasqualetti50), three cross-sectional studies(Reference Milward, Bruce and Knuiman51Reference Lam, Kritz-Silverstein and Barrett-Connor53), forty-five case–control studies(Reference Bartzokis, Sultzer and Cummings54Reference Squitti, Quattrocchi and Forno98), thirty autopsy studies(Reference Loeffler, DeMaggio and Juneau65, Reference Squitti, Quattrocchi and Forno98Reference Connor, Tucker and Johnson126), five uncontrolled studies(Reference Landeghem, Sikström and Beckman127Reference Salustri, Barbati and Ghidoni131) and one case study(Reference Squitti, Cassetta and Dal Forno132). A total of two studies were published twice(Reference Kessler, Bayer and Bach37, Reference Kessler, Pajonk and Bach38, Reference Lannfelt, Blennow and Zetterberg46, Reference Faux, Ritchie and Gunn47), one study was both a case–control and an autopsy study(Reference Squitti, Quattrocchi and Forno98).

Randomised-controlled trials

Supplementation of metals

There is only one study in which Fe or Cu was supplemented as a single component in patients with AD. Kessler et al. (Reference Kessler, Bayer and Bach37, Reference Kessler, Pajonk and Bach38) conducted a prospective, randomised, double-blind and placebo-controlled phase II clinical trial with patients suffering from mild AD. They received either 8 mg Cu daily (n 18) or placebo (n 17) as an add-on to donepezil over 12 months. The treatment showed no benefits to cognitive function measured by the Mini Mental State Examination (MMSE) test and the AD assessment scale-cognitive subscale (ADAS-cog)(Reference Kessler, Bayer and Bach37). Nevertheless, the Cu treatment was associated with a stabilising effect on the cerebrospinal fluid (CSF) levels of Aβ42(Reference Kessler, Bayer and Bach37), whose reduction is used as a biomarker for AD(Reference Hampel, Frank and Broich133). CSF-Aβ42 declined by only 10 % in the Cu group compared with 30 % in the placebo group(Reference Kessler, Pajonk and Bach38).

In the Age-Related Eye Disease Study, 2166 participants aged 65 years or older, whose cognitive status had not been determined, received daily antioxidants, Zn and Cu, antioxidants plus Zn and Cu or placebo for an average of almost 7 years, following which they completed a cognitive battery(Reference Yaffe, Clemons and McBee39). Compared with the placebo treatment, supplementation of antioxidants with or without Zn and Cu did not show an effect on cognitive performance(Reference Yaffe, Clemons and McBee39). However, since a cognitive test at baseline was lacking, the results represent only a cross-sectional comparison of treatment groups.

Further studies aimed to identify the effects of supplementation of multiple nutritional factors (including Cu and/or Fe) on cognitive function in the elderly or on the development of AD(Reference Chandra40Reference Wouters-Wesseling, Wagenaar and Rozendaal44). The outcomes of these studies, however, do not permit deciphering the effects that derive from single components.

Depletion of metals

Contrary to the supplementation of metals, four randomised, placebo-controlled studies(Reference Ritchie, Bush and Mackinnon45Reference McLachlan, Kruck and Kalow49) and one uncontrolled study(Reference Regland, Lehmann and Abedini134) tested different chelators in AD based on the laboratory findings that the formation of amyloid plaques and Aβ neurotoxicity rely on interaction with Cu, Fe and Zn.

There have been two Cochrane systematic reviews(Reference Jenagaratnam and McShane35, Reference Sampson, Jenagaratnam and McShane36) that dealt with the literature concerning the chelating agent clioquinol (PBT-1, iodochlorhydroxyquin) and that concluded an absence of evidence of a beneficial effect. Both the reviews refer to the study of Ritchie et al. (Reference Ritchie, Bush and Mackinnon45), a randomised, double-blind phase II study in which eighteen donepezil-treated patients with AD received placebo or clioquinol for 36 weeks. No significant difference in cognition between the groups could be determined as measured on the ADAS-cog(Reference Ritchie, Bush and Mackinnon45). Initially, PBT-1 had been tested in twenty patients with mild-to-moderate AD, who had received 20 or 80 mg clioquinol for 21 d(Reference Regland, Lehmann and Abedini134). Slight cognitive improvements were reported only in the high-dose group, and there was no placebo-treated control group(Reference Regland, Lehmann and Abedini134).

PBT-2, a second-generation 8-OH quinoline metal protein-attenuating compound, has been tested in a randomised, double-blind study with seventy-eight patients with AD, who were under treatment with acetylcholinesterase inhibitors(Reference Ritchie, Bush and Mackinnon45). The patients received 50 mg PBT-2, 250 mg PBT-2 or placebo per day for 12 weeks. There was no improvement in MMSE or ADAS-cog scores, though CSF-Aβ42 level decreased in a PBT-2-dose-dependent manner(Reference Lannfelt, Blennow and Zetterberg46). In the 250 mg PBT-2 group, two tests on executive function showed improvement over placebo. The primary outcome of the study (safety and tolerability) was met(Reference Lannfelt, Blennow and Zetterberg46, Reference Faux, Ritchie and Gunn47).

The efficacy of the Cu chelating agent d-penicillamine was tested in a randomised, double-blind and placebo-controlled study with thirty-four AD patients over 6 months(Reference Squitti, Rossini and Cassetta48). Although the extent of oxidative stress decreased, no significant differences in change of cognitive functioning between the intervention group and the placebo group could be determined(Reference Squitti, Rossini and Cassetta48).

In an earlier randomised, double-blind and placebo-controlled study(Reference McLachlan, Kruck and Kalow49), desferrioxamine (DFO), a siderophore (Greek: Fe carrier) with a chelating function and an affinity for Cu2+, Zn2+, Fe3+ and Al3+ (Reference Martell and Smith135), had been tested in patients with moderate AD. A total of forty-eight patients received DFO once daily, five times a week for 24 months, placebo or no treatment. Activities of daily living were assessed and video-monitored, revealing a treatment-associated reduction in the rate of decline of daily living skills(Reference McLachlan, Kruck and Kalow49). The no-treatment group was reported to deteriorate twice as rapidly as the DFO-treated group.

Prospective studies

In the study conducted by Morris et al. (Reference Morris, Evans and Tangney32), in a community-based prospective setting, 3718 elderly participants were assessed for cognitive function via a home interview, which included four cognitive tests. Daily diet was measured by a FFQ at 3-year intervals for 6 years. It has been reported that dietary intakes of Cu and Fe were not associated with cognitive function after adjustment for confounders. However, a diet high in Cu combined with a high dietary intake of saturated fats was associated with a much faster rate of cognitive decline, while this interaction was not apparent with intakes of Fe or Zn or cholesterol. Those participants in the highest fifth of Cu intake (derived from high Cu doses in vitamin supplements), combined with a diet high in saturated fats, lost cognition at a rate of 19 years in a period of 6 years. Thus, the cognitive decline was three times higher than expected(Reference Morris, Evans and Tangney32). In contrast, Cu intake was not associated with cognitive change when the diet was not high in saturated fats.

This is corroborated by an observational study on eighty-one subjects with mild-to-moderate AD, clinically followed after 1 year(Reference Squitti, Bressi and Pasqualetti50). Consistent with the findings by Morris et al. (2006), hyperlipidaemic patients with higher levels of Cu were more prone to greater cognitive decline. In addition, free serum Cu levels at baseline were associated with a more severe cognitive decline in MMSE scores over time(Reference Squitti, Bressi and Pasqualetti50). This association was independent of lipid serum levels.

Cross-sectional studies

A cross-sectional study with 800 community-dwelling Australians found no significant association between serum ferritin and cognitive function, measured by the Cambridge Cognitive Test(Reference Milward, Bruce and Knuiman51). In participants with (n 51) or without (n 749) dementia, serum ferritin was not related to cognitive function at either point of measurement(Reference Milward, Bruce and Knuiman51).

In a Spanish study with 260 non-institutionalised elderly, Ortega et al. (Reference Ortega, Requejo and Andrés52) reported that better cognitive function measured by MMSE was associated with greater dietary intake of Fe and other nutrients including vitamin C, Zn and thiamine, while it became worse when the amount of energy supplied by fats and cholesterol increased.

The Rancho Bernardo study found in a sample of 1451 participants that both low and high plasma Fe concentrations were associated with lower performance in certain cognitive tests including total and long-term recall(Reference Lam, Kritz-Silverstein and Barrett-Connor53). In women, Cu and Fe concentrations (n 849) inversely correlated with scores in the long- and short-term recall of the Buschke and Fuld Selective Reminding Test and with performance on the Blessed Information-Memory-Concentration Test(Reference Lam, Kritz-Silverstein and Barrett-Connor53).

Autopsy studies

Studies analysing post-mortem brain tissue samples reveal consistently increased levels of Fe in the brains of AD patients compared with controls(Reference Deibel, Ehmann and Markesbery99, Reference Loeffler, Connor and Juneau102, Reference Connor, Menzies and Martin103, Reference House, St Pierre and McLean105Reference Atamna and Frey110, Reference Cornett, Markesbery and Ehmann124). Particularly, Fe was found at elevated levels in the hippocampus, the amygdala and in parts of the cortex. There is, moreover, a change in the protein level involved in the Fe homeostasis: ferritin, an intracellular Fe-storage-protein, is increased in microglia(Reference Jellinger, Paulus and Grundke-Iqbal111) and senile plaques(Reference Grundke-Iqbal, Fleming and Tung112) with a fivefold increase in the cortical ratio of its protein subunits, the heavy and the light chain(Reference Connor, Snyder and Arosio123). The latter was disease and brain-region specific and may provide evidence of a dysfunction of the Fe homeostasis on the level of gene expression. Additionally, transferrin (TF) is found in senile plaques at increased concentration(Reference Connor, Snyder and Beard104), and the Fe regulatory protein 2, a cytoplasmic mRNA binding protein, was strongly increased in the AD brain and associated with intraneuronal lesions, including neurofibrillary tangles, senile plaque neurites and neuropil threads(Reference Smith, Wehr and Harris113). Only a few studies report unchanged or decreased Fe levels(Reference Corrigan, Reynolds and Ward114Reference Andrási, Farkas and Scheibler116, Reference Cornett, Ehmann and Wekstein125), but they either did not use short post-mortem interval tissue specimens from well-characterised AD brains(Reference Corrigan, Reynolds and Ward114Reference Andrási, Farkas and Scheibler116) or analysed the pituitary gland, which is a predictor of environmental Hg exposure, but less relevant for the association between AD and Fe(Reference Cornett, Ehmann and Wekstein125).

In the senile plaques of AD patients, Fe and Cu as well as Zn were found at highly elevated levels(Reference Smith, Harris and Sayre100, Reference Sayre, Perry and Harris101, Reference Lovell, Robertson and Teesdale121). In the plaque-free neuropil of patients with AD, Cu and Fe concentrations were approximately two and four times higher, respectively, than in the neuropil of healthy brains(Reference Lovell, Robertson and Teesdale121).

With respect to Cu, autopsy studies reported decreased(Reference Deibel, Ehmann and Markesbery99, Reference Andrási, Farkas and Scheibler116, Reference Plantin, Lying-Tunell and Kristensson117) or unchanged levels(Reference Squitti, Quattrocchi and Forno98, Reference Deibel, Ehmann and Markesbery99, Reference Corrigan, Reynolds and Ward114, Reference Ward and Mason115, Reference Tandon, Ni and Ding118, Reference Religa, Strozyk and Cherny136) in the hippocampus, cerebellum, cortex or amygdala in patients with AD compared with controls (Table 1). Loeffler et al. (Reference Loeffler, LeWitt and Juneau119) found that brain Cu levels tend to increase with age, whereas a relative increase in AD brain tissue was significant only for the frontal cortex and in comparison with young controls, but not in other brain areas nor compared with age-matched controls. In contrast, a relative increase in ceruloplasmin, a Cu-carrying protein that is involved in Fe metabolism, was reported in the CSF, hippocampus, entorhinal cortex, frontal cortex and putamen of AD patients(Reference Loeffler, DeMaggio and Juneau65, Reference Loeffler, LeWitt and Juneau119). The Cu attributable to ceruloplasmin, however, was calculated to account for < 1 % of the regional brain Cu and may therefore represent a compensatory effect due to oxidative stress rather than a surrogate for brain Cu levels(Reference Loeffler, LeWitt and Juneau119). Also, other research groups found relatively decreased(Reference Connor, Tucker and Johnson126) or unchanged(Reference Squitti, Quattrocchi and Forno98, Reference Castellani, Smith and Nunomura120) levels of ceruloplasmin.

Table 1 Alterations of copper or iron in post-mortem brain tissue samples of patients with Alzheimer's disease compared with samples of healthy controls

Case–control studies

Apart from post-mortem analysis, ferritin Fe can also be measured non-invasively with MRI in living brains. MRI studies revealed an increased level of Fe in the brains of patients with AD, particularly in the three basal ganglia regions, putamen, caudate and globus pallidus(Reference Bartzokis, Sultzer and Cummings54, Reference Bartzokis and Tishler56Reference Kirsch, McAuley and Holshouser59. In patients with an early onset of AD, ferritin Fe was found to be particularly elevated in the basal ganglia(Reference Bartzokis, Tishler and Shin57). Another non-invasive method, phase imaging, enables differentiation of the severity of Fe deposition in different brain regions. A case–control study applying phase imaging found higher levels of Fe deposits in the hippocampus of AD patients than in controls(Reference Ding, Chen and Ling60). In addition, the phase values correlated with MMSE and the duration of disease in patients with AD(Reference Ding, Chen and Ling60).

Reports on systemic levels of Fe in subjects with AD are inconsistent; two studies found decreased plasma Fe levels(Reference Basun, Forssell and Wetterberg61, Reference Vural, Demirin and Kara62), whereas others found unchanged concentrations in CSF and serum compared with controls(Reference Molina, Jiménez-Jiménez and Aguilar63, Reference Hershey, Hershey and Varnes64, Reference Squitti, Ghidoni and Scrascia93). The CSF level of TF has been reported to be normal in patients with AD(Reference Loeffler, DeMaggio and Juneau65), whereas the CSF ferritin level was increased(Reference Kuiper, Mulder and Van Kamp66). Another study revealed that the ratio of ceruloplasmin:TF is increased in AD patients and correlated positively with peroxide levels and negatively with serum Fe concentrations(Reference Squitti, Salustri and Siotto97).

A number of further case–control studies investigated an association of certain genes that are involved in Fe homeostasis and the risk of developing AD. Mutations in the haemochromatosis (HFE) gene (e.g. C282Y and H63D), which cause an autosomal recessive disorder that is associated with a deregulation of the Fe metabolism, haemochromatosis(Reference Bomford137), have been reported to be associated with an increased risk of developing AD(Reference Pulliam, Jennings and Kryscio67Reference Sampietro, Caputo and Casatta69). Sampietro et al. (Reference Sampietro, Caputo and Casatta69), for example, compared the HFE genotypes of 107 patients with late-onset AD with that of ninety-nine healthy controls, and found that the frequency of the HEF-H63D mutation was highest in the patients with an early time of disease onset. This finding, however, is not undisputed(Reference Guerreiro, Bras and Santana70Reference Candore, Licastro and Chiappelli72), and a recent meta-analysis of eight(Reference Kehrer4) studies failed to find an association between haemochromatosis genotypes and AD (mild cognitive impairment)(Reference Ellervik, Birgens and Tybjærg Hansen34).

A second Fe-related genotype of current interest is subtype C2 of TF (TF C2), a plasma glycoprotein transporting Fe(Reference Ponka, Beaumont and Richardson138). TF C2 was demonstrated to occur at an increased frequency in patients with AD(Reference Zambenedetti, De Bellis and Biunno73, Reference Landeghem, Sikström and Beckman127, Reference van Rensburg, Carstens and Potocnik128). The study by Zambenedetti et al. (Reference Zambenedetti, De Bellis and Biunno73) involved 132 patients with a diagnosis of probable AD, and two age-matched control subjects for each patient. The report displayed that the risk of AD increases more than five times in apoE ɛ4/ɛ4 carriers and roughly 1·5 times (P = 0·07) in TF C2 carriers with a significant interaction between the two alleles. Moreover, comparative genotyping of healthy controls and patients with AD or mild cognitive impairment revealed that epistatic interaction between the TF C2 gene and the C282Y mutation in HFE leads to an increased risk of AD, which is exacerbated in carriers of apoE ɛ4/ɛ4(Reference Kauwe, Bertelsen and Mayo89, Reference Robson, Lehmann and Wimhurst90).

Conversely, some studies found no association between TF C2 and a higher risk of AD(Reference Kim, Jhoo and Lee85Reference Lleo, Blesa and Angelopoulos88). For instance, in a study with 221 AD patients and 167 controls from a Basque region, an association between apoE ɛ4 allele and increased risk of AD combined with a young age at onset was found, but no association between TF C2 or HFE mutation and disease susceptibility(Reference Blázquez, De Juan and Ruiz-Martínez87). Another group reported that homozygosity of the TF C1 allele, but not TF C2, has a role as a potential risk factor(Reference Zhang, Yang and Zhang91).

In relation to Cu, a number of recent studies reported increased serum Cu(Reference Squitti, Lupoi and Pasqualetti74Reference Smorgon, Mari and Atti80, Reference Jeandel, Nicolas and Dubois82, Reference Gonzalez, Martin and Cacho83, Reference Babiloni, Squitti and Del Percio95, Reference Ozcankaya and Delibas96), plasma Cu(Reference Basun, Forssell and Wetterberg61, Reference Vural, Demirin and Kara62, Reference Arnal, Cristalli and de Alaniz94) or CSF(Reference Basun, Forssell and Wetterberg61) levels of Cu in patients with AD or mild cognitive impairment(Reference Squitti, Ghidoni and Scrascia93) compared with healthy controls. In these studies, total Cu(Reference Brewer, Kanzer and Zimmerman78, Reference Smorgon, Mari and Atti80, Reference Arnal, Cristalli and de Alaniz94, Reference Squitti, Lupoi and Pasqualetti139Reference Agarwal, Kushwaha and Tripathi141), ceruloplasmin(Reference Squitti, Pasqualetti and Dal Forno140, Reference Agarwal, Kushwaha and Tripathi141) as well as ‘free’ Cu (ceruloplasmin non-bound Cu)(Reference Brewer, Kanzer and Zimmerman78, Reference Squitti, Ghidoni and Scrascia93, Reference Squitti, Pasqualetti and Dal Forno140, Reference Zappasodi, Salustri and Babiloni142, Reference Squitti, Barbati and Rossi143) have been found at increased rates. The free Cu fraction correlated positively with CSF biomarkers of AD including Aβ and hyperphosphorylated τ(Reference Squitti, Barbati and Rossi143) and inversely with MMSE(Reference Squitti, Barbati and Rossi143). On the other hand, some researchers found unchanged total Cu serum(Reference Basun, Forssell and Wetterberg61, Reference Molina, Jiménez-Jiménez and Aguilar63, Reference Snaedal, Kristinsson and Gunnarsdottir81, Reference Jeandel, Nicolas and Dubois82, Reference Ozcankaya and Delibas96, Reference Shore, Henkin and Nelson144) and CSF(Reference Molina, Jiménez-Jiménez and Aguilar63, Reference Hershey, Hershey and Varnes64, Reference Squitti, Barbati and Rossi76) concentrations, while only one Turkish study reported relatively decreased Cu serum levels in AD subjects(Reference Vural, Demirin and Kara62). A recent meta-analysis has summarised the aforementioned case studies and reported evidence of increased Cu levels in serum and no difference between patients with AD and controls in CSF Cu levels, whereas data regarding the plasma levels did not allow conclusions to be drawn(Reference Bucossi, Ventriglia and Panetta33).

We found two studies that revealed that carriers of the apoE ɛ4 allele, which has been described as a genetic risk factor for AD(Reference Haan, Shemanski and Jagust145), demonstrate higher levels of circulating Cu than non-carriers(Reference Gonzalez, Martin and Cacho83, Reference Squitti, Ventriglia and Barbati84). In addition, the temporal α-1 electroencephalography activity is more strongly associated with free serum Cu in apoE ɛ4 carriers than in non-carriers(Reference Zappasodi, Salustri and Babiloni77), while the Cu, Zn superoxide dismutase activity in erythrocytes is not correlated with the apoE genotype in AD patients(Reference Rossi, Squitti and Pasqualetti55). AD patients treated with d-penicillamine for 24 weeks had reduced superoxide dismutase activity in erythrocytes(Reference Rossi, Squitti and Pasqualetti55).

Further studies

Further studies of relevance encompass uncontrolled studies and case reports; two uncontrolled studies revealed an association between higher levels of free Cu in serum and worse performance in MMSE, both in AD patients (n 81)(Reference Squitti, Bressi and Pasqualetti50) and in healthy subjects (n 64)(Reference Salustri, Barbati and Ghidoni131), respectively. Other studies with AD patients revealed an association between reduced total plasma Cu levels and worse cognitive performance (ADAS-cog scores)(Reference Pajonk, Kessler and Supprian130) or CSF biomarker levels characteristic for AD (Aβ42 >375 pg/ml, total τ < 445 pg/ml and phosphorylated τ < 61 pg/ml)(Reference Kessler, Pajonk and Meisser129). In a case study with a pair of elderly monozygotic twins that were discordant for AD but with very similar habits and lifestyle, serum Cu and total peroxide levels were 44 % higher in the twin with AD(Reference Squitti, Cassetta and Dal Forno132).

Discussion

Randomised-controlled trials

In the placebo-controlled, randomised trials, there is no clear evidence that supplementation or depletion of Cu or Fe brings beneficial effects on the cognitive performance of patients with AD. All supplementation trials administered Cu as an add-on to normal medication or in combination with other nutrients, which would hamper drawing of any conclusion even if there was a beneficial effect. The depletion studies were small-sampled (n 20–78) and ran over short periods (3–24 months). The trial by Squitti et al. (Reference Squitti, Rossini and Cassetta48) was conducted over 24 weeks; this was not sufficient time to detect a cognitive decline in the placebo group. Implicitly, no final conclusion is possible on the clinical benefit of d-penicillamine without a longer study period. This also applies to the PBT-2 trial over 12 weeks in which MMSE and ADAS-cog scores diverged for the placebo and intervention groups, but effects remained insignificant(Reference Lannfelt, Blennow and Zetterberg46). The early (1993) DFO trial included AD patients diagnosed by ADAS-cog and ran over 2 years. Unfortunately, the dropout rate for performing different cognitive tests (e.g. Wechsler Memory Scale) was too high to analyse anything but the video-recorded behaviour over time, which could be biased by the scoring procedure and behaviour changes induced by the method itself. Furthermore, it is worth noting that DFO is a charged molecule that is rapidly metabolised and does not easily pass the blood–brain barrier, which may limit its applicability; it also is unclear whether the beneficial effects of DFO relied on the chelation of Fe, Al or other metals. In summary, the current evidence arising from randomised controlled trials on the association between Cu or Fe and AD is inconclusive and awaits longer studies with larger samples.

Prospective studies

The paucity of clinical data regarding nutritional trials is countered by a longitudinal study by Morris et al. (Reference Morris, Evans and Tangney32). The study had a large sample (n 3718), a median follow-up of 5·5 years and deduced cognitive scores from four different tests, but may be limited since the mixed effect models were not adjusted for the apoE genotype that influences both lipid(Reference Fotuhi, Mohassel and Yaffe146) and Cu(Reference Zappasodi, Salustri and Babiloni77) metabolism in AD. In addition, dietary Cu intake was calculated by multiplying the daily intake of food with the Cu content of foods, which depends on the soil and can vary regionally. However, it is unlikely that this analytical step heavily biased the results, as the findings of the study were strongest for supplementary Cu intake. While no association was found for cognitive decline and Cu and dietary cholesterol(Reference Morris, Evans and Tangney32), a trial with the cholesterol-lowering drug atorvastatin found that AD patients treated with atorvastatin (n 32) over 1 year showed significant improvements in cognitive performance compared with placebo, and that the treatment was also associated with a reduction in circulating Cu levels in blood(Reference Sparks, Petanceska and Sabbagh147). The second prospective study(Reference Squitti, Bressi and Pasqualetti50) was small-sampled (n 81), with a short follow-up and explored the predictability of cognitive decline in relationship with Cu levels. It provided no information on the size of the effect that simultaneous hyperlipidaemia and high Cu levels have on cognitive impairment or whether apoE influences the association. In summary, the prospective studies suggest that a diet that is rich in both Cu and saturated fats fosters cognitive decline in elderly subjects.

Cross-sectional studies

The findings by Ortega et al. (Reference Ortega, Requejo and Andrés52) are based on comparing groups with MMSE scores ≥ 28 and < 28. The association of Fe intake and MMSE scores was not adjusted for age, which lowered the scores. Also, the conventional cut-off point for dementia is 24(Reference Petersen, Smith and Waring148), so it is difficult to deduce what the findings mean for the association of dietary Fe with AD. In contrast, another study found systemic Fe and Cu levels to be associated with scores of certain cognitive tests but measured neither MMSE scores nor the prevalence of AD(Reference Lam, Kritz-Silverstein and Barrett-Connor53). The third cross-sectional study(Reference Milward, Bruce and Knuiman51) did not find an association between dementia and serum ferritin levels. However, serum ferritin may not be a reliable proxy for brain Fe(Reference House, St Pierre and Milward149). In summary, the current evidence of cross-sectional studies between an association of AD and Cu or Fe levels is inconclusive.

Autopsy studies

In general, the heterogeneous outcomes of the autopsy studies might have been influenced by several factors. First, there may be differences in the underlying tissue samples according to non-matched differences in the patients including age, sex and genetic or environmental factors such as exposure to toxic elements. Second, the small sample sizes of the studies (nine to twenty-one AD patients; ten to seventeen controls) come with increased variability. Third, it is possible that fixation of brain tissue samples with formalin(Reference Lovell, Robertson and Teesdale29, Reference Samudralwar, Diprete and Ni109, Reference Deibel, Ehmann and Markesbery150) influences the results(Reference Schrag, Dickson and Jiffry151). Fourth, the trace elements were measured by different methods such as inductively coupled MS(Reference Corrigan, Reynolds and Ward114), instrumental neutron activation analyses(Reference Deibel, Ehmann and Markesbery99, Reference Andrási, Farkas and Scheibler116) or radiochemical neutron activation analysis(Reference Tandon, Ni and Ding118). Fifth, there are dissimilarities in the preparation of the post-mortem tissues. For instance, some studies used short-term post-mortem interval tissue specimens and samples from AD cases confirmed by histology(Reference Andrási, Farkas and Scheibler116, Reference Deibel, Ehmann and Markesbery150), while a few did not(Reference Corrigan, Reynolds and Ward114), some measured Cu as μg Cu/g dry brain weight(Reference Deibel, Ehmann and Markesbery99, Reference Plantin, Lying-Tunell and Kristensson117), others referred to wet weight(Reference Tandon, Ni and Ding118, Reference Religa, Strozyk and Cherny136). Any conclusion reached should take these methodological heterogeneities into account. It may nonetheless be summarised that the autopsy studies almost consistently revealed that Fe is increased and that homeostatic Fe regulation is disrupted in the brains of patients with AD. The high levels of Cu, Fe and Zn in the amyloid plaques indicate the central roles of the metals in the formation of the central histological features of AD (for details, see Bush(Reference Bush16)). The situation with Cu levels in brains of AD patients is less clear. Most studies found no significant differences in Cu concentrations between AD patients and controls. Some of those that suggest otherwise compared AD patients with young controls(Reference Loeffler, LeWitt and Juneau119) and used formalin fixation and no short-term post-mortem interval samples (>48 h)(Reference Andrási, Farkas and Scheibler116). However, definitive answers on whether Cu is unchanged in the brain during AD remain a matter of scientific debate.

Case–control studies

The presented case–control studies provide four lines of evidence. First, they confirm the findings of autopsy studies that Fe is increased in the brains of AD patients in comparison with controls by applying phase imaging or MRI. Second, seven case–control studies compared Fe, TF and ferritin levels in plasma, serum and CSF of patients with AD with controls. The studies' heterogeneity may arise from differences in the sample collection and preparation, in the methods used for Fe measurement, in the diagnosis of AD, in the characteristics of the study populations and in the limited number of studies. These all hinder drawing general conclusions on systemic Fe levels in AD. Third, genes associated with Fe metabolism (HFE, TF C2) were analysed for an association with AD. For HFE, a meta-analysis negates such an association; for TF C2, the results are inconsistent and necessitate additional scrutiny. Fourth, case–control studies analysed alterations in Cu levels in plasma, serum and CSF of AD patients in comparison with controls. A current meta-analysis reported increased Cu levels in the serum of AD patients and unchanged CSF levels. The apoE ɛ4 genotype was associated with higher Cu levels than in non-carriers and altered brain activities, suggesting that the increased risk of apoE ɛ4 carriers for AD may partly be based on a role in the effects induced by the dyshomeostasis of Cu.

Further studies

Apart from general limitations that are associated with uncontrolled studies and case reports, the interesting study by Pajonk et al. (Reference Pajonk, Kessler and Supprian130) may be further limited by its short follow-up of 8 weeks, and the fact that the analysis excluded all patients in the highest tertile of plasma Cu levels. The results of further studies(Reference Kessler, Pajonk and Meisser129, Reference Salustri, Barbati and Ghidoni131) highlight the current scientific debate on whether it is more appropriate to address total Cu or free Cu levels in the study of mental decline(Reference Salustri, Barbati and Ghidoni131).

Summary

In conclusion, the current trials provide no conclusive evidence that depletion or supplementation of Cu or Fe is beneficial for AD; prospective studies found that a diet concurrently high in Cu and saturated fats may foster cognitive decline in age; Fe has been consistently found at elevated levels in the brains of AD sufferers by both autopsy and case–control studies. The specific outcomes for Cu are more conflicting; while evidence suggests that the systemic Cu level is increased in patients with AD, further research is needed to define the alterations of Cu in the brain during AD. Also, the relevance of certain genes including TF C2 or apoE awaits further investigation.

Molecular basis

Is the prospective studies' finding biologically plausible? The molecular effects of dietary Cu found in cells and animals are complex and understood only in part. They appear to depend on whether only Cu or Cu and further nutritional factors are experimentally altered.

In the case of a single elevation in Cu levels, the amyloidogenic pathway is inhibited(Reference Bayer, Schäfer and Simons152, Reference Borchardt, Camakaris and Cappai153). One possible explanation for this is a Cu-induced change in APP processing. APP is an integral membrane protein with two Cu binding domains(Reference Multhaup, Schlicksupp and Hesse154), which is cut by endonucleases(Reference Mattson27) into lipid rafts, membrane microdomains enriched in cholesterol and sphingolipids(Reference Hung, Robb and Volitakis155, Reference Simons and Ikonen156). When Cu binds to APP, a conformational change is induced that is thought to affect the clustering of APP in the cell membrane, which, in turn, alters the rate at which APP is processed(Reference Kong, Miles and Crespi157). In consequence, the concentration of Aβ decreases, which would be in line with the findings of some clinical studies(Reference Kessler, Pajonk and Bach38, Reference Kessler, Pajonk and Meisser129, Reference Pajonk, Kessler and Supprian130), but in contradiction to others(Reference Squitti, Lupoi and Pasqualetti74, Reference Squitti, Barbati and Rossi76). Moreover, the processing of the APP depends on the flotillin-2-related endocytosis of APP(Reference Ehehalt, Keller and Haass158, Reference Schneider, Rajendran and Honsho159), which depends on cholesterol(Reference Schneider, Rajendran and Honsho159) and Cu(Reference Hung, Robb and Volitakis155). Increased Cu levels attenuate Aβ synthesis via the inhibition of APP endocytosis(Reference Hung, Robb and Volitakis155).

On the other hand, Cu overload has been reported to result in the overexpression of APP(Reference Armendariz, Gonzalez and Loguinov160), lipid peroxidation, generation of the reactive aldehyde 4-hydrox-2-nonenal and oxidative stress(Reference Hayashi, Shishido and Nakayama161), thus aggravating the vicious circle of AD pathogenesis. A comprehensive understanding of how Cu is involved in AD awaits further research. However, if – in addition to increased Cu intake – further nutritional parameters such as intake of cholesterol or SFA are experimentally altered, the molecular effects appear to be distinct. Hence, the addition of trace amounts (0·12 parts per million) of Cu in distilled water to cholesterol-fed rabbits can induce a reduction in cognitive abilities and increased levels of amyloid plaques(Reference Sparks and Schreurs162).

In vitro experiments have shown that Aβ can form cation channels in the lipid bilayer membrane(Reference Lin, Bhatia and Lal163). Enrichment of cholesterol reduces membrane fluidity and results in the destabilisation of the Aβ channels and an exclusion of Aβ in a metal- and pH-dependent manner(Reference Curtain, Ali and Smith164). In the light of the aforementioned pathogenic effects of Cu overload, additional presence of Cu may further foster neurotoxicity via soluble Aβ and lipid oxidation. Furthermore, hypercholesterolaemia is thought to simultaneously heighten the brain levels of Aβ and Fe(Reference Ghribi, Golovko and Larsen165), which exacerbates oxidative stress(Reference Zecca, Youdim and Riederer31). Hence, the membrane integrity and stability of Aβ in the membrane are delicately balanced, and even small changes in the concentrations of metals, cholesterol or saturated fats may be able to influence the pathogenesis of AD. This conclusion might be limited since brain Fe and Cu homeostases depend on a multitude of molecular players and processes (see for reviews, Hung et al. (Reference Hung, Bush and Cherny30); Zecca et al. (Reference Zecca, Youdim and Riederer31)) that have not all been taken into account in the present review. With regard to clinical evidence, the cholesterol-promoting effect of a high dietary intake ratio of SFA:PUFA should be kept in mind(Reference Keys, Anderson and Grande166Reference Mensink and Katan168). Fig. 1 provides an integration of the effects of Cu and cholesterol into the pathology of AD.

Fig. 1 Effects of cholesterol and Cu on the amyloid β pathology. Considering, in step (1), that cholesterol and Cu levels rise, cholesterol will be integrated into the membrane whose fluidity, in turn, is reduced. The membrane-bound Aβ protein dissociates (2)(Reference Curtain, Ali and Smith164) to form extracellular amyloid plaques at which Fe2+ and Cu+ generate H2O2, which results in lipid peroxidation and the subsequent generation of 4-hydroxynonenal (4HNE), a neurotoxic aldehyde. In the cell, the free Aβ exhibits diverse pathogenic mechanisms (3) including mitochondrial oxidative stress, decreased production of ATP, production of H2O2 in the mitochondria, the Fe- and Cu-catalysed generation of the hydroxyl radical, that induces oxidative stress in the endoplasmic reticulum(Reference Mattson27). Finally (4), the cholesterol-enriched diet can lead to apoptosis, DNA damage, blood–brain barrier disruption, as well as dysregulation at the level of Fe regulatory proteins(Reference Ghribi, Golovko and Larsen165).

However, we are missing a lot of information that could help us see the complete picture. For instance, the complex homeostases of brain Fe and Cu and the relationship of systemic to brain levels are only understood incompletely. Also, the relevance of apoE in the interplay of Fe, Cu and lipid metabolism during AD is unclear. Supplementary experiments in mural AD models at different life stages could give answers to questions of whether the temporal pattern of dietary combinations are relevant to the development of AD and could lay the ground for additional clinical trials.

Practical implications

In a recent prospective study with elderly people, Gu et al. (Reference Gu, Nieves and Stern169) identified a dietary pattern that was strongly associated with a lower risk of developing AD. It included higher intakes of salad dressing, nuts, fish, tomatoes, poultry, cruciferous vegetables, fruits, and dark and green leafy vegetables and a lower intake of high-fat dairy products, red meat and butter(Reference Gu, Nieves and Stern169). The identified dietary pattern contains few SFA and is likely to be associated with a low intake of Fe(Reference Dwyer, Zacharski and Balestra170, Reference Buijsse, Feskens and Moschandreas171). The same could be true for a Mediterranean diet (high intake of vegetables, legumes, fruits, fish, nuts and cereals, but a low intake of saturated lipids, meat and poultry, and a moderate intake of ethanol(Reference Willett, Sacks and Trichopoulou172)), which has been reported to be associated with a reduced risk of AD(Reference Scarmeas, Stern and Tang24). Against the clear clinical evidence of elevated Fe levels in AD, it is tempting to assume that the benefits of a Mediterranean diet on AD do not exclusively rely on a distinct lipid intake but also on a lower level of Fe intake. In contrast, the foods with the highest contents of Cu (beef liver, oysters, molluscs, certain nuts, almonds and cocoa(Reference Lindow, Elvehjem and Peterson173, Reference Lurie, Holden and Schubert174)) are neither typical nor atypical for a Mediterranean diet. Its benefits for AD(Reference Scarmeas, Stern and Tang24) therefore are probably not based on differences in the intake of Cu.

Although circulating Cu relates to the nutritional status of Cu(Reference Solomons175), the origin of the free Cu fraction is still under discussion, potentially relying on inflammation(Reference Akiyama, Barger and Barnum176) or an increased efflux from cortical cells(Reference Bush16). The alteration of systemic and brain Cu level in AD therefore depends on diet to a degree that is not yet known. However, it is startling that the participants on the highest quintile of Cu intake in the Morris Study took Cu in vitamin or mineral supplements(Reference Morris, Evans and Tangney32). This gives significance to the dietary intake of Cu in the elderly regardless of the current status of understanding.

In this light, it is of note that dietary intakes of different metals and their physiological levels in the body are not independent of each other and other factors. Thus, an elevation in Fe levels can be secondary to a high-Cu diet as Cu facilitates Fe intake(Reference Dunlap, James and Hume177), and hypercholesterolaemia might be induced by misbalancing dietary metal intake(Reference Squitti, Salustri and Siotto97). Moreover, the inverse correlation of the blood ceruloplasmin:TF ratio(Reference Squitti, Salustri and Siotto97) may also suggest that brain Fe accumulation is related to systemic alterations, which in turn depend on diet.

Therefore, the practical implications could be to avoid Cu-containing supplements and a high intake of SFA or excessive diet of Fe. Fe deficiency in developed countries is most common in certain subgroups of the population including toddlers and women of childbearing age(Reference Looker, Dallman and Carroll178). Data from the nutritional survey 2007–2008, National Health and Nutrition Examination Survey III, showed that the average nutritional intake of Cu is between 1·3 and 1·6 mg/d (1·1–1·3 mg/d) for adult men (women) in different age cohorts(179), whereas the dietary reference intake is 0·9 mg/d of Cu for adults(180). For Fe, the dietary reference intake recommended by the National Academy of Sciences is 8 (15–18) mg/d for adults (women aged 14–50 years)(180), while the actual intake is 15·6–18·1 mg/d for adult men and 12·6–13·2 mg/d for adult women at different ages(179). That means that the intake of Cu and Fe is up to 100 % higher than recommended. All efforts to reduce Fe or Cu levels should target the metals' physiological windows in order to avoid deficiency but achieve levels that are below the threshold levels critical for AD. What these thresholds look like with regard to AD is speculative with respect to the heterogeneity of different case–control studies. For instance, some studies found total Cu serum levels of >13 μg/l for the controls(Reference Agarwal, Kushwaha and Tripathi79, Reference Smorgon, Mari and Atti80), while others reported levels of < 10 μg/l both, for controls and AD patients(Reference Molina, Jiménez-Jiménez and Aguilar63, Reference Zappasodi, Salustri and Babiloni77). Also, AD is characterised by multiple aetiology and therapeutic ranges of metal reduction may not be generalised without considering further (dietary) parameters.

Despite the eschewal of high dietary intakes of Fe and Cu, various alternative approaches to lowering metal levels in the brain are under investigation. Several molecules acting as chelating agents are currently being developed(Reference Lee, Friedman and Angel181Reference Mandel, Amit and Bar-Am183), while some nutritional constituents have also been examined in relation to their metal-chelating activities.

The green tea polyphenol ( − )-epigallocatechin-3 gallate is being discussed as entailing protective effects on the progression of AD by different mechanisms, including an Fe- chelating function(Reference Weinreb, Mandel and Amit184). Curcumin, a polyphenolic diketone responsible for the yellow colour of turmeric, is also suggested to exert a protective effect against AD by binding Fe2+ and Cu2+ though the binding affinity for Zn2+ is small(Reference Jiao, Wilkinson and Christine Pietsch185, Reference Baum and Ng186). There are more chelating agents being discussed for the treatment of AD. For instance, ethylenediaminetetraacetic acid has been reported to induce improvement in patients with AD(Reference Amit, Avramovich-Tirosh and Youdim182). Pharmaceutical compounds that exert their anti-inflammatory effects by interaction with Cu-, Fe- or Zn-dependent proteins have also been found to lower the risk for developing AD(Reference McGeer, Schulzer and McGeer187), and a number of other molecules that act as chelating agents are currently being developed(Reference Malecki and Connor188). An alternative approach of reducing the level of stored Fe in patients with AD has been hypothesised recently with the use of calibrated phlebotomy that could reduce stored Fe without causing anaemia(Reference Dwyer, Zacharski and Balestra189). All practical implications, however, should be taken cautiously in light of the limited clinical evidence and the multiple causality of AD.

Conclusion

In conclusion, the present systematic review suggests that a diet rich in Cu and Fe might aggravate the detrimental effects of a high intake of cholesterol and SFA on the risk of developing AD. The association is biologically plausible. However, since the relationship between dietary metal and fat intake and dementia is clinically not well examined, additional studies are necessary to further address which nutritional patterns best fit to certain risk groups in the population.

Acknowledgements

The authors are grateful to Majella Horan, European University Viadrina, Institute of Transcultural Health Studies for proofreading. No financial or other conflicts of interest exist for any of the authors. The present study was supported by the Samueli Institute, Alexandria, VA, USA. M. L. analysed the information, performed literature searches and drafted the manuscript. H. W. assisted with the analysis of critical information, and contributed to the drafting of the manuscript and to the interpretation of data.

References

1 Mertz, W (1981) The essential trace elements. Science 213, 13321338.CrossRefGoogle ScholarPubMed
2 Muñoz, M, Villar, I & García-Erce, JA (2009) An update on iron physiology. World J Gastroenterol 15, 46174626.CrossRefGoogle ScholarPubMed
3 Linder, M & Hazegh-Azam, M (1996) Copper biochemistry and molecular biology. Am J Clin Nutr 63, 797S811S.Google ScholarPubMed
4 Kehrer, JP (2000) The Haber–Weiss reaction and mechanisms of toxicity. Toxicology 149, 4350.CrossRefGoogle ScholarPubMed
5 Rayman, MP (2000) The importance of selenium to human health. Lancet 356, 233241.CrossRefGoogle ScholarPubMed
6 Brewer, GJ (2010) Risks of copper and iron toxicity during aging in humans. Chem Res Toxicol 23, 319326.CrossRefGoogle ScholarPubMed
7 Brewer, GJ (2007) Iron and copper toxicity in diseases of aging, particularly atherosclerosis and Alzheimer's disease. Exp Biol Med 232, 323335.Google ScholarPubMed
8 Altamura, S & Muckenthaler, MU (2009) Iron toxicity in diseases of aging: Alzheimer's disease, Parkinson's disease and atherosclerosis. J Alzheimers Dis 16, 879895.CrossRefGoogle ScholarPubMed
9 Weinberg, ED & Miklossy, J (2008) Iron withholding: a defense against disease. J Alzheimers Dis 13, 451463.CrossRefGoogle ScholarPubMed
10 Gerlach, M, Ben-Shachar, D, Riederer, P, et al. (1994) Altered brain metabolism of iron as a cause of neurodegenerative diseases? J Neurochem 63, 793807.CrossRefGoogle ScholarPubMed
11 Gaggelli, E, Kozlowski, H, Valensin, D, et al. (2006) Copper homeostasis and neurodegenerative disorders (Alzheimer's, prion, and Parkinson's diseases and amyotrophic lateral sclerosis. Chem Rev 106, 19952044.CrossRefGoogle ScholarPubMed
12 Hallgren, B & Sourander, P (1958) The effect of age on the non-haemin iron in the human brain. J Neurochem 3, 4151.CrossRefGoogle ScholarPubMed
13 Bartzokis, G, Beckson, M, Hance, DB, et al. (1997) MR evaluation of age-related increase of brain iron in young adult and older normal males. Magn Reson Imaging 15, 2935.CrossRefGoogle ScholarPubMed
14 Zecca, L, Gallorini, M, Schünemann, V, et al. (2001) Iron, neuromelanin and ferritin content in the substantia nigra of normal subjects at different ages: consequences for iron storage and neurodegenerative processes. J Neurochem 76, 17661773.CrossRefGoogle ScholarPubMed
15 Speziali, M & Orvini, E (2003) Metals distribution and regionalization in the brain. In Metal Ions and Neurodegenerative Disorders, pp. 1565 [Zatta, P, editor]. Singapore: World Scientific Publishing Co Inc.CrossRefGoogle Scholar
16 Bush, AI (2003) The metallobiology of Alzheimer's disease. Trends Neurosci 26, 207214.CrossRefGoogle ScholarPubMed
17 Watt, NT, Whitehouse, IJ & Hooper, NM (2011) The role of zinc in Alzheimer's disease. Int J Alzheimers Dis 2011, 971021.Google Scholar
18 Alzheimer's Disease International (2009) World Alzheimer Report. http://www.alz.co.uk/research/files/WorldAlzheimerReport.pdf (cited 2010 01·09·2010).Google Scholar
19 WHO (2009) WHO World Alzheimer Report. Geneva: WHO.Google Scholar
20 Brookmeyer, R, Johnson, E, Ziegler-Graham, K, et al. (2007) Forecasting the global burden of Alzheimer's disease. Alzheimers Dement 3, 186191.CrossRefGoogle ScholarPubMed
21 Campion, D, Dumanchin, C, Hannequin, D, et al. (1999) Early-onset autosomal dominant Alzheimer disease: prevalence, genetic heterogeneity, and mutation spectrum. Am J Hum Genet 65, 664670.CrossRefGoogle ScholarPubMed
22 Brouwers, N, Sleegers, K & Van Broeckhoven, C (2008) Molecular genetics of Alzheimer's disease: an update. Ann Med 40, 562583.CrossRefGoogle ScholarPubMed
23 Butterfield, DA, Castegna, A, Pocernich, CB, et al. (2002) Nutritional approaches to combat oxidative stress in Alzheimer's disease. J Nutr Biochem 13, 444461.CrossRefGoogle ScholarPubMed
24 Scarmeas, N, Stern, Y, Tang, M, et al. (2006) Mediterranean diet and risk for Alzheimer's disease. Ann Neurol 59, 912921.CrossRefGoogle ScholarPubMed
25 Solfrizzi, V, Panza, F & Capurso, A (2003) The role of diet in cognitive decline. J Neural Transm 110, 95110.CrossRefGoogle ScholarPubMed
26 Braak, H & Braak, E (1991) Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol (Berl) 82, 239259.CrossRefGoogle ScholarPubMed
27 Mattson, MP (2004) Pathways towards and away from Alzheimer's disease. Nature 430, 631639.CrossRefGoogle ScholarPubMed
28 Bush, AI & Tanzi, RE (2008) Therapeutics for Alzheimer's disease based on the metal hypothesis. Neurotherapeutics 5, 421432.CrossRefGoogle ScholarPubMed
29 Lovell, M, Robertson, J, Teesdale, W, et al. (1998) Copper, iron and zinc in Alzheimer's disease senile plaques. J Neurol Sci 158, 4752.CrossRefGoogle ScholarPubMed
30 Hung, YH, Bush, AI & Cherny, RA (2010) Copper in the brain and Alzheimer's disease. J Biol Inorg Chem 15, 6176.CrossRefGoogle ScholarPubMed
31 Zecca, L, Youdim, MBH, Riederer, P, et al. (2004) Iron, brain ageing and neurodegenerative disorders. Nat Rev Neurosci 5, 863873.CrossRefGoogle ScholarPubMed
32 Morris, MC, Evans, DA, Tangney, CC, et al. (2006) Dietary copper and high saturated and trans fat intakes associated with cognitive decline. Arch Neurol 63, 10851088.CrossRefGoogle ScholarPubMed
33 Bucossi, S, Ventriglia, M, Panetta, V, et al. (2010) Copper in Alzheimer's disease: a meta-analysis of serum, plasma, and cerebrospinal fluid studies. J Alzheimer's Dis 23, 111.Google Scholar
34 Ellervik, C, Birgens, H, Tybjærg Hansen, A, et al. (2007) Hemochromatosis genotypes and risk of 31 disease endpoints: meta analyses including 66,000 cases and 226,000 controls. Hepatology 46, 10711080.CrossRefGoogle Scholar
35 Jenagaratnam, L & McShane, R (2006) Clioquinol for the treatment of Alzheimer's Disease. Cochrane Database of Systematic Reviews issue 1, CD005380.CrossRefGoogle ScholarPubMed
36 Sampson, EL, Jenagaratnam, L & McShane, R (2008) Metal protein attenuating compounds for the treatment of Alzheimer's disease. Cochrane Database of Systematic Reviews issue 1, CD005380.CrossRefGoogle ScholarPubMed
37 Kessler, H, Bayer, TA, Bach, D, et al. (2008) Intake of copper has no effect on cognition in patients with mild Alzheimer's disease: a pilot phase 2 clinical trial. J Neural Transm 115, 11811187.CrossRefGoogle ScholarPubMed
38 Kessler, H, Pajonk, FG, Bach, D, et al. (2008) Effect of copper intake on CSF parameters in patients with mild Alzheimer's disease: a pilot phase 2 clinical trial. J Neural Transm 115, 16511659.CrossRefGoogle ScholarPubMed
39 Yaffe, K, Clemons, TE, McBee, WL, et al. (2004) Impact of antioxidants, zinc, and copper on cognition in the elderly: a randomized, controlled trial. Neurology 63, 17051707.Google ScholarPubMed
40 Chandra, RK (2001) Effect of vitamin and trace-element supplementation on cognitive function in elderly subjects. Nutrition 17, 709712.CrossRefGoogle ScholarPubMed
41 Gariballa, S & Forster, S (2007) Effects of dietary supplements on depressive symptoms in older patients: a randomised double-blind placebo-controlled trial. Clin Nutr 26, 545551.CrossRefGoogle ScholarPubMed
42 Planas, M, Conde, M, Audivert, S, et al. (2004) Micronutrient supplementation in mild Alzheimer disease patients. Clin Nutr 23, 265272.CrossRefGoogle ScholarPubMed
43 McNeill, G, Avenell, A, Campbell, M, et al. (2007) Effect of multivitamin and multimineral supplementation on cognitive function in men and women aged 65 years and over: a randomised controlled trial. Nutr J 6, 10.CrossRefGoogle Scholar
44 Wouters-Wesseling, W, Wagenaar, L, Rozendaal, M, et al. (2005) Effect of an enriched drink on cognitive function in frail elderly persons. J Gerontol A Biol Sci Med Sci 60, 265270.CrossRefGoogle ScholarPubMed
45 Ritchie, C, Bush, A, Mackinnon, A, et al. (2003) Metal-protein attenuation with iodochlorhydroxyquin (clioquinol) targeting A {beta} amyloid deposition and toxicity in Alzheimer disease: a pilot phase 2 clinical trial. Arch Neurol 60, 16851691.CrossRefGoogle ScholarPubMed
46 Lannfelt, L, Blennow, K, Zetterberg, H, et al. (2008) Safety, efficacy, and biomarker findings of PBT2 in targeting Abeta as a modifying therapy for Alzheimer's disease: a phase IIa, double-blind, randomised, placebo-controlled trial. Lancet Neurolo 7, 779786.CrossRefGoogle ScholarPubMed
47 Faux, NG, Ritchie, CW, Gunn, A, et al. (2010) PBT2 Rapidly improves cognition in Alzheimer's disease: additional phase II analyses. J Alzheimer's Dis 20, 509516.CrossRefGoogle ScholarPubMed
48 Squitti, R, Rossini, PM, Cassetta, Ed-penicill, et al. (2002) lamine reduces serum oxidative stress in Alzheimer's disease patients. Eur J Clin Invest 32, 5159.CrossRefGoogle Scholar
49 McLachlan, DR, Kruck, TP, Kalow, W, et al. (1991) Intramuscular desferrioxamine in patients with Alzheimer's disease. Lancet 337, 13041308.CrossRefGoogle Scholar
50 Squitti, R, Bressi, F, Pasqualetti, P, et al. (2009) Longitudinal prognostic value of serum “free” copper in patients with Alzheimer disease. Neurology 72, 5055.CrossRefGoogle ScholarPubMed
51 Milward, EA, Bruce, DG, Knuiman, MW, et al. (2010) A cross-sectional community study of serum iron measures and cognitive status in older adults. J Alzheimer's Dis 20, 617623.CrossRefGoogle ScholarPubMed
52 Ortega, RM, Requejo, AM, Andrés, P, et al. (1997) Dietary intake and cognitive function in a group of elderly people. Am J Clin Nutr 66, 803809.CrossRefGoogle Scholar
53 Lam, P, Kritz-Silverstein, D, Barrett-Connor, E, et al. (2008) Plasma trace elements and cognitive function in older men and women: the Rancho Bernardo study. J Nutr Health Aging 12, 2227.CrossRefGoogle ScholarPubMed
54 Bartzokis, G, Sultzer, D, Cummings, J, et al. (2000) In vivo evaluation of brain iron in Alzheimer disease using magnetic resonance imaging. Arch Gen Psychiatry 57, 4753.CrossRefGoogle ScholarPubMed
55 Rossi, L, Squitti, R, Pasqualetti, P, et al. (2002) Red blood cell copper, zinc superoxide dismutase activity is higher in Alzheimer's disease and is decreased by-penicillamine. Neurosci Lett 329, 137140.CrossRefGoogle Scholar
56 Bartzokis, G & Tishler, TA (2000) MRI evaluation of basal ganglia ferritin iron and neurotoxicity in Alzheimer's and Huntingon's disease. Cell Mol Biol (Noisy-le-grand) 46, 821833.Google ScholarPubMed
57 Bartzokis, G, Tishler, TA, Shin, IL, et al. (2004) Brain ferritin iron as a risk factor for age at onset in neurodegenerative diseases. Ann N Y Acad Sci 1012, 224236.CrossRefGoogle ScholarPubMed
58 Pankhurst, Q, Hautot, D, Khan, N, et al. (2008) Increased levels of magnetic iron compounds in Alzheimer's disease. J Alzheimer's Dis 13, 4952.CrossRefGoogle ScholarPubMed
59 Kirsch, W, McAuley, G, Holshouser, B, et al. (2009) Serial susceptibility weighted MRI measures brain iron and microbleeds in dementia. J Alzheimer's Dis 17, 599609.CrossRefGoogle ScholarPubMed
60 Ding, B, Chen, KM, Ling, HW, et al. (2009) Correlation of iron in the hippocampus with MMSE in patients with Alzheimer's disease. J Magn Reson Imaging 29, 793798.CrossRefGoogle ScholarPubMed
61 Basun, H, Forssell, LG, Wetterberg, L, et al. (1991) Metals and trace elements in plasma and cerebrospinal fluid in normal aging and Alzheimer's disease. J Neural Transm Park Dis Dement Sect 3, 231258.Google ScholarPubMed
62 Vural, H, Demirin, H, Kara, Y, et al. (2010) Alterations of plasma magnesium, copper, zinc, iron and selenium concentrations and some related erythrocyte antioxidant enzyme activities in patients with Alzheimer's disease. J Trace Elem Med Biol 24, 169173.CrossRefGoogle ScholarPubMed
63 Molina, JA, Jiménez-Jiménez, FJ, Aguilar, MV, et al. (1998) Cerebrospinal fluid levels of transition metals in patients with Alzheimer's disease. J Neural Transm 105, 479488.CrossRefGoogle ScholarPubMed
64 Hershey, C, Hershey, L, Varnes, A, et al. (1983) Cerebrospinal fluid trace element content in dementia: clinical, radiologic, and pathologic correlations. Neurology 33, 13501352.CrossRefGoogle ScholarPubMed
65 Loeffler, D, DeMaggio, A, Juneau, P, et al. (1994) Ceruoplasmin is increased in cerebrospinal fluid in Alzheimer's disease but not Parkinson's disease. Alzheimer Dis Assoc Disord 8, 190197.CrossRefGoogle Scholar
66 Kuiper, MA, Mulder, C, Van Kamp, GJ, et al. (1994) Cerebrospinal fluid ferritin levels of patients with Parkinson's disease, Alzheimer's disease, and multiple system atrophy. J Neural Transm Park Dis Dement Sect 7, 109114.CrossRefGoogle ScholarPubMed
67 Pulliam, JF, Jennings, CD, Kryscio, RJ, et al. (2003) Association of HFE mutations with neurodegeneration and oxidative stress in Alzheimer's disease and correlation with APOE. Am J Med Genet B Neuropsychiatr Genet 119, 4853.CrossRefGoogle Scholar
68 Moalem, S, Percy, ME, Andrews, DF, et al. (2000) Are hereditary hemochromatosis mutations involved in Alzheimer disease? Am J Med Genet 93, 5866.3.0.CO;2-L>CrossRefGoogle ScholarPubMed
69 Sampietro, M, Caputo, L, Casatta, A, et al. (2001) The hemochromatosis gene affects the age of onset of sporadic Alzheimer's disease. Neurobiol Aging 22, 563568.CrossRefGoogle ScholarPubMed
70 Guerreiro, RJ, Bras, JM, Santana, I, et al. (2006) Association of HFE common mutations with Parkinson's disease, Alzheimer's disease and mild cognitive impairment in a Portuguese cohort. BMC Neurol 6, 24.CrossRefGoogle Scholar
71 Berlin, D, Chong, G, Chertkow, H, et al. (2004) Evaluation of HFE (hemochromatosis) mutations as genetic modifiers in sporadic AD and MCI. Neurobiol Aging 25, 465474.CrossRefGoogle ScholarPubMed
72 Candore, G, Licastro, F, Chiappelli, M, et al. (2003) Association between the HFE mutations and unsuccessful ageing: a study in Alzheimer's disease patients from Northern Italy. Mech Ageing Dev 124, 525528.CrossRefGoogle ScholarPubMed
73 Zambenedetti, P, De Bellis, GL, Biunno, I, et al. (2003) Transferrin C2 variant does confer a risk for Alzheimer's disease in Caucasians. J Alzheimer's Dis 5, 423427.CrossRefGoogle Scholar
74 Squitti, R, Lupoi, D, Pasqualetti, P, et al. (2002) Elevation of serum copper levels in Alzheimer's disease. Neurology 59, 11531161.CrossRefGoogle ScholarPubMed
75 Squitti, R, Pasqualetti, P, Dal Forno, G, et al. (2005) Excess of serum copper not related to ceruloplasmin in Alzheimer disease. Neurology 64, 10401046.CrossRefGoogle Scholar
76 Squitti, R, Barbati, G, Rossi, L, et al. (2006) Excess of nonceruloplasmin serum copper in AD correlates with MMSE, CSF beta-amyloid, and h-tau. Neurology 67, 7682.CrossRefGoogle ScholarPubMed
77 Zappasodi, F, Salustri, C, Babiloni, C, et al. (2008) An observational study on the influence of the APOE-epsilon4 allele on the correlation between ‘free’ copper toxicosis and EEG activity in Alzheimer disease. Brain Res 1215, 183189.CrossRefGoogle ScholarPubMed
78 Brewer, GJ, Kanzer, SH, Zimmerman, EA, et al. (2010) Copper and ceruloplasmin abnormalities in Alzheimer's disease. Am J Alzheimer's Dis Other Demen 25, 490497.CrossRefGoogle ScholarPubMed
79 Agarwal, R, Kushwaha, SS, Tripathi, CB, et al. (2008) Serum copper in Alzheimer's disease and vascular dementia. Ind J Clin Biochem 23, 369374.CrossRefGoogle ScholarPubMed
80 Smorgon, C, Mari, E, Atti, A, et al. (2004) Trace elements and cognitive impairment: an elderly cohort study. Arch Gerontol Geriatr 393402.CrossRefGoogle ScholarPubMed
81 Snaedal, J, Kristinsson, J, Gunnarsdottir, S, et al. (2000) Copper, ceruloplasmin and superoxide dismutase in patients with Alzheimer's disease. Dement Geriatr Cogn Disord 9, 239242.CrossRefGoogle Scholar
82 Jeandel, C, Nicolas, MB, Dubois, F, et al. (1989) Lipid peroxidation and free radical scavengers in Alzheimer's disease. Gerontology 35, 275282.CrossRefGoogle ScholarPubMed
83 Gonzalez, C, Martin, T, Cacho, J, et al. (1999) Serum zinc, copper, insulin and lipids in Alzheimer's disease epsilon4 apolipoprotein E allele carriers. Eur J Clin Invest 29, 637642.CrossRefGoogle Scholar
84 Squitti, R, Ventriglia, M, Barbati, G, et al. (2007) ‘Free’ copper in serum of Alzheimer's disease patients correlates with markers of liver function. J Neural Transm 114, 15891594.CrossRefGoogle ScholarPubMed
85 Kim, KW, Jhoo, JH, Lee, JH, et al. (2001) Transferrin C2 variant does not confer a risk for Alzheimer's disease in Koreans. Neurosci Lett 308, 4548.CrossRefGoogle ScholarPubMed
86 Rondeau, V, Iron, A, Letenneur, L, et al. (2006) Analysis of the effect of aluminum in drinking water and transferrin C2 allele on Alzheimer's disease. Eur J Neurol 13, 10221025.CrossRefGoogle ScholarPubMed
87 Blázquez, L, De Juan, D, Ruiz-Martínez, J, et al. (2007) Genes related to iron metabolism and susceptibility to Alzheimer's disease in Basque population. Neurobiol Aging 28, 19411943.CrossRefGoogle ScholarPubMed
88 Lleo, A, Blesa, R, Angelopoulos, C, et al. (2002) Transferrin C2 allele, haemochromatosis gene mutations, and risk for Alzheimer's disease. Br Med J 72, 820821.Google ScholarPubMed
89 Kauwe, JS, Bertelsen, S, Mayo, K, et al. (2010) Suggestive synergy between genetic variants in TF and HFE as risk factors for Alzheimer's disease. Am J Med Genet B Neuropsychiatr Genet 153, 955959.CrossRefGoogle Scholar
90 Robson, KJ, Lehmann, DJ, Wimhurst, VL, et al. (2004) Synergy between the C2 allele of transferrin and the C282Y allele of the haemochromatosis gene (HFE) as risk factors for developing Alzheimer's disease. J Med Genet 41, 261265.CrossRefGoogle ScholarPubMed
91 Zhang, P, Yang, Z, Zhang, C, et al. (2003) Association study between late-onset Alzheimer's disease and the transferrin gene polymorphisms in Chinese. Neurosci Lett 349, 209211.CrossRefGoogle ScholarPubMed
92 Shore, D, Henkin, R, Nelson, N, et al. (1984) Hair and serum copper, zinc, calcium, and magnesium concentrations in Alzheimer-type dementia. J Am Geriatr Soc 32, 892895.CrossRefGoogle ScholarPubMed
93 Squitti, R, Ghidoni, R, Scrascia, F, et al. (2011) Free copper distinguishes mild cognitive impairment subjects from healthy elderly individuals. J Alzheimer's Dis 23, 239248.CrossRefGoogle ScholarPubMed
94 Arnal, N, Cristalli, DO, de Alaniz, MJ, et al. (2010) Clinical utility of copper, ceruloplasmin, and metallothionein plasma determinations in human neurodegenerative patients and their first-degree relatives. Brain Res 1319, 118130.CrossRefGoogle ScholarPubMed
95 Babiloni, C, Squitti, R, Del Percio, C, et al. (2007) Free copper and resting temporal EEG rhythms correlate across healthy, mild cognitive impairment, and Alzheimer's disease subjects. Clin Neurophysiol 118, 12441260.CrossRefGoogle ScholarPubMed
96 Ozcankaya, R & Delibas, N (2002) Malondialdehyde, superoxide dismutase, melatonin, iron, copper, and zinc blood concentrations in patients with Alzheimer disease: cross-sectional study. Croat Med J 43, 2832.Google ScholarPubMed
97 Squitti, R, Salustri, C, Siotto, M, et al. (2010) Ceruloplasmin/transferrin ratio changes in Alzheimer's disease. Int J Alzheimers Dis 2011, 231595.Google ScholarPubMed
98 Squitti, R, Quattrocchi, CC, Forno, GD, et al. (2007) Ceruloplasmin (2-D PAGE) pattern and copper content in serum and brain of Alzheimer disease patients. Biomark Insights 1, 205213.Google ScholarPubMed
99 Deibel, MA, Ehmann, WD & Markesbery, WR (1996) Copper, iron, and zinc imbalances in severly degenerated brain regions in Alzheimer's disease: possible relation to oxidative stress. J Neurol Sci 143, 137142.CrossRefGoogle Scholar
100 Smith, MA, Harris, PL, Sayre, LM, et al. (1997) Iron accumulation in Alzheimer disease is a source of redox-generated free radicals. Proc Natl Acad Sci U S A 94, 98669868.CrossRefGoogle ScholarPubMed
101 Sayre, LM, Perry, G, Harris, PL, et al. (2000) In situ oxidative catalysis by neurofibrillary tangles and senile plaques in Alzheimer's disease: a central role for bound transition metals. J Neurochem 74, 270279.CrossRefGoogle ScholarPubMed
102 Loeffler, DA, Connor, JR, Juneau, PL, et al. (1995) Transferrin and iron in normal, Alzheimer's disease, and Parkinson's disease brain regions. J Neurochem 65, 710716.CrossRefGoogle ScholarPubMed
103 Connor, J, Menzies, S, Martin, S, et al. (1992) A histochemical study of iron, transferrin, and ferritin in Alzheimer's diseased brains. J Neurosci Res 31, 7583.CrossRefGoogle ScholarPubMed
104 Connor, JR, Snyder, BS, Beard, JL, et al. (1992) Regional distribution of iron and iron-regulatory proteins in the brain in aging and Alzheimer's disease. J Neurosci Res 31, 327335.CrossRefGoogle ScholarPubMed
105 House, MJ, St Pierre, TG & McLean, C (2008) 1·4T study of proton magnetic relaxation rates, iron concentrations, and plaque burden in Alzheimer's disease and control postmortem brain tissue. Magn Reson Med 60, 4152.CrossRefGoogle ScholarPubMed
106 Leite, RE, Jacob-Filho, W, Saiki, M, et al. (2008) Determination of trace elements in human brain tissues using neutron activation analysis. J Radioanal Nucl Chem 278, 581584.CrossRefGoogle Scholar
107 Panayi, AE, Spyrou, NM, Ubertalli, LC, et al. (2000) Differences in trace element concentrations between the right and left hemispheres of human brain using INAA. J Radioanal Nucl Chem 244, 205207.CrossRefGoogle Scholar
108 Andrási, E, Farkas, É, Gawlik, D, et al. (2000) Brain iron and zinc contents of German patients with Alzheimer disease. J Alzheimer's Dis 2, 1726.CrossRefGoogle ScholarPubMed
109 Samudralwar, DL, Diprete, CC, Ni, BF, et al. (1995) Elemental imbalances in the olfactory pathway in Alzheimer's disease. J Neurol Sci 130, 139145.CrossRefGoogle ScholarPubMed
110 Atamna, H & Frey, WH 2nd (2004) A role for heme in Alzheimer's disease: heme binds amyloid and has altered metabolism. Proc Natl Acad Sci U S A 101, 1115311158.CrossRefGoogle ScholarPubMed
111 Jellinger, K, Paulus, W, Grundke-Iqbal, I, et al. (1990) Brain iron and ferritin in Parkinson's and Alzheimer's diseases. J Neural Transm Park Dis Dement Sect 2, 327340.CrossRefGoogle ScholarPubMed
112 Grundke-Iqbal, I, Fleming, J, Tung, YC, et al. (1990) Ferritin is a component of the neuritic (senile) plaque in Alzheimer dementia. Acta Neuropathol (Berl) 81, 105110.CrossRefGoogle ScholarPubMed
113 Smith, M, Wehr, K, Harris, P, et al. (1998) Abnormal localization of iron regulatory protein in Alzheimer's disease. Brain Res 788, 232236.CrossRefGoogle ScholarPubMed
114 Corrigan, FM, Reynolds, GP & Ward, NI (1993) Hippocampal tin, aluminum and zinc in Alzheimer's disease. BioMetals 6, 149154.CrossRefGoogle ScholarPubMed
115 Ward, NI & Mason, JA (1987) Neutron activation analysis techniques for identifying elemental status in Alzheimer's disease. J Radioanal Nucl Chem 113, 515526.CrossRefGoogle Scholar
116 Andrási, E, Farkas, É, Scheibler, H, et al. (1995) Al, Zn, Cu, Mn and Fe levels in brain in Alzheimer's disease. Arch Gerontol Geriatr 21, 8997.CrossRefGoogle ScholarPubMed
117 Plantin, LO, Lying-Tunell, U & Kristensson, K (1987) Trace elements in the human central nervous system studied with neutron activation analysis. Biol Trace Elem Res 13, 6975.CrossRefGoogle ScholarPubMed
118 Tandon, L, Ni, BF, Ding, XX, et al. (1994) RNAA for arsenic, cadmium, copper, and molybdenum in CNS tissues from subjects with age-related neurodegenerative diseases. J Radioanal Nucl Chem 179, 331339.CrossRefGoogle Scholar
119 Loeffler, DA, LeWitt, PA, Juneau, PL, et al. (1996) Increased regional brain concentrations of ceruloplasmin in neurodegenerative disorders. Brain Res 738, 265274.CrossRefGoogle ScholarPubMed
120 Castellani, RJ, Smith, MA, Nunomura, A, et al. (1999) Is increased redox-active iron in Alzheimer disease a failure of the copper-binding protein ceruloplasmin? Free Radic Biol Med 26, 15081512.CrossRefGoogle ScholarPubMed
121 Lovell, MA, Robertson, JD, Teesdale, WJ, et al. (1998) Copper, iron and zinc in Alzheimer's disease senile plaques. J Neurol Sci 158, 4752.CrossRefGoogle ScholarPubMed
122 Miller, LM, Wang, Q, Telivala, TP, et al. (2006) Synchrotron-based infrared and X-ray imaging shows focalized accumulation of Cu and Zn co-localized with beta-amyloid deposits in Alzheimer's disease. J Struct Biol 155, 3037.CrossRefGoogle Scholar
123 Connor, JR, Snyder, BS, Arosio, P, et al. (1995) A quantitative analysis of isoferritins in select regions of aged, parkinsonian, and Alzheimer's diseased brains. J Neurochem 65, 717724.CrossRefGoogle ScholarPubMed
124 Cornett, CR, Markesbery, WR & Ehmann, WD (1998) Imbalances of trace elements related to oxidative damage in Alzheimer's disease brain. Neurotoxicology 19, 339345.Google ScholarPubMed
125 Cornett, CR, Ehmann, WD, Wekstein, DR, et al. (1998) Trace elements in Alzheimer's disease pituitary glands. Biol Trace Elem Res 62, 107114.CrossRefGoogle ScholarPubMed
126 Connor, JR, Tucker, P, Johnson, M, et al. (1993) Ceruloplasmin levels in the human superior temporal gyrus in aging and Alzheimer's disease. Neurosci Lett 159, 8890.CrossRefGoogle ScholarPubMed
127 Landeghem, GF, Sikström, C, Beckman, LE, et al. (1998) Transferrin C2, metal binding and Alzheimer's disease. Neuroreport 9, 177179.CrossRefGoogle ScholarPubMed
128 van Rensburg, SJ, Carstens, ME, Potocnik, FC, et al. (1993) Increased frequency of the transferrin C2 subtype in Alzheimer's disease. Neuroreport 4, 12691271.CrossRefGoogle ScholarPubMed
129 Kessler, H, Pajonk, FG, Meisser, P, et al. (2006) Cerebrospinal fluid diagnostic markers correlate with lower plasma copper and ceruloplasmin in patients with Alzheimer's disease. J Neural Transm 113, 17631769.CrossRefGoogle ScholarPubMed
130 Pajonk, FG, Kessler, H, Supprian, T, et al. (2005) Cognitive decline correlates with low plasma concentrations of copper in patients with mild to moderate Alzheimer's disease. J Alzheimer's Dis 8, 2327.CrossRefGoogle ScholarPubMed
131 Salustri, C, Barbati, G, Ghidoni, R, et al. (2010) Is cognitive function linked to serum free copper levels? A cohort study in a normal population. Clin Neurophysiol 121, 502507.CrossRefGoogle Scholar
132 Squitti, R, Cassetta, E, Dal Forno, G, et al. (2004) Copper perturbation in 2 monozygotic twins discordant for degree of cognitive impairment. Arch Neurol 61, 738743.CrossRefGoogle ScholarPubMed
133 Hampel, H, Frank, R, Broich, K, et al. (2010) Biomarkers for Alzheimer's disease: academic, industry and regulatory perspectives. Nat Rev Drug Discov 9, 560574.CrossRefGoogle ScholarPubMed
134 Regland, B, Lehmann, W, Abedini, I, et al. (2000) Treatment of Alzheimer's disease with clioquinol. Dement Geriatr Cogn Disord 12, 408414.CrossRefGoogle Scholar
135 Martell, AE & Smith, RM (1974) Critical Stability Constants. New York, NY: Plenum Press.Google Scholar
136 Religa, D, Strozyk, D, Cherny, RA, et al. (2006) Elevated cortical zinc in Alzheimer disease. Neurology 67, 6975.CrossRefGoogle ScholarPubMed
137 Bomford, A (2002) Genetics of haemochromatosis. Lancet 360, 16731681.CrossRefGoogle ScholarPubMed
138 Ponka, P, Beaumont, C & Richardson, DR (1998) Function and regulation of transferrin and ferritin. Semin Hematol 35, 3554.Google ScholarPubMed
139 Squitti, R, Lupoi, D, Pasqualetti, P, et al. (2002) Elevation of serum copper levels in Alzheimer's disease. Neurology 59, 11531161.CrossRefGoogle ScholarPubMed
140 Squitti, R, Pasqualetti, P, Dal Forno, G, et al. (2005) Excess of serum copper not related to ceruloplasmin in Alzheimer disease. Neurology 64, 10401046.CrossRefGoogle Scholar
141 Agarwal, R, Kushwaha, S, Tripathi, C, et al. (2008) Serum copper in Alzheimer's disease and vascular dementia. Ind J Clin Biochem 23, 369374.CrossRefGoogle ScholarPubMed
142 Zappasodi, F, Salustri, C, Babiloni, C, et al. (2008) An observational study on the influence of the APOE-[epsilon] 4 allele on the correlation between ‘free’ copper toxicosis and EEG activity in Alzheimer disease. Brain Res 1215, 183189.CrossRefGoogle Scholar
143 Squitti, R, Barbati, G, Rossi, L, et al. (2006) Excess of nonceruloplasmin serum copper in AD correlates with MMSE, CSF {beta}-amyloid, and h-tau. Neurology 67, 7682.CrossRefGoogle ScholarPubMed
144 Shore, D, Henkin, RI, Nelson, NR, et al. (1984) Hair and serum copper, zinc, calcium, and magnesium concentrations in Alzheimer-type dementia. J Am Geriatr Soc 32, 892895.CrossRefGoogle ScholarPubMed
145 Haan, MN, Shemanski, L, Jagust, WJ, et al. (1999) The role of APOE epsilon4 in modulating effects of other risk factors for cognitive decline in elderly persons. JAMA 282, 4046.CrossRefGoogle ScholarPubMed
146 Fotuhi, M, Mohassel, P & Yaffe, K (2009) Fish consumption, long-chain omega-3 fatty acids and risk of cognitive decline or Alzheimer disease: a complex association. Nat Clin Pract Neurol 5, 140152.Google ScholarPubMed
147 Sparks, DL, Petanceska, S, Sabbagh, M, et al. (2005) Cholesterol, copper and Abeta in controls, MCI, AD and the AD cholesterol-lowering treatment trial (ADCLT). Curr Alzheimer Res 2, 527539.CrossRefGoogle ScholarPubMed
148 Petersen, RC, Smith, GE, Waring, SC, et al. (1999) Mild cognitive impairment: clinical characterization and outcome. Arch Neurol 56, 303308.CrossRefGoogle ScholarPubMed
149 House, MJ, St Pierre, TG, Milward, EA, et al. (2010) Relationship between brain R(2) and liver and serum iron concentrations in elderly men. Magn Reson Med 63, 275281.CrossRefGoogle ScholarPubMed
150 Deibel, M, Ehmann, W & Markesbery, W (1996) Copper, iron, and zinc imbalances in severely degenerated brain regions in Alzheimer's disease: possible relation to oxidative stress. J Neurol Sci 143, 137142.CrossRefGoogle ScholarPubMed
151 Schrag, M, Dickson, A, Jiffry, A, et al. (2010) The effect of formalin fixation on the levels of brain transition metals in archived samples. Biometals 23, 11231127.CrossRefGoogle ScholarPubMed
152 Bayer, T, Schäfer, S, Simons, A, et al. (2003) Dietary Cu stabilizes brain superoxide dismutase 1 activity and reduces amyloid A production in APP23 transgenic mice. Proc Natl Acad Sci U S A 100, 1418714192.CrossRefGoogle Scholar
153 Borchardt, T, Camakaris, J, Cappai, R, et al. (1999) Copper inhibits beta-amyloid production and stimulates the non-amyloidogenic pathway of amyloid-precursor-protein secretion. Biochem J 344, Pt 2, 461467.CrossRefGoogle ScholarPubMed
154 Multhaup, G, Schlicksupp, A, Hesse, L, et al. (1996) The amyloid precursor protein of Alzheimer's disease in the reduction of copper (II) to copper (I). Science 271, 14061409.CrossRefGoogle Scholar
155 Hung, YH, Robb, EL, Volitakis, I, et al. (2009) Paradoxical condensation of copper with elevated beta-amyloid in lipid rafts under cellular copper deficiency conditions. J Biol Chem 284, 2189921907.CrossRefGoogle ScholarPubMed
156 Simons, K & Ikonen, E (1997) Functional rafts in cell membranes. Nature 387, 569572.CrossRefGoogle ScholarPubMed
157 Kong, GK, Miles, LA, Crespi, GA, et al. (2008) Copper binding to the Alzheimer's disease amyloid precursor protein. Eur Biophys J 37, 269279.CrossRefGoogle Scholar
158 Ehehalt, R, Keller, P, Haass, C, et al. (2003) Amyloidogenic processing of the Alzheimer-amyloid precursor protein depends on lipid rafts. J Cell Biol 160, 113123.CrossRefGoogle Scholar
159 Schneider, A, Rajendran, L, Honsho, M, et al. (2008) Flotillin-dependent clustering of the amyloid precursor protein regulates its endocytosis and amyloidogenic processing in neurons. J Neurosci 28, 28742882.CrossRefGoogle ScholarPubMed
160 Armendariz, AD, Gonzalez, M & Loguinov, AV (2004) Gene expression profiling in chronic copper overload reveals upregulation of Prnp and App. Physiol Genomics 20, 4554.CrossRefGoogle ScholarPubMed
161 Hayashi, T, Shishido, N, Nakayama, K, et al. (2007) Lipid peroxidation and 4-hydroxy-2-nonenal formation by copper ion bound to amyloid-beta peptide. Free Radic Biol Med 43, 15521559.CrossRefGoogle ScholarPubMed
162 Sparks, DL & Schreurs, BG (2003) Trace amounts of copper in water induce beta-amyloid plaques and learning deficits in a rabbit model of Alzheimer's disease. Proc Natl Acad Sci U S A 100, 1106511069.CrossRefGoogle Scholar
163 Lin, H, Bhatia, R & Lal, R (2001) Amyloid beta protein forms ion channels: implications for Alzheimer's disease pathophysiology. FASEB J 15, 24332444.CrossRefGoogle ScholarPubMed
164 Curtain, CC, Ali, FE, Smith, DG, et al. (2003) Metal ions, pH, and cholesterol regulate the interactions of Alzheimer's disease amyloid-beta peptide with membrane lipid. J Biol Chem 278, 29772982.CrossRefGoogle ScholarPubMed
165 Ghribi, O, Golovko, MY, Larsen, B, et al. (2006) Deposition of iron and amyloid plaques is associated with cortical cellular damage in rabbits fed with long term cholesterol enriched diets. J Neurochem 99, 438449.CrossRefGoogle ScholarPubMed
166 Keys, A, Anderson, JT & Grande, F (1965) Serum cholesterol response to changes in the diet: IV. Particular saturated fatty acids in the diet. Metabolism 14, 776787.CrossRefGoogle ScholarPubMed
167 Hegsted, DM, McGandy, RB, Myers, ML, et al. (1965) Quantitative effects of dietary fat on serum cholesterol in man. Am J Clin Nutr 17, 281295.CrossRefGoogle ScholarPubMed
168 Mensink, RP & Katan, MB (1992) Effect of dietary fatty acids on serum lipids and lipoproteins. A meta-analysis of 27 trials. Arterioscler Thromb 12, 911919.CrossRefGoogle ScholarPubMed
169 Gu, Y, Nieves, JW, Stern, Y, et al. (2010) Food combination and Alzheimer disease risk. Arch Neurol 67, 699706.CrossRefGoogle ScholarPubMed
170 Dwyer, BE, Zacharski, LR, Balestra, DJ, et al. (2010) Potential role of iron in a Mediterranean-style diet. Arch Neurol 67, 12861288.CrossRefGoogle Scholar
171 Buijsse, B, Feskens, EJ, Moschandreas, J, et al. (2007) Oxidative stress, and iron and antioxidant status in elderly men: differences between the Mediterranean south (Crete) and northern Europe (Zutphen). Eur J Cardiovasc Prev Rehabil 14, 495500.CrossRefGoogle ScholarPubMed
172 Willett, WC, Sacks, F, Trichopoulou, A, et al. (1995) Mediterranean diet pyramid: a cultural model for healthy eating. Am J Clin Nutr 61, 1402S1406S.CrossRefGoogle ScholarPubMed
173 Lindow, CW, Elvehjem, CA & Peterson, WH (1929) The copper content of plant and animal foods. J Biol Chem 82, 465471.CrossRefGoogle Scholar
174 Lurie, DG, Holden, JM, Schubert, A, et al. (1989) The copper content of foods based on a critical evaluation of published analytical data. J Food Compos Anal 2, 298316.CrossRefGoogle Scholar
175 Solomons, N (1979) On the assessment of zinc and copper nutriture in man. Am J Clin Nutr 32, 856871.CrossRefGoogle ScholarPubMed
176 Akiyama, H, Barger, S, Barnum, S, et al. (2000) Inflammation and Alzheimer's disease. Neurobiol Aging 21, 383421.CrossRefGoogle ScholarPubMed
177 Dunlap, WM, James, GW & Hume, DM (1974) Anemia and neutropenia caused by copper deficiency. Ann Intern Med 80, 470476.Google ScholarPubMed
178 Looker, AC, Dallman, PR, Carroll, MD, et al. (1997) Prevalence of iron deficiency in the United States. JAMA 277, 973976.CrossRefGoogle ScholarPubMed
179 What we eat in America, NHANES 2007–2008 (database on the Internet) 2007–2008 http://www.ars.usda.gov/SP2UserFiles/Place/12355000/pdf/0708/Table_1_NIN_GEN_07.pdf (cited Nov 26th 2010).Google Scholar
180 National Academy of Sciences (2004) Dietary reference tables http://iom.edu/en/Global/News%20Announcements/~/media/Files/Activity%20Files/Nutrition/DRIs/DRISummaryListing2.ashx (cited 2010 Nov 25).Google Scholar
181 Lee, JY, Friedman, JE, Angel, I, et al. (2004) The lipophilic metal chelator DP-109 reduces amyloid pathology in brains of human beta-amyloid precursor protein transgenic mice. Neurobiol Aging 25, 13151321.CrossRefGoogle ScholarPubMed
182 Amit, T, Avramovich-Tirosh, Y, Youdim, MB, et al. (2008) Targeting multiple Alzheimer's disease etiologies with multimodal neuroprotective and neurorestorative iron chelators. FASEB J 22, 12961305.CrossRefGoogle ScholarPubMed
183 Mandel, S, Amit, T, Bar-Am, O, et al. (2007) Iron dysregulation in Alzheimer's disease: multimodal brain permeable iron chelating drugs, possessing neuroprotective-neurorescue and amyloid precursor protein-processing regulatory activities as therapeutic agents. Prog Neurobiol 82, 348360.CrossRefGoogle ScholarPubMed
184 Weinreb, O, Mandel, S, Amit, T, et al. (2004) Neurological mechanisms of green tea polyphenols in Alzheimer's and Parkinson's diseases. J Nutr Biochem 15, 506516.CrossRefGoogle ScholarPubMed
185 Jiao, Y, Wilkinson, J 4th, Christine Pietsch, E, et al. (2006) Iron chelation in the biological activity of curcumin. Free Radic Biol Med 40, 11521160.CrossRefGoogle ScholarPubMed
186 Baum, L & Ng, A (2004) Curcumin interaction with copper and iron suggests one possible mechanism of action in Alzheimer's disease animal models. J Alzheimer's Dis 6, 367377.CrossRefGoogle ScholarPubMed
187 McGeer, PL, Schulzer, M & McGeer, EG (1996) Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer's disease: a review of 17 epidemiologic studies. Neurology 47, 425432.CrossRefGoogle ScholarPubMed
188 Malecki, EA & Connor, JR (2002) The case for iron chelation and/or antioxidant therapy in Alzheimer's disease. Drug Dev Res 56, 526530.CrossRefGoogle Scholar
189 Dwyer, BE, Zacharski, LR, Balestra, DJ, et al. (2009) Getting the iron out: phlebotomy for Alzheimer's disease? Med Hypotheses 72, 504509.CrossRefGoogle ScholarPubMed
Figure 0

Table 1 Alterations of copper or iron in post-mortem brain tissue samples of patients with Alzheimer's disease compared with samples of healthy controls

Figure 1

Fig. 1 Effects of cholesterol and Cu on the amyloid β pathology. Considering, in step (1), that cholesterol and Cu levels rise, cholesterol will be integrated into the membrane whose fluidity, in turn, is reduced. The membrane-bound Aβ protein dissociates (2)(164) to form extracellular amyloid plaques at which Fe2+ and Cu+ generate H2O2, which results in lipid peroxidation and the subsequent generation of 4-hydroxynonenal (4HNE), a neurotoxic aldehyde. In the cell, the free Aβ exhibits diverse pathogenic mechanisms (3) including mitochondrial oxidative stress, decreased production of ATP, production of H2O2 in the mitochondria, the Fe- and Cu-catalysed generation of the hydroxyl radical, that induces oxidative stress in the endoplasmic reticulum(27). Finally (4), the cholesterol-enriched diet can lead to apoptosis, DNA damage, blood–brain barrier disruption, as well as dysregulation at the level of Fe regulatory proteins(165).