1

Journal of Alzheimer’s Disease xx (20xx) x–xx DOI 10.3233/JAD-141194 IOS Press

4

5

roo f

3

Towards a Unified Vision of Copper Involvement in Alzheimer’s Disease: A Review Connecting Basic, Experimental, and Clinical Research

or P

2

Review

uth

1

6

Amit Pala,1 , Mariacristina Siottob,1 , Rajendra Prasada and Rosanna Squittic,d,∗

7

a Department

8

b Don

11

Handling Associate Editor: Roberta Ghidoni

cte

9

dA

10

of Biochemistry, PGIMER, Chandigarh, India Carlo Gnocchi Foundation ONLUS, Milan, Italy c Fatebenefratelli Foundation, AFaR Division; Fatebenefratelli Hospital, Isola Tiberina, Rome, Italy d Laboratorio di Neurodegenerazione, IRCCS San Raffaele Pisana, Italy

Accepted 29 August 2014

23

Keywords: Alzheimer’s disease, ceruloplasmin, copper, metal, subtype

24

INTRODUCTION

15 16 17 18 19 20 21

25 26

co

14

Un

13

rre

22

Abstract. Copper is an essential micronutrient for physiological cell functioning and central nervous system (CNS) development. Indeed, it is a cofactor of many proteins and enzymes in a number of molecular pathways, including energy generation, oxygen transportation, hematopoiesis, cellular growth and metabolism, and signal transduction. This is because it serves as a catalyst of reduction-oxidation (redox) reactions in these processes. When copper is kept under control, bound to special proteins, it yields key properties. However, when it spirals out of control, it is exchanged among small compounds (it is loosely bound to them), and its redox activity makes it dangerous for cell viability, promoting oxidative stress. Copper homeostasis in the CNS is securely synchronized, and perturbations in brain copper levels are known to underlie the pathoetiology of wide a spectrum of common neurodegenerative disorders, including Alzheimer’s disease. The main objective of this review is to provide some of the most relevant evidence gleaned from recent studies conducted on animal models and humans, and to discuss the evidence as it pertains to a new concept: Aberrant copper metabolism, which appears to have a genetic basis, is a modifiable risk factor accelerating Alzheimer’s disease and initiation/progression of cognitive deficits in a percentage of susceptible persons.

12

As life expectancies in industrialized countries continue to increase, cognitive and brain aging are becoming key issues in human health. The cognitive 1 These

authors contributed equally to this work. ∗ Correspondence to: Rosanna Squitti, PhD, Department of Neuroscience, Fatebenefratelli Foundation, AFaR Division; Fatebenefratelli Hospital, Isola Tiberina 00186, Rome, Italy. Tel.: +39 66837385; Fax: +39 66837300; E-mail: [email protected].

decline that occurs during aging negatively impacts quality of life (albeit with varying degrees of severity) by disrupting learning and memory abilities. The present review specifically addresses three important topics. That is, it: 1) provides information about aberrant copper metabolism as a causative risk factor in the initiation/progression of Alzheimer’s disease (AD), connecting experimental results to clinical evidence; 2) summarizes the acquired evidence about a copper subtype of AD and discusses its estimated

ISSN 1387-2877/14/$27.50 © 2014 – IOS Press and the authors. All rights reserved

27 28 29 30 31 32 33 34 35 36

44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79

80 81

82 83 84 85

roo f

43

or P

42

chondrial oxidative phosphorylation, neurotransmitter synthesis and denaturation, free radical detoxification, and pigment formation [6, 7]. Conversely, excess copper is highly cytotoxic, as it can readily interact with oxygen to produce extremely damaging free hydroxyl radicals unless homeostasis is tightly regulated [7, 8]. Copper toxicity occurs as a consequence of the generation of reactive oxygen species (ROS) by copper ions via Fenton or Haber–Weiss reactions, which are responsible for lipid peroxidation in membranes, protein oxidation, and the cleavage of DNA and RNA molecules [9–12]. As the central nervous system (CNS) is mainly composed of polyunsaturated lipids (lipids are a major constituent of neuronal membranes), it is particularly susceptible to oxidative stress and cellular damage. In addition, the high flux of ROS produced during reduction-oxidation (redox) reactions is extremely damaging to CNS functioning [13–15]. It is intriguing to note that numerous investigations have implicated copper dyshomeostasis directly or indirectly in the pathogenesis of several neurological and neurodegenerative diseases, like AD, Parkinson’s disease, Wilson’s disease aceruloplasminemia, Menkes disease, amyotrophic lateral sclerosis, Huntington’s disease, occipital horn syndrome, and prion disease (reviewed in [4, 16–20]). Even though some of these are diseases characterized by true copper defects and others are diseases where copper influences only the course of the disease, they all show the relevance of copper physiology in humans. They also show that, in case of a dysmetabolism, copper eventually leads to a disorder in which the neurological and neurodegenerative connotation is strongly represented. Emblematic of copper toxicosis is Wilson’s disease [21]. Wilson’s disease (or hepatolenticular degeneration) is an autosomal recessive genetic disorder in which copper, in the form of a copper not bound to ceruloplasmin (Non-Cp-Cu) is increased in general circulation and accumulates in organs and tissues. Deposits in the cornea, named Kayser-Fleischer rings, are typical. Clinical symptoms can have a liver, neurological, or psychiatric presentation. Mutations in the Wilson’s disease protein (ATP7B) gene cause the disease. A single abnormal copy of the gene is present in 1 in 100 people. Nevertheless, the accumulated evidence is in agreement over the basic role copper toxicity plays in the initiation/development of neurodegenerative diseases, especially AD. AD is a multifactorial disease caused by host of genetic and environmental factors. It is the most common cause of dementia, and it is characterized by the progressive neurodegeneration associated with

dA

41

cte

40

rre

39

annual incidence; and 3) discuss human copper intake in terms of its content in drinking water, as well as in foods. To address the first topic: evidence from milestone studies in in vitro and animal models of copper toxicity have been provided. They focus on copper toxicity, which induces AD-like pathology in diverse animal models via drinking water supplemented with trace levels of copper. It is important to define the terminology that we have used throughout the review. Copper in the diet represents the exclusive way copper comes into the body, since that from inhalation or contact is generally negligible. Food, supplements, and, in a minor percentage, drinking water are considered as sources for intake of copper in humans [1]. In experimental models, the most used approach to increase copper in the diet is to add it in drinking water. In this review, we were interested in providing a unified vision of copper involvement in AD, merging data from experimental models, to those coming from clinical studies, which are data referred to humans, in which copper ingestion is truly from the diet and not merely through drinking water. So, with the expression ‘copper in the diet’ we want to determine the amount of copper intake into the body and we consider copper ingested with drinking water as a part of the copper from diet. This terminology appears to be relevant when comparing evidence from experimental models and data from humans, which need a more holistic interpretation, which is included in the expression ‘copper in the diet’. Subsequently, addressing the second topic, we will show data on humans who demonstrate the association of copper with cognitive worsening. This is not a generalized feature of AD, but it is exhibited by a percentage of patients, who can be distinguished in a subtype of AD [2–5]. These people, who have an aberrant metabolism in handling copper that precedes the clinical diagnosis of AD, are at a higher risk with respect to copper intake. To address the third topic, information about water quality in terms of copper content, as well as copper in foods will be provided. We will try to discuss this data in the framework of recent evidence from large study data-set.

co

38

Un

37

A. Pal et al. / Non-Cp-Cu in AD

uth

2

COPPER TOXICITY AND CENTRAL NERVOUS SYSTEM Copper is an essential trace element that plays a key role in the growth and development of the body. Copper takes part in diverse aspects of metabolism, including connective tissue synthesis, iron metabolism, mito-

86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137

A. Pal et al. / Non-Cp-Cu in AD

146 147 148 149 150 151 152 153 154 155

156 157 158 159

160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186

EVIDENCE SUPPORTING A CAUSATIVE ROLE OF COPPER IN ALZHEIMER’S DISEASE: IN VITRO AND ANIMAL STUDIES Salient studies conducted in the late 1990 s demonstrated the strict interaction of the A␤ peptide with the metals copper, iron and zinc, which react as catalysts. It has been demonstrated that copper reacts with A␤ in reduction-oxidation (redox) cycles, which have as by-products H2 O2 and oxidative stress (Fenton’s and Haber Weiss chemistry). A␤ oligomer formation and precipitation within plaques is prompted in these reactions, along with lipid peroxidation [30–35] amyloid-␤ protein precursor (A␤PP) binds and reduces copper from Cu(II) to Cu(I), triggering oxidative chain reactions that produce H2 O2 and OH•. An additional milestone study demonstrated that A␤ and metals are packed together in plaques within the brain, and chelating agents, which sequester metals, can dissolve these plaques in autoptic brain samples [36]. Results from these studies have provided the basics of the potential effects of ‘copper toxicity’ in AD. Plentiful and diverse animal studies support this role of copper as a toxic agent in AD pathology. In an innovative study of late 1990 s, which suggested direct involvement of the APP gene in copper metabolism, hepatic and cerebral cortex (brain region particularly involved in AD) copper content were shown to be significantly increased in APP−/− and amyloid precursorlike protein (APLP2−/− ) knockout mice [35]. Thus, it is conceivable that APP gene expression modulates

roo f

145

or P

144

uth

143

dA

142

hepatic and cerebral cortex copper levels, but the mechanism leading to increased brain copper content (due to APP gene knockout) is not currently known. Serum copper levels were not significantly more altered in APP−/− and APLP2−/− mice than in wild-type mice. This suggests that a failure of copper excretion from the brain to the blood or, more likely, that an imbalance of copper distribution between the fractions bound and unbound to Cp could take part in the deposition of excessive copper levels in the brain in this knockout model. This will be explained better below. Several findings in experimental animal models account for the observation that the ingestion of inorganic copper has some deleterious effects on CNS functioning. Along with seminal studies demonstrating that the A␤PP is an important regulator of brain copper homeostasis [35], it was demonstrated that it also potentiates the A␤ mediated neurotoxicity by augmenting the oxidative stress [32]. A review of the animal models provides the most direct evidence in favor of the toxic burden brought by copper in AD etiology. In a seminal study published in 2003, Sparks and Schreurs demonstrated that, in a cholesterol fed rabbit model of AD, adding trace amounts of 0.12 ppm (0.12 mg/L) copper to distilled drinking water resulted in significantly enhanced cognitive waning. It also exacerbated amyloid plaque deposition to that of control animals [37]. This finding was somehow provocative, as it led to concerns about the content of copper in drinking water leached from copper pipe-lines. Although cholesterol is vital for neuronal transmission, synaptic plasticity, and cell function, it is also a well-established risk factor for development of atherosclerosis and AD [30]. Cholesterol to 7-hydroxy cholesterol oxidation, caused by A␤, is extremely toxic for neurons [38]. In the cholesterol fed rabbit model, plasma Cp levels, measured using o-dianisidine dihydrochloride as a substrate, suggest an increase in Cp levels, though statistical significance was not attained drinking distilled water supplemented with copper. This suggests that a Non-Cp-Cu increase is a vehicle of copper within the brain. In a different investigation, Sparks reconfirmed his earlier finding, reporting that other animal models, like spontaneously hypercholesterolemic Watanabe rabbits, cholesterol-fed beagles and rabbits, and PS1 and APP transgenic mice showed considerably increased brain levels of A␤ when exposed to drinking water with added copper (0.12 ppm) [39]. Notably, noncholesterol fed PS1 and APP transgenic mice models of AD demonstrated significantly enhanced levels of A␤ due to copper exposure via drinking water. Thus,

cte

141

rre

140

senile plaque deposition and neurofibrillary tangles in the brain. Although the current focus of AD research is on the accumulation of amyloid-␤ (A␤) and tau [22, 23], numerous studies argued that a deregulation of copper metabolism contributes to these pathogenetic pathways and can be a risk factor accelerating the disease cascade [24–26]. We have juxtaposed these two diseases, as they seem to share some common features. As we will detail later, a percentage of AD patients have Non-Cp-Cu higher than normal reference values, have some genetic variants in the ATP7B gene which associates with an increased risk of the disease, and also have increased levels of Cp apo-form in general circulation [27, 28]. A recently published case report notes the presence of Kayser-Fleisher rings in an AD diagnosed patient, studied with [18 F]fluorodeoxyglucose positron emission tomography (PET), and 11 C-labelled Pittsburgh Compound-B PET [29].

co

139

Un

138

3

187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238

246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271

272 273 274

275 276 277 278 279 280 281 282 283 284 285 286 287 288

roo f

245

or P

244

from the gastro-intestinal tract to general circulation, there is no doubt that Non-Cp-Cu is the fraction of blood copper bioavailable for tissues and organs, since it is not structurally bound to Cp (which is actually a master protein in iron metabolism), and it is a low molecular weight copper that can easily be bound and exchanged among the micronutrients, peptide and albumin, for cell uptake. Although this physiological knowledge gives a primary role to Non-Cp-Cu as a causative agent for the modification induced by copper ingestion on A␤ toxicity shown in all the animal models described above, none of these studies ever searched for and provided evidence of an increase of Non-CpCu as a vehicle of this metal, able to pass through the blood-brain barrier (BBB) filter, reach the brain, and partake in the AD cascade. In their recent study, Singh and colleagues [43] demonstrated exactly this simple path for copper, showing what was expected but never proven in AD research by actually connecting two pieces of evidence: one resulting from experimental models investigating the causation of copper in models of AD cascade, and one coming from clinical observational studies and showing Non-Cp-Cu involvement in the clinical picture, prognosis, and prediction of clinical worsening in humans. Ultimately, this yielded unique information: Non-Cp-Cu is a causative risk factor for AD. Indeed, Singh et al. [43] studied normal mice (wild type) and a mouse model of AD (A␤PP transgenic mice) exposed to 0.13 ppm of copper sulfate for 90 days levels of copper via drinking water, which doubled plasma concentrations of Non-Cp-Cu. This caused either a reduction of cerebrospinal fluid A␤ clearance across the BBB in wild-type mice, or an identical effect, along with an increase in A␤ production in the transgenic mice. Thus, we are proposing the reevaluation of a concept already expressed intuitively in the literature [42, 44]. The concept is that Non-Cp-Cu is a causative risk factor for AD. However, we believe this concept pertains only to a certain proportion of AD cases, i.e., those who have a mild metabolic dysfunction of copper, which can be revealed by measuring Non-Cp-Cu in general circulation. This percentage of AD cases can be considered a subtype of AD, as described below.

dA

243

cte

242

rre

241

undoubtedly this mouse model of AD exhibited vulnerability to trace amounts of copper in drinking water even in the absence of cholesterol in the diet. This observation highlights the fact that both cholesterol and copper are causative factors that interact together to further enhance formation of A␤ plaques. They also have significant clinical insinuations, since they provide the rationale for the critical need to reevaluate safe drinking water copper levels. Another study confirmed the findings of Sparks and colleagues [40]. The authors demonstrated that Kunming strain mice fed with high-cholesterol diet and distilled water containing 0.21 ppm copper exhibited significantly increased APP mRNA level coupled with activation of caspase-3 in the brain, suggesting apoptosis mediated neurotoxicity. Strikingly, copper also increased cholesterol-induced learning and memory impairment in mice [40]. Moreover, it has been reported that, in Sprague-Dawley rats, which underwent bilateral common carotid artery occlusion (2VO) and were administered with 250 ppm copper containing water for 3 months, chronic copper toxicity exacerbated memory impairment induced by 2VO coupled with an augmented expression of brain A␤PP and ␤-site A␤PP-cleaving enzyme 1 (BACE1) at both mRNA and protein levels. However, these copper-aggravated changes were ameliorated after copper was withdrawn from the drinking water [41]. As a whole, these experimental animal models demonstrated the toxicity mediated by copper in the AD cascade, showing that increased level of copper ingested with drinking water, or more generally through the diet, affects AD neuropathology. TOWARD A UNIFIED VISION OF COPPER INVOLVEMENT IN ALZHEIMER’S DISEASE

co

240

In humans it is well established that copper ingested through the diet reaches the liver through the portal circulation [1]. Grossly simplifying the metabolic fate of copper absorption, retention, and excretion (detailed in sophisticated studies carried out in metabolic research units using isotopic tracers [1]), it can be said that, after the liver absorption of the quantity necessary for biological needs, copper comes back to the general circulation in the form of Cp or of a Non-Cp-Cu. Some authors [42] argue that the copper taken in by drinking water as inorganic copper comes directly into the bloodstream without passing through the liver, becoming immediately part of the Non-Cp-Cu pool. Regardless of the exact path of the inorganic copper

Un

239

A. Pal et al. / Non-Cp-Cu in AD

uth

4

ASSOCIATION OF COPPER AND ALZHEIMER’S DISEASE: HUMAN STUDIES As we have described above, A␤PP and A␤ itself have been shown to react with copper producing oxidative stress [45–47], eventually damaging biomolecules

289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331 332

333 334 335

336 337 338

A. Pal et al. / Non-Cp-Cu in AD

347 348 349 350 351 352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390

roo f

346

or P

345

uth

344

dA

343

decline and abnormal levels of the exchangeable fraction of low molecular weight Non-Cp-Cu in serum along with its connection with copper intake (reviewed above). This low molecular weight copper has very low concentrations in human physiology: lower than 1.6 ␮mol/L (i.e., 5–15% of total serum copper, which instead ranges between 11–24.4 ␮mol/L [59, 60]). In a series of investigations, it was demonstrated that the average of serum Non-Cp-Cu concentrations are increased in AD [61]. Mini-Mental State Examination (MMSE), as well as other neuropsychological tests of AD subjects, shows correlation between Non-CpCu levels and cognitive performance (the higher the serum Non-Cp-Cu levels, the lower the MMSE score) [25, 61, 62]. It has been estimated that it is filterable and in humans 3% of Non-Cp-Cu crosses the BBB [25]. Moreover, Non-Cp-Cu levels were predictive of MMSE score worsening in AD patients in a year study follow-up [26]; and that Non-Cp-Cu increases in mild cognitive impairment (MCI) [63]—the status which is considered a precursor of AD, due to the high statistical rate of progression from MCI to AD—predicting the rate of progression from MCI to AD [64]. As a whole, these results indicate that the levels of Non-Cp-Cu are higher in AD patients than in healthy individuals, and that this pool of copper is filterable (as exemplified by Wilson’s disease and by the experimental model of AD toxicity) and has effects on cognition (Fig. 1). These three facts were confirmed by the study of James et al. [53].

cte

342

rre

341

and membranes. The Lovell et al. study, which has been considered a milestone study in the field for many years (creating a bias in reference citation), after an accurate revision of its methods, has been judged with many flaws and shortcomings [48]. Detailed information is reported in a recent meta-analysis which reviewed all the data available through 2011, and demonstrated a significant decrease of copper in many areas and, generally, in the brain of AD patients [49]. Two subsequent studies recently confirmed this finding [50, 51]. In their study, Rembach and colleagues [51], found a decrease in the copper present in the soluble AD brain extract (but still bound to proteins), along with a lack of a copper increase in the formic acid fraction, which contained AD plaques that showed no increases of copper. Even though counterintuitive, the evidence of a decrease of copper in the brain is perfectly in agreement with an increase of systemic Non-Cp-Cu. This notion is well supported by animal models of Wilson’s disease. More precisely, the Long Evans Cinnamon (LEC) rats, which have natural mutations in the ATP7B gene, and are then an ecological model of Wilson’s disease, have decreased levels of total copper in their brain and increased systemic Non-Cp-Cu [52], growing the number of shared features between AD and Wilson’s disease. Moreover, the study by James et al. [53], examining in depth the copper distribution in the AD brain, reported either a decrease of total copper in some specific areas of the AD brain, or an increase of the labile (resembling systemic NonCp-Cu) in the same areas. This confirmed a systemic copper dysfunction in AD, in terms of deregulation of Non-Cp-Cu in diverse body districts. In this line, numerous clinical investigations in living patients carried out so far [53–56] have revealed that Cp and copper levels are disturbed in AD, indicating systemic homeostasis deregulation in AD (systemic reviews in [56, 57]). This knowledge has consistently increased in the last 3 years, strongly supporting the original hypothesis of copper toxicity in AD described earlier, based on the biochemical properties of the A␤ peptide and its interaction with metals. Various studies have been published about the link between copper and cognition loss/AD. In a milestone study by Morris et al. [58], it was observed that people who eat a diet rich in saturated and trans-fats and have a higher copper intake experienced a faster rate of cognitive decline. These persons were assumed to be taking copper supplements, by virtue of the fact that they were in the highest quintile of copper intake [58]. Of specific importance are a number of previous findings in human subjects, which provide direct relationships between cognitive

co

340

Un

339

5

COPPER SUBTYPE OF AD Literature on this specific topic has increased in recent years [2–4], even though evidence reported previously certainly sustains this new concept in AD. Generally speaking, to prove the existence of a subtype of a disease, four different strategies should be taken into account: i) identification of a bimodal distribution of the biomarker of interest in the patient population; ii) the response to a specific therapy; iii) cluster analysis; and iv) the identification of a specific biomarker that distinguishes the cases, classifying them in sub-groups that are then compared for demographic, clinical, genetic, or biological variables. When the two hypothetical peaks of the subpopulation are very close, a clear graphical bimodal distribution can be hard to be visualized, since the peaks appear overlapped. Moreover, even in case of clear graphical bimodal distribution, additional evaluations should be done to avoid bias related to sex and age or

391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420

421

422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439

A. Pal et al. / Non-Cp-Cu in AD

dA

uth

or P

roo f

6

rre

cte

Fig. 1. The increase of systemic Non-Cp-Cu in AD causes an imbalance of copper in the brain. Once in the brain, Non-Cp-Cu can interact with physiological amyloid-␤ (A␤), forming clusters of metal-toxic soluble A␤ that evolve in diffuse amyloid and, finally, in toxic plaques. Non-Cp-Cu can also readily interact with reactive oxygen species (ROS), which are responsible for lipid peroxidation in membranes of neuron, protein oxidation, and cleavage of DNA and RNA molecules.

451

Genetic evidence

442 443 444 445 446 447 448 449

452 453 454 455 456 457 458

Un

441

co

450

additional potential confounders. In this case, the other approaches can be more informative. Regarding AD, the application of the procedure described in the fourth approach has recently been discussed. In the study by Murray et al. [65], the authors used the density and the distribution of neurofibrillary tangle counts in the brain at autopsy as a criterion to distinguish subgroups of AD, and then compared these subgroups for various neuropathological features. With regard to copper, we have applied a ‘copper criterion’ to distinguish AD patients in three recent studies, as follows.

440

Recent studies have revealed loci of susceptibility for AD in the master gene pertinent to the Non-Cp-Cu regulation pathway, namely ATP7B [66–70]. One specific study investigated the effect of these ATP7B gene variants on the variation of the biochemical levels of Non-Cp-Cu in AD [70]. In this latter study, after the subject samples were stratified into two classes on the

basis of the Non-Cp-Cu (≤than 1.6 ␮mol/L and > than 1.6 ␮mol/L), we have shown that AD patients with higher levels of Non-Cp-Cu have a higher frequency of some ATP7B variants than AD patients with lower levels of Non-Cp-Cu, thus distinguishing a copper subtype of AD. Prognosis of AD In a study exploring whether a deregulation of NonCp-Cu pool could predict AD clinical worsening, we stratified the AD patients on the basis of a cut-off of Non-Cp-Cu at baseline to better distinguish the two groups (2.1 ␮mol/L). Those patients with levels of Non-Cp-Cu higher than this cut-off had an increased probability to worsen after a year of follow-up than those patients who had lower Non-Cp-Cu values. NonCp-Cu can be then identified as a specific biomarker that distinguishes AD patients into two diverse subgroups that differ in terms of clinical worsening [26].

459 460 461 462 463 464

465

466 467 468 469 470 471 472 473 474 475 476

A. Pal et al. / Non-Cp-Cu in AD

485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527

roo f

484

or P

483

uth

482

dA

481

Non-Cp-Cu levels in AD are not ascribable to any known recognizable condition; ii) ATP7B gene variants increase either the AD risk; iii) or serum Non-Cp-Cu levels in a percentage of AD [70], a primitive mild altered copper metabolism on a genetic basis can be demonstrated in AD. Nevertheless, hard work is still needed to identify the exact loci for AD susceptibility in the ATP7B gene or other genes pertinent to copper metabolism (work in progress). Regarding the exact value of the cut-off better distinguishing AD patients from healthy controls, we have obtained some preliminary data that define more accurately the normal reference values for Non-Cp-Cu, estimated in a cohort of 250 subjects by means of a new patented device to measure Non-Cp-Cu (C4D test, Colabufo, N. and R. Squitti, P.E. European Patent Office (EPO) (RO/EP), 2012). The upper reference limit, (95%) for the healthy population, has been set to 1.91 ␮mol/L (its 90% confidence interval is equal to 1.78–2.06; Code EDMA, 11 02 01 06 00 Copper Cu). Abnormal Non-Cp-Cu is a modifiable risk factor, so it can have relevant effects in contrasting the AD epidemic. In fact, we have calculated the estimated incidence of the copper subtype of AD, starting with the data estimating that there are 90 million inhabitants older than 60 in the region defined as Euro A [76]. In this population, the prevalence of AD is 5.4%, while the annual incidence, i.e., the new cases per year, is nine in a thousand of people. This incidence for those subjects with Non-Cp-Cu higher than normal values is 8% (estimated incidence), or ten times higher. For MCI, that has an incidence of conversion to AD of 15%, the estimated incidence in subjects with Non-Cp-Cu higher than normal values increases to 64%. We are now collecting data to run cluster analyses to substantiate these findings, and we are preparing to run a clinical trial with an anti-copper agent to definitely address the same issues.

cte

480

rre

479

In a recent article [64], Non-Cp-Cu has been revealed as an index in the earliest clinical stages of AD in predicting an increase in the hazard conversion rate and a faster rate of progression from MCI to AD. As it has already been demonstrated for AD patients [26], after the stratification of the MCI subjects on the basis of a cut-off of Non-Cp-Cu at the baseline (1.6 ␮mol/L), the two subgroups differed in terms of their conversion to AD. Those patients with levels of Non-Cp-Cu higher than this cut-off had a three-fold increased hazard of the conversion rate to AD. As a whole, these data allow us to define a copper subtype of AD. If evaluated in the framework of the recent literature, this new concept also allows us to overcome the controversy fuelling a debate that was prevalent in 2006–2012, concerning the issue of whether copper is increased in AD living patients. In fact, meta-analyses, collecting scattered and heterogeneous results into one single piece of evidence, have repetitively and univocally demonstrated that copper and Non-Cp-Cu are increased in AD [56, 71–73]. However, it has to be considered that copper abnormalities are not a condition exhibited by all the AD patients. Taking this into consideration, we can account for the previously controversial results on the basis of the percentage of this copper AD subtype, which has been included in each study. The percentage also varies on the basis of the lifestyle, dietary habits, genetic make-up, ethnicity, and geographic area of the subject sample investigated. Of note is the finding that cognitive function is inversely correlated with Non-Cp-Cu levels, even in healthy subjects [74]. In our 15 years of experience in AD research, we have found that the percentage of AD patients with abnormal levels of Non-Cp-Cu is around 60%, and we have discussed this issue in a previous article published in this same Journal (information available at http://www.j-alz.com/node/182) [56]. In these subjects, the copper dysfunction exhibited cannot be ascribed to recognized conditions that increase NonCp-Cu, such as concomitant Wilson’s disease, acute liver failure of any etiology, chronic cholestasis, and copper intoxication, or supplement (we have also excluded additional diseases known to affect metal metabolism). Moreover, we have demonstrated in recent years the association of some genetic variants of ATP7B with AD [66–70], which have some effects on the concentrations of Non-Cp-Cu in general circulation [70]. Other authors have confirmed our findings [75]. On the basis of these three facts: i) increased

co

478

MCI conversion to AD

Un

477

7

DIETARY COPPER CONTENT: DRINKING WATER AND FOODS Copper enters the body exclusively through dietary intake (food, dietary supplements, drinking water, beverages). Drinking water copper concentrations range from a few ␮g/L to 4.8 mg/L. Taking into consideration the Singh et al. finding [43], it is intriguing to note that 1.3 ppm (1.3 mg/L) copper in drinking water is permissible by the US Environmental Protection Agency, in comparison to the 0.17 ppm (0.17 mg/L) permitted by the California

528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565

566 567

568 569 570 571 572 573 574 575 576

584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613 614 615 616 617 618

619 620 621 622

623 624 625

roo f

583

Age/status

Infants Children 1–3 y 4–6 y Males 9–13 y 14–18 y >19 y Females 9–13 y 14–18 y >19 y During pregnancy During lactation

or P

582

Table 1 Dietary reference intake of copper in various age groups as defined by the Food and Nutrition Board of the Institute of Medicine at the National Academies of Sciences. RDAs (Recommended Dietary Allowances) are reported.

THE INTERPLAY BETWEEN GENETICS AND DIETARY COPPER INTAKE IN BODY COPPER TOXICOSIS OR ACCUMULATION IN HUMANS It is established that whole-body copper metabolism is regulated by a variable efficiency of copper absorption and through excretion of endogenous copper into

RDA (␮g/d) – 340 440 700 890 900 700 890 900 1000 1300

Table 2 List of top-10 food with the highest bioavailable copper content.

Food

dA

581

cte

580

rre

579

co

578

Environmental Protection Agency. However, the World Health Organization (WHO) interim drinking water guidance level for copper remains at 2 mg/L. As reported by the WHO, the minimal acceptable intake of copper is about 0.9 mg/day, and the recommended dietary allowance (RDA) is 1–1.3 mg/day. The average daily intake per person has been estimated at around roughly 2–3 mg/day (WHO, 1996. Copper. Trace elements in human nutrition and health. World Health Organization, Geneva, pp. 123–43) [77]; WHO, 2004,WHO/SDE/WSH/03.04/88 [78]). The Chicago Health and Aging project evaluated more the 3,700 subjects during a 9-year longitudinal study, through four cognitive tests administered during in-home interviews at 3-year intervals for 6 years, and a dietary assessment was performed with a food frequency questionnaire. The article reports that persons with a median copper intake of 2.75 mg/day, which is associated with a diet rich in saturated and trans fats, had a faster rate of cognitive decline, estimated to be around the equivalent of 19 or more years of age [61]. The Iowa Women’s Health Study showed that the ingestion of inorganic copper in the form of copper supplements was associated with high AD mortality [79]. In fact, most multi-vitamin/mineral dietary supplements contain approximately 2 mg of inorganic copper/pill [10], which further escalates the copper pool in the body. Thus, the intake of copper with dietary supplements usually exceeds the RDA [77, 78]. Moreover, a recent study demonstrated a strong association between the annual mortality of AD and copper concentration in the soil of mainland China [80]. This suggests a relation between the soil and the quality of the water, as well the product of agriculture in those regions. Finally, the Tolerable Upper Intake Level of copper has been set at 10 mg/day, and some foods easily exceed this amount (Tables 1 and 2; Food and Nutrition Board of the Institute of Medicine, The National Academies, 2001; information available at http://www.dsld.nlm.nih.gov/dsld/docs/Dietary Refer ence Intakes Recommended Intakes for Individuals. pdf).

Un

577

A. Pal et al. / Non-Cp-Cu in AD

uth

8

Animal liver (veal) Oysters Sesame seeds Cocoa Nuts (cashew) Seafood (Calamari) Sunflower seeds Sun dried tomatoes Pumpkin Dried herbs (basil)

Copper content (mg)/100 g serving 15 mg 1–8 mg 4.1 mg 3.8 mg 2.2 mg 2.1 mg 1.8 mg 1.4 mg 1.4 mg 1.4 mg

the gastrointestinal tract. In healthy individuals, when dietary copper is high and more is absorbed, endogenous excretion increases, protecting against excess accumulation of copper in the body [1]. These mechanisms of protections can be disturbed on a genetic basis. It is well known that Wilson’s disease is caused by mutations in the ATP7B gene. However, genetic make-up on the individual, along with environmental factors (lifestyle), can have some effects on the disease presentation. In this regard, some studies in animal models of Wilson’s disease, along with case reports of this disease, show diverse presentation of the disease, especially within a family sharing the same mutation (better discussed in [5]). Accordingly, the data gleaned about the LEC rats may be called into question. LEC rats have an early and rapid onset of fulminant hepatitis on a high dietary copper regimen. But they survive long-term on a low-copper dietary regimen [81]. Comparing this basic evidence to the complexity and longevity of a human life, it can be

626 627 628 629 630 631 632 633 634 635 636 637 638 639 640 641 642 643 644 645

A. Pal et al. / Non-Cp-Cu in AD

647 648 649

appreciated the burden of the environmental factors (copper in the diet, comprehending the inorganic copper) in accelerating the onset and progression of a disease that occurs in old age.

sion, or in terms of possible interventions intended to prevent or improve conditions associated with AD risk.

roo f

646

9

ACKNOWLEDGMENTS

657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694

or P

656

uth

655

dA

654

AP and RP are thankful to the Indian Council of Medical Research (ICMR, New Delhi) for the financial assistance as JRF/SRF to Dr. Amit Pal [3/1/3/JRF2009/HRD-13 (11279)]. RS thanks the National Research Council “Aging Program 2012-2014”; MIUR Cod. 1182 /Ric/V o prot. 2010SH7H3F ‘Functional connectivity and neuroplasticity in physiological and pathological aging [ConnAge]; FISM – Fondazione Italiana Sclerosi Multipla – Cod.2011/R/32 Fatigue Relief in Multiple Sclerosis by transcranial Direct Current Stimulation (tDCS): can we Differentiate stimulation Targets within the primary sensorimotor cortices? [FaReMuS DiCDiT]; Ministry of Health Cod. GR-2008-1138642 Promoting recovery from Stroke: Individually enriched therapeutic intervention in Acute phase [ProSIA]; Ricerca Corrente, Italian Ministry of Health; Canox4drug SpA 2013-2016 ‘Non-Ceruloplasmin copper in Alzheimer’s disease’ (Prot. 30/2013). Authors apologize to any colleagues whose work was not cited in this review because of length restrictions. Authors’ disclosures available online (http://www.jalz.com/disclosures/view.php?id=2528).

cte

653

rre

652

In vitro and animal models demonstrated that copper acts as a causative factor in A␤ toxicity, and numerous human studies on living patients showed the association of copper with the clinical picture of AD, in terms of cognitive performances, cerebrospinal fluid markers of AD, and clinical worsening in a percentage of AD patients. Moreover, recent genetic data, along with meta-analyses sustain a systemic deregulation of copper in AD. This finds support in a local decrease of total copper in the brain, as is also exemplified in model of Wilson’s disease. This evidence demonstrates the existence of a subtype of AD, which can be typified by abnormal levels of serum Non-Cp-Cu. Higher levels of copper in the diet, primarily if they are taken in in the form of inorganic copper (vitamin/mineral supplements), together with the consumption of a high fat diet, in people with peculiar genetic variants (APOE4, APP, ATP7B) appear to be linked with an increased risk of AD. These concepts are dire, if one considers that, for example, 100 grams of liver contains 14 mg of copper (Table 2). Even though endogenous mechanisms of copper balance protect against excess accumulation of copper in the body [1], these mechanisms can be disturbed in AD and have a relevance in consideration of the fact that MCI subjects with abnormal serum NonCp-Cu levels had a threefold increased hazard of the conversion rate to AD [64]. The outlined findings discussed in this review suggest that modifications in drinking water/dietary supplements, as well as diet in those subjects with an ascertained copper dysfunction can be a safe and inexpensive strategy to prevent or delay the onset of neurodegeneration/AD [5, 82]. However, the research community is still in a dilemma over the minimum safe levels of copper in drinking water or vitamin/mineral supplements. Even though RDA data have been released, it is obvious that they are routinely overlooked by many when planning their diets [77]. More research in healthy/AD animal models is warranted for further insights into the pathophysiological mechanisms of copper toxicity [83]. Research on humans should also be conducted, either in terms of testing the efficacy of treatments that can reestablish normal copper physiology to halting or delaying AD progres-

co

651

SUMMARY AND CONCLUSIONS

Un

650

REFERENCES [1] [2]

[3]

[4]

[5]

[6]

[7]

Turnlund JR (1998) Human whole-body copper metabolism. Am J Clin Nutr 67, 960S-964S. Squitti R (2014) Copper subtype of Alzheimer’s disease (AD): Meta-analyses, genetic studies and predictive value of non-ceruloplasmim copper in mild cognitive impairment conversion to full AD. J Trace Elem Med Biol, doi: 10.1016/j.jtemb.2014.06.018 Squitti R, Polimanti R (2012) Copper hypothesis in the missing hereditability of sporadic Alzheimer’s disease: ATP7B gene as potential harbor of rare variants. J Alzheimers Dis 29, 493-501. Squitti R, Polimanti R (2013) Copper phenotype in Alzheimer’s disease: Dissecting the pathway. Am J Neurodegener Dis 2, 46-56. Squitti R, Siotto M, Polimanti R (2014) Low-copper diet as a preventive strategy for Alzheimer’s disease. Neurobiol Aging 35(Suppl 2), S40-S50. Crisponi G, Nurchi VM, Fanni D, Gerosa C, Nemolato S, Faa G (2010) Copper-related diseases: From chemistry to molecular pathology. Coord Chem Rev 254, 876-889. Madsen E, Gitlin JD (2007) Copper and iron disorders of the brain. Annu Rev Neurosci 30, 317-337.

695 696

697

698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722

723

724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745

752

[10]

753 754 755 756

[11]

757 758

[12]

759 760 761 762

[13]

763 764 765 766

[14]

767 768 769 770

[15]

771 772 773

[16]

774 775

[17]

776 777 778

[18]

779 780 781 782

[19]

783 784 785

[20]

786 787 788

[21]

789 790 791

[22]

792 793 794

[23]

795 796

[24]

797 798 799

[25]

800 801 802 803 804

[26]

805 806 807 808 809 810

[27]

[29]

[30]

[31]

[32]

roo f

751

cerebrospinal fluid of Alzheimer’s disease patients. Biometals 21, 367-372. Squitti R, Quattrocchi CC, Salustri C, Rossini PM (2008) Ceruloplasmin fragmentation is implicated in ‘free’ copper deregulation of Alzheimer’s disease. Prion 2, 23-27. Amtage F, Birnbaum D, Reinhard T, Niesen W, Weiller C, Mader I, Meyer P, Rijntjes M (2014) Estrogen intake and copper depositions: Implications for Alzheimer’s disease? Case Rep Neurol 6, 181-187. Atwood CS, Moir RD, Huang X, Scarpa RC, Bacarra NM, Romano DM, Hartshorn MA, Tanzi RE, Bush AI (1998) Dramatic aggregation of Alzheimer abeta by Cu(II) is induced by conditions representing physiological acidosis. J Biol Chem 273, 12817-12826. Huang HC, Jiang ZF. Amyloid-beta protein precursor family members: A review from homology to biological function. J Alzheimers Dis 26, 607-626. Huang X, Atwood CS, Hartshorn MA, Multhaup G, Goldstein LE, Scarpa RC, Cuajungco MP, Gray DN, Lim J, Moir RD, Tanzi RE, Bush AI (1999) The A beta peptide of Alzheimer’s disease directly produces hydrogen peroxide through metal ion reduction. Biochemistry 38, 7609-7616. Huang X, Cuajungco MP, Atwood CS, Hartshorn MA, Tyndall JD, Hanson GR, Stokes KC, Leopold M, Multhaup G, Goldstein LE, Scarpa RC, Saunders AJ, Lim J, Moir RD, Glabe C, Bowden EF, Masters CL, Fairlie DP, Tanzi RE, Bush AI (1999) Cu(II) potentiation of alzheimer abeta neurotoxicity. Correlation with cell-free hydrogen peroxide production and metal reduction. J Biol Chem 274, 37111-37116. White AR, Multhaup G, Maher F, Bellingham S, Camakaris J, Zheng H, Bush AI, Beyreuther K, Masters CL, Cappai R (1999) The Alzheimer’s disease amyloid precursor protein modulates copper-induced toxicity and oxidative stress in primary neuronal cultures. J Neurosci 19, 9170-9179. White AR, Reyes R, Mercer JF, Camakaris J, Zheng H, Bush AI, Multhaup G, Beyreuther K, Masters CL, Cappai R (1999) Copper levels are increased in the cerebral cortex and liver of APP and APLP2 knockout mice. Brain Res 842, 439-444. Cherny RA, Legg JT, McLean CA, Fairlie DP, Huang X, Atwood CS, Beyreuther K, Tanzi RE, Masters CL, Bush AI (1999) Aqueous dissolution of Alzheimer’s disease Abeta amyloid deposits by biometal depletion. J Biol Chem 274, 23223-23228. 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, 11065-11069. Nelson TJ, Alkon DL (2005) Oxidation of cholesterol by amyloid precursor protein and beta-amyloid peptide. J Biol Chem 280, 7377-7387. Sparks DL, Friedland R, Petanceska S, Schreurs BG, Shi J, Perry G, Smith MA, Sharma A, Derosa S, Ziolkowski C, Stankovic G (2006) Trace copper levels in the drinking water, but not zinc or aluminum influence CNS Alzheimerlike pathology. J Nutr Health Aging 10, 247-254. Lu J, Zheng YL, Wu DM, Sun DX, Shan Q, Fan SH (2006) Trace amounts of copper induce neurotoxicity in the cholesterol-fed mice through apoptosis. FEBS Lett 580, 67306740. Mao X, Ye J, Zhou S, Pi R, Dou J, Zang L, Chen X, Chao X, Li W, Liu M, Liu P (2012) The effects of chronic copper exposure on the amyloid protein metabolisim associated genes’ expression in chronic cerebral hypoperfused rats. Neurosci Lett 518, 14-18.

or P

750

[28]

[33]

dA

[9]

cte

749

rre

748

Cox DW, Moore SD (2002) Copper transporting P-type ATPases and human disease. J Bioenerg Biomembr 34, 333338. Mehta R, Templeton DM, O’Brien PJ (2006) Mitochondrial involvement in genetically determined transition metal toxicity II. Copper toxicity. Chem Biol Interact 163, 77-85. Georgopoulos PG, Roy A, Yonone-Lioy MJ, Opiekun RE, Lioy PJ (2001) Environmental copper: Its dynamics and human exposure issues. J Toxicol Environ Health B Crit Rev 4, 341-394. Sayre LM, Perry G, Smith MA (1999) Redox metals and neurodegenerative disease. Curr Opin Chem Biol 3, 220-225. Prasad R, Kaur G, Nath R, Walia BN (1996) Molecular basis of pathophysiology of Indian childhood cirrhosis: Role of nuclear copper accumulation in liver. Mol Cell Biochem 156, 25-30. Braughler JM, Hall ED (1989) Central nervous system trauma and stroke. I. Biochemical considerations for oxygen radical formation and lipid peroxidation. Free Radic Biol Med 6, 289301. Carri MT, Ferri A, Cozzolino M, Calabrese L, Rotilio G (2003) Neurodegeneration in amyotrophic lateral sclerosis: The role of oxidative stress and altered homeostasis of metals. Brain Res Bull 61, 365-374. Ozcelik D, Uzun H (2009) Copper intoxication; antioxidant defenses and oxidative damage in rat brain. Biol Trace Elem Res 127, 45-52. Desai V, Kaler SG (2008) Role of copper in human neurological disorders. Am J Clin Nutr 88, 855S-858S. Zheng W, Monnot AD (2012) Regulation of brain iron and copper homeostasis by brain barrier systems: Implication in neurodegenerative diseases. Pharmacol Ther 133, 177-188. Kozlowski H, Luczkowski M, Remelli M, Valensin D. Copper, zinc and iron in neurodegenerative diseases (Alzheimer’s, Parkinson’s and prion diseases). Coord Chem Rev 256, 21292141. Jomova K, Vondrakova D, Lawson M, Valko M (2010) Metals, oxidative stress and neurodegenerative disorders. Mol Cell Biochem 345, 91-104. Tiffany-Castiglioni E, Hong S, Qian Y (2011) Copper handling by astrocytes: Insights into neurodegenerative diseases. Int J Dev Neurosci 29, 811-818. Roberts EA, Schilsky ML, American Association for Study of Liver D (2008) Diagnosis and treatment of Wilson disease: An update. Hepatology 47, 2089-2111. Ballatore C, Lee VM, Trojanowski JQ (2007) Tau-mediated neurodegeneration in Alzheimer’s disease and related disorders. Nat Rev Neurosci 8, 663-672. LaFerla FM, Green KN, Oddo S (2007) Intracellular amyloidbeta in Alzheimer’s disease. Nat Rev Neurosci 8, 499-509. Brewer GJ (2009) The risks of copper toxicity contributing to cognitive decline in the aging population and to Alzheimer’s disease. J Am Coll Nutr 28, 238-242. Squitti R, Barbati G, Rossi L, Ventriglia M, Dal Forno G, Cesaretti S, Moffa F, Caridi I, Cassetta E, Pasqualetti P, Calabrese L, Lupoi D, Rossini PM (2006) Excess of nonceruloplasmin serum copper in AD correlates with MMSE, CSF [beta]-amyloid, and h-tau. Neurology 67, 76-82. Squitti R, Bressi F, Pasqualetti P, Bonomini C, Ghidoni R, Binetti G, Cassetta E, Moffa F, Ventriglia M, Vernieri F, Rossini PM (2009) Longitudinal prognostic value of serum “free” copper in patients with Alzheimer disease. Neurology 72, 50-55. Capo CR, Arciello M, Squitti R, Cassetta E, Rossini PM, Calabrese L, Rossi L (2008) Features of ceruloplasmin in the

co

[8]

747

Un

746

A. Pal et al. / Non-Cp-Cu in AD

uth

10

[34]

[35]

[36]

[37]

[38]

[39]

[40]

[41]

811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875

A. Pal et al. / Non-Cp-Cu in AD

883 884

[44]

885 886 887

[45]

888 889 890 891

[46]

892 893 894 895 896

[47]

897 898 899 900

[48]

901 902 903

[49]

904 905 906 907 908

[50]

909 910 911 912 913

[51]

914 915 916 917 918

[52]

919 920 921 922 923

[53]

924 925 926 927

[54]

928 929 930 931

[55]

932 933 934 935

[56]

936 937 938 939 940

[57]

[59]

[60]

[61]

[62]

[63]

roo f

882

or P

881

uth

880

in Alzheimer’s disease and mild cognitive impairment: A meta-analysis. Neurobiol Dis 59, 100-110. Morris MC, Evans DA, Tangney CC, Bienias JL, Schneider JA, Wilson RS, Scherr PA (2006) Dietary copper and high saturated and trans fat intakes associated with cognitive decline. Arch Neurol 63, 1085-1088. Jacobs D, Oxley DK, DeMott WR (2002) Laboratory Test Handbook: Concise with Disease Index, 2nd ed., Lexi-Comp, Hudson, OH. Walshe JM, Clinical Investigations Standing Committee of the Association of Clinical B (2003) Wilson’s disease: The importance of measuring serum caeruloplasmin nonimmunologically. Ann Clin Biochem 40, 115-121. Squitti R, Pasqualetti P, Dal Forno G, Moffa F, Cassetta E, Lupoi D, Vernieri F, Rossi L, Baldassini M, Rossini PM (2005) Excess of serum copper not related to ceruloplasmin in Alzheimer disease. Neurology 64, 1040-1046. Squitti R, Lupoi D, Pasqualetti P, Dal Forno G, Vernieri F, Chiovenda P, Rossi L, Cortesi M, Cassetta E, Rossini PM (2002) Elevation of serum copper levels in Alzheimer’s disease. Neurology 59, 1153-1161. Squitti R, Ghidoni R, Scrascia F, Benussi L, Panetta V, Pasqualetti P, Moffa F, Bernardini S, Ventriglia M, Binetti G, Rossini PM (2011) Free copper distinguishes mild cognitive impairment subjects from healthy elderly individuals. J Alzheimers Dis 23, 239-248. Squitti R, Ghidoni R, Siotto M, Ventriglia M, Benussi L, Paterlini A, Magri M, Binetti G, Cassetta E, Caprara D, Vernieri F, Rossini PM, Pasqualetti P (2014) Value of serum nonceruloplasmin copper for prediction of mild cognitive impairment conversion to Alzheimer disease. Ann Neurol 75, 574-580. Murray ME, Graff-Radford NR, Ross OA, Petersen RC, Duara R, Dickson DW (2011) Neuropathologically defined subtypes of Alzheimer’s disease with distinct clinical characteristics: A retrospective study. Lancet Neurol 10, 785-796. Bucossi S, Mariani S, Ventriglia M, Polimanti R, Gennarelli M, Bonvicini C, Pasqualetti P, Scrascia F, Migliore S, Vernieri F, Rossini PM, Squitti R (2011) Association between the c. 2495 A>G ATP7B polymorphism and sporadic Alzheimer’s disease. Int J Alzheimers Di 2011, 973692s. Bucossi S, Polimanti R, Mariani S, Ventriglia M, Bonvicini C, Migliore S, Manfellotto D, Salustri C, Vernieri F, Rossini PM, Squitti R (2012) Association of K832R and R952K SNPs of Wilson’s disease gene with Alzheimer’s disease. J Alzheimers Dis 29, 913-919. Bucossi S, Polimanti R, Ventriglia M, Mariani S, Siotto M, Ursini F, Trotta L, Scrascia F, Callea A, Vernieri F, Squitti R (2013) Intronic rs2147363 variant in ATP7B transcription factor-binding site associated with Alzheimer’s disease. J Alzheimers Dis 37, 453-459. Squitti R, Polimanti R, Bucossi S, Ventriglia M, Mariani S, Manfellotto D, Vernieri F, Cassetta E, Ursini F, Rossini PM (2013) Linkage disequilibrium and haplotype analysis of the ATP7B gene in Alzheimer’s disease. Rejuvenation Res 16, 3-10. Squitti R, Polimanti R, Siotto M, Bucossi S, Ventriglia M, Mariani S, Vernieri F, Scrascia F, Trotta L, Rossini PM (2013) ATP7B variants as modulators of copper dyshomeostasis in Alzheimer’s disease. Neuromolecular Med 15, 515-522. Bucossi S, Ventriglia M, Panetta V, Salustri C, Pasqualetti P, Mariani S, Siotto M, Rossini PM, Squitti R. Copper in Alzheimer’s disease: A meta-analysis of serum,plasma, and cerebrospinal fluid studies. J Alzheimers Dis 24, 175-185. Schrag M, Mueller C, Zabel M, Crofton A, Kirsch WM, Ghribi O, Squitti R, Perry G. Oxidative stress in blood in

dA

[43]

[58]

[64]

[65]

cte

879

rre

878

Brewer GJ (2008) The risks of free copper in the body and the development of useful anticopper drugs. Curr Opin Clin Nutr Metab Care 11, 727-732. Singh I, Sagare AP, Coma M, Perlmutter D, Gelein R, Bell RD, Deane RJ, Zhong E, Parisi M, Ciszewski J, Kasper RT, Deane R (2013) Low levels of copper disrupt brain amyloidbeta homeostasis by altering its production and clearance. Proc Natl Acad Sci U S A 110, 14771-14776. Brewer GJ (2011) Issues raised involving the copper hypotheses in the causation of Alzheimer’s disease. Int J Alzheimers Di 2011, 537528. Multhaup G, Schlicksupp A, Hesse L, Beher D, Ruppert T, Masters CL, Beyreuther K (1996) The amyloid precursor protein of Alzheimer’s disease in the reduction of copper(II) to copper(I). Science 271, 1406-1409. White AR, Multhaup G, Galatis D, McKinstry WJ, Parker MW, Pipkorn R, Beyreuther K, Masters CL, Cappai R (2002) Contrasting, species-dependent modulation of coppermediated neurotoxicity by the Alzheimer’s disease amyloid precursor protein. J Neurosci 22, 365-376. Parthasarathy S, Yoo B, McElheny D, Tay W, Ishii Y (2014) Capturing a reactive state of amyloid aggregates: NMR-based characterization of copper-bound Alzheimer disease amyloid beta-fibrils in a redox cycle. J Biol Chem 289, 9998-10010. Lovell MA, Robertson JD, Teesdale WJ, Campbell JL, Markesbery WR (1998) Copper, iron and zinc in Alzheimer’s disease senile plaques. J Neurol Sci 158, 47-52. Schrag M, Mueller C, Oyoyo U, Smith MA, Kirsch WM. Iron, zinc and copper in the Alzheimer’s disease brain: A quantitative meta-analysis. Some insight on the influence of citation bias on scientific opinion. Prog Neurobiol 94, 296306. Graham SF, Nasaruddin MB, Carey M, Holscher C, McGuinness B, Kehoe PG, Love S, Passmore P, Elliott CT, Meharg AA, Green BD (2014) Age-associated changes of brain copper, iron, and zinc in Alzheimer’s disease and dementia with Lewy bodies. J Alzheimers Dis, doi: 10.3233/JAD-140684 Rembach A, Hare DJ, Lind M, Fowler CJ, Cherny RA, McLean C, Bush AI, Masters CL, Roberts BR (2013) Decreased copper in Alzheimer’s disease brain is predominantly in the soluble extractable fraction. Int J Alzheimers Di 2013, 623241. Fujiwara N, Iso H, Kitanaka N, Kitanaka J, Eguchi H, Ookawara T, Ozawa K, Shimoda S, Yoshihara D, Takemura M, Suzuki K (2006) Effects of copper metabolism on neurological functions in Wistar and Wilson’s disease model rats. Biochem Biophys Res Commun 349, 1079-1086. James SA, Volitakis I, Adlard PA, Duce JA, Masters CL, Cherny RA, Bush AI (2012) Elevated labile Cu is associated with oxidative pathology in Alzheimer disease. Free Radic Biol Med 52, 298-302. Arnal N, Cristalli DO, de Alaniz MJ, Marra CA. Clinical utility of copper, ceruloplasmin, and metallothionein plasma determinations in human neurodegenerative patients and their first-degree relatives. Brain Res 1319, 118-130. Lopez N, Tormo C, De Blas I, Llinares I, Alom J (2013) Oxidative stress in Alzheimer’s disease and mild cognitive impairment with high sensitivity and specificity. J Alzheimers Dis 33, 823-829. Squitti R, Simonelli I, Ventriglia M, Siotto M, Pasqualetti P, Rembach A, Doecke J, Bush AI (2014) Meta-analysis of serum non-ceruloplasmin copper in Alzheimer’s disease. J Alzheimers Dis 38, 809-822. Schrag M, Mueller C, Zabel M, Crofton A, Kirsch WM, Ghribi O, Squitti R, Perry G (2013) Oxidative stress in blood

co

[42]

877

Un

876

[66]

[67]

[68]

[69]

[70]

[71]

[72]

11

941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005

[74]

1012 1013 1014 1015

[75]

1016 1017 1018 1019 1020

[76]

1021 1022 1023 1024 1025

[77]

1026 1027

[78]

[81]

[82]

[83]

roo f

1011

[80]

Mursu J, Robien K, Harnack LJ, Park K, Jacobs DR Jr (2011) Dietary supplements and mortality rate in older women: The Iowa Women’s Health Study. Arch Intern Med 171, 16251633. Shen XL, Yu JH, Zhang DF, Xie JX, Jiang H (2014) Positive relationship between mortality from Alzheimer’s disease and soil metal concentration in mainland China. J Alzheimers Dis, doi: 10.3233/JAD-140153. Siaj R, Sauer V, Stoppeler S, Spiegel HU, Kohler G, Zibert A, Schmidt HH (2012) Dietary copper triggers onset of fulminant hepatitis in the Long-Evans cinnamon rat model. World J Gastroenterol 18, 5542-5550. Barnard ND, Bush AI, Ceccarelli A, Cooper J, de Jager CA, Erickson KI, Fraser G, Kesler S, Levin SM, Lucey B, Morris MC, Squitti R (2014) Dietary and lifestyle guidelines for the prevention of Alzheimer’s disease. Neurobiol Aging 35S2, S74-S78. Pal A, Kumar A, Prasad R (2014) Predictive association of copper metabolism proteins with Alzheimer’s disease and Parkinson’s disease: A preliminary perspective. Biometals 27, 25-31.

or P

1010

[79]

dA

1009

cte

[73]

rre

1008

Alzheimer’s disease and mild cognitive impairment: A metaanalysis. Neurobiol Dis 59, 100-110. Ventriglia M, Bucossi S, Panetta V, Squitti R. Copper in Alzheimer’s disease: A meta-analysis of serum, plasma, and cerebrospinal fluid studies. J Alzheimers Dis 30, 981-984. Salustri C, Barbati G, Ghidoni R, Quintiliani L, Ciappina S, Binetti G, Squitti R (2010) Is cognitive function linked to serum free copper levels? A cohort study in a normal population. Clin Neurophysiol 121, 502-507. Liu HP, Lin WY, Wang WF, Tsai CH, Wu WC, Chiou MT, Shen CP, Wu BT, Tsai FJ (2013) Genetic variability in coppertransporting P-type adenosine triphosphatase (ATP7B) is associated with Alzheimer’s disease in a Chinese population. J Biol Regul Homeost Agents 27, 319-327. Ferri CP, Prince M, Brayne C, Brodaty H, Fratiglioni L, Ganguli M, Hall K, Hasegawa K, Hendrie H, Huang Y, Jorm A, Mathers C, Menezes PR, Rimmer E, Scazufca M, Alzheimer’s Disease I (2005) Global prevalence of dementia: A Delphi consensus study. Lancet 366, 2112-2117. WHO (1996) Copper. In Trace Elements in Human Nutrition and Health. World Health Organization, Geneva, pp. 123-143. WHO (2004) Copper. In Copper in Drinking-water. World Health Organization, Geneva, pp. 1-23.

co

1007

Un

1006

A. Pal et al. / Non-Cp-Cu in AD

uth

12

1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048

Towards a unified vision of copper involvement in Alzheimer's disease: a review connecting basic, experimental, and clinical research.

Copper is an essential micronutrient for physiological cell functioning and central nervous system (CNS) development. Indeed, it is a cofactor of many...
285KB Sizes 0 Downloads 5 Views