You are on page 1of 7

Biochimica et Biophysica Acta 1822 (2012) 657–663

Contents lists available at SciVerse ScienceDirect

Biochimica et Biophysica Acta


journal homepage: www.elsevier.com/locate/bbadis

Review

Antioxidants in Down syndrome☆


Ira T. Lott ⁎
Department of Pediatrics and Neurology, School of Medicine, University of California Irvine (UCI), Orange, CA 92868, USA

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

Article history: Individuals with Down syndrome (DS) have high levels of oxidative stress throughout the lifespan. Mouse
Received 31 October 2011 models of DS share some structural and functional abnormalities that parallel findings seen in the human
Received in revised form 13 December 2011 phenotype. Several of the mouse models show evidence of cellular oxidative stress and have provided a plat-
Accepted 14 December 2011
form for antioxidant intervention. Genes that are overexpressed on chromosome 21 are associated with
Available online 21 December 2011
oxidative stress and neuronal apoptosis. The lack of balance in the metabolism of free radicals generated dur-
Keywords:
ing processes related to oxidative stress may have a direct role in producing the neuropathology of DS includ-
Antioxidant ing the tendency to Alzheimer disease (AD). Mitochondria are often a target for oxidative stress and are
Oxidative stress considered to be a trigger for the onset of the AD process in DS. Biomarkers for oxidative stress have been
Down syndrome described in DS and in AD in the general population. However, intervention trials using standard antioxidant
Alzheimer disease supplements or diets have failed to produce uniform therapeutic effect. This chapter will examine the biolog-
Antioxidant clinical trial ical role of oxidative stress in DS and its relationship to abnormalities in both development and aging within
Mitochondrial disorder the disorder. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.
© 2011 Elsevier B.V. All rights reserved.

1. Introduction publications were not excluded if considered relevant to the topics.


The reference list of articles identified by this search was selected for
Oxidative stress is part of the fundamental biology of Down syn- relevance.
drome (DS). As the most common genetic cause of intellectual disabili-
ty, DS is due to trisomy of all or part of the chromosome 21. With few
exceptions, all of the genes of known function are located on the long 2. Mouse models of DS and oxidative stress
arm of chromosome 21. The gene sequence of chromosome 21 has
been carried out with 99.7% coverage [1]. Among the more than 500 Mouse models have been used, in part, to determine how the
genes on chromosome 21, functional information exists on about 150 genes on human chromosome 21 lead to the tissue-wide abnormali-
genes. Of these, there are 18 that effect gene expression and over 20 ties seen in DS [6]. There is 80% synteny between human chromo-
that effect metabolic functioning [2]. Chromosome 21 contains several some 21 (Hsa 21) and mouse chromosome 16 meaning that specific
genes that have been implicated in oxidative stress related to neurode- genes are localized on the same chromosome in both the human
generation including Cu/Zn superoxide dismutase (SOD1), amyloid and mouse species. Approximately 175 genes are highly conserved
precursor protein (APP), Ets-2 transcription factors, Down Syndrome between Hsa 21 and the mouse genome although the correspondence
Critical Region 1 (DSCR1) stress-inducible factor, beta-site APP cleaving varies with the different models [7]. The mouse strains labeled
enzyme (BACE) and S100 [3–5]. In this chapter, investigations underly- Ts65Dn, Ts1Yey, Ts1Cje, and Ts1Rh are trisomic for the segments of
ing oxidative stress in DS will be discussed with an emphasis on those mouse chromosome 16 that are homologous to Hsa 21. The relation-
which have led to therapeutic intervention trials. The scope of the review ship of these models to one another and to genes on mouse chromo-
included a search of PUBMED, MEDLINE AND APAPsychNET Direct some 16 is shown in Fig. 1 [8]. The most commonly studied mouse
(1970–2011). The term “Down Syndrome” was used in combination model is Ts65Dn which shares a number of structural and functional
with the appropriate review target in the individual sections of this neurological similarities with individuals who have DS. These include
report (i.e. mouse models, dementia, clinical trials, etc.). A principle abnormalities in basal forebrain cholinergic neurons and synapses,
focus was on selected publications over the past 5 years, but older reduction of cell density in the hippocampus of aged mice, reduced
dendritic branching in layer III pyramidal cells, and abnormalities
in the ability to induce long-term potentiation [9–12]. The Ts65Dn
mice also show spatial and non-spatial learning disabilities [13]. The
☆ This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treat-
advantages of using mouse models for understanding various abnor-
ment in Disease.
⁎ UC Irvine Medical Center, Department of Pediatrics ZC 4482, 101 The City Dr.,
malities in DS such as oxidative stress are numerous and include the
Orange, CA 92868, USA. Tel.: + 1 714 456 5333; fax: + 1 714 456 8466. ability to study a sufficient number of animals in a short time, genetic
E-mail address: itlott@uci.edu. engineering for individual genes or chromosomal regions, control for

0925-4439/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.bbadis.2011.12.010
658 I.T. Lott / Biochimica et Biophysica Acta 1822 (2012) 657–663

Mouse syntenic regions


The lack of balance in the metabolism of ROS appears to have a di-
rect role in producing the neuropathology of DS [23–25]. Oxidative
damage has been observed in brain tissue from individuals with DS
[26,27]. In the brain of fetuses with DS, two forms of glycation end
products indicative of cellular oxidative stress were increased [28].
Cortical neurons from fetuses with DS show intracellular accumula-
tion of ROS that lead to neuronal apoptosis [29]. These findings sug-
gest that glycoxidation occurs very early in the life of individuals
with DS. Early deposition of oxidized Aβ has also been seen within
MMU16 plaques from a 31 year old individual with DS, a finding which sug-
gests that oxidative modification of Aβ may be an early event in the
pathogenesis of AD [30]. Oxidative stress is also manifested in the
brain of patients with AD in the general population [31,32]. Cultured
human cortical neurons have provided a model in which oxidative
stress may be treated in DS. Peptides released from naturally occur-
ring glial proteins have been shown to possess potent antioxidative
MMU17 effects in cultured DS cortical neurons [33].

MMU10
4. Genes encoded on chromosome 21 and their relationship to
oxidative stress
Fig. 1. Schematic representation of mouse models of trisomy 21. The first bar on the left
represents mouse chromosomal regions (MMU10, MMU16, and MMU17) that are syn- SOD1 has been shown to be a part of many physiological processes
tenic to human chromosome 21 (Hsa21). This bar is proportional to the number of such as transcription regulation, protein activation, bioenergy output,
genes carried by the fragment. The approximate mapping of the genes is shown on
and cell proliferation. Simultaneous disorders of SOD1 and the uncou-
the left of the bar. All, except one, models are syntenic to MMU16. The estimated
number of syntenic genes is indicated on the right of each bar for each mouse model. pling of proteins related to mitochondrial dysfunction have been im-
Reprinted from American Journal of Medical Genetics Part C (Seminars in Medical plicated in the pathogenesis of AD and may precede Aβ deposition
Genetics), 154C, Roubertoux P and Carlier M, Mouse Models of Cognitive Disabilities [34,35]. SOD1 gene is located just above the DS minimal critical region
in Trisomy 21 (Down Syndrome), 400–416, 2010, with permission from John Wiley and more than 99% are trisomic for this gene [36]. The Down syn-
and Sons.
drome critical region is located on the long arm (q) of chromosome
21 and is considered to be responsible for some, if not all, of the fea-
genetic background, low cost of screening, ability to study both devel- tures of the DS phenotype.
oping and mature subjects, modeling therapeutic interventions and This cytosolic enzyme catalyzes the dismutation of superoxide
the lack of postmortem delays [6,14]. radicals that are formed during oxidative metabolism to hydrogen
Oxidative stress represents an imbalance between the production peroxide and oxygen. Under normal circumstances, the hydrogen
of reactive oxygen species (ROS) and the ability of a biological system peroxide is removed by glutathione peroxidase or catalase. However,
to detoxify the reactive intermediates or to repair the resulting brain levels of both of these enzymes maybe low in mammalian
damage. Toxic effects may be caused through the production of lipid species causing an imbalance between the first and second step in
peroxidation products and these compounds are increased in the superoxide catabolism which then results in an increase in hydroxyl
brains of two mouse models of DS called Ts1Cje and Ts2Cje [15]. radicals [37]. Hydroxyl radicals are highly reactive compounds that
The TsiCje model excludes APP and SOD1 yet shows evidence of oxi- can damage neuronal membranes, cellular structures and organelles
dative stress as characterized by decreased mitochondrial membrane [38]. In DS, the exact role played by SOD1 overexpression is not
potential and other biomarkers [16]. In Ts65Dn mice, supplementa- clear. For example, congenital heart disease which is seen in up to
tion of the antioxidant α-tocopherol from the embryonic stage on- 50% of children with DS does not appear to be associated with abnor-
ward showed improvement of spatial learning and alertness in the malities in redox metabolism [39]. However, circulating endothelial
treated mice but no amelioration of background hyperactivity [17]. progenitor cells involved in angiogenesis in DS have an increased sus-
There was also improvement in hypocellularity in the dentate gyrus ceptibility to oxidative stress which is thought to be related to the
in the treated mice. In another study, markers of oxidative stress immune disorder associated with this syndrome [40]. Proliferation
were found to be elevated in Ts65Dn brain and treatment with α- defects and gene expression dysregulation seen in neural progenitor
tocopherol was associated with improved performance on spatial cells in a mouse model of DS (Ts1Cje) required neither overexpres-
working memory tasks and an attenuation in cholinergic neuronal sion of SOD1 or APP [41]. In double APP/SOD1 transgenic mice, SOD1
pathology in forebrain [18]. A nootropic drug with antioxidant prop- overexpression prevented toxicity from APP and significantly in-
erties showed mixed results in cognition and behavior of Ts65Dn creased the lifespan of the animals [42]. The relationship of genes
mice [19]. Thus, several treatment approaches to oxidative stress encoded on chromosome 21 to oxidative stress has been previously
have shown some benefit in mouse models of DS. reviewed [25].
Ets-2 is a protein that is encoded by Ets-2 gene and functions as a
3. Biological foundation of oxidative stress in individuals with DS transcription factor that is overexpressed on chromosome 21. Ets-2
has been involved in multiple cellular processes including differenti-
It is the area of Alzheimer disease (AD) that oxidative stress in DS ation, maturation and signaling cascades [43,44]. It appears to be
seems to have the most significance. By 40 years of age, virtually markedly upregulated by oxidative stress. In fetal cortical cells Ets-2
all individuals with DS show the characteristic neuropathological induces a stereotyped pattern of apoptotic changes leading to neuro-
changes of AD including beta-amyloid (Aβ) plaques and neurofibril- nal cell death [4]. In AD, increased expression of Ets-2 is associated
lary tangles [20]. The accumulation of Aβ plaques in brain is seen with pathological markers suggesting that oxidative stress-driven
across the lifespan in DS with appearance by 8 years of age or before Ets-2 upregulation contributes to neuronal death in both DS and AD.
[21]. In most individuals with DS, the rate of Aβ deposition increases In Ts65Dn mouse, the Ets-2 tumor repressor gene has been related
markedly between 35 and 45 years and becomes associated with to a reduced incidence of solid tumors, one of the multiple complex
neurofibrillary tangles [22]. genetic mechanisms that underlie cancer pathogenesis in DS [45,46].
I.T. Lott / Biochimica et Biophysica Acta 1822 (2012) 657–663 659

APP is involved in the development of AD and DS [47,48]. Aβ plasma 5. Mitochondria and oxidative stress in DS
levels are increased in DS and soluble forms of Aβ have been seen down
to 21 weeks of gestational age [49,50]. Oxidized Aβ is seen in a high per- Mitochondria are subcellular organelles that contain enzymes for
centage of diffuse and core plaques within the brains of individuals with ATP production, calcium homeostasis, ROS, and apoptotic signaling
DS and oxidative modification of Aβ may be an early event in plaque [59]. As such, they are a target for oxidative stress. Dysfunction of
biogenesis. Animal exposure studies to Aβ demonstrate that the neuro- mitochondria has surfaced as one of the most discussed hypotheses
biological factors that foster AD may have their origins in early life and for a trigger in AD.
result from interplay between the environment, epigenetic factors and Within the general population and to some extent in DS, the
oxidative stress [51]. Environmental influences occurring during brain strongest genetic association with dementia is the ε-4 allele of the
development inhibit DNA methyltransferases, thus hypomethylating apolipoprotein gene (Apoε4) [60,61]. Cleavage of Apoε4 yields pep-
promoters of genes associated with AD such as APP. This early life im- tides that increase oxidative stress within the mitochondria [62].
printing may act later in life to increase the levels of APP and Aβ. In- Mitochondrial dysfunction has been observed in individuals with
creased Aβ levels may promote the production of ROS, damage DNA, AD as manifested by defects in oxidative phosphorylation specifically
and accelerate neurodegenerative events. However, the resulting pic- in the respiratory complex IV [63,64]. The Aβ peptide itself can
ture is complex since other AD-associated genes may be repressed rath- elevate oxidative stress by entering the mitochondria and producing
er than overexpressed. Thus, hypo- or hypermethylation of AD-specific ROS [65,66]. Mitochondrial dysfunction is one of the earliest events
genes and their subsequent effect on Aβ production may be one of the in the brains of mutant mice overexpressing APP as well as tau and
factors accounting for the variability of the incidence of AD-type presenilin1 [65,67].
dementia in individuals with DS. Somatic mitochondrial DNA mutations may be elevated 15-fold in
DSCR1 is a stress-inducible factor located on chromosome 21 that AD brain [68]. In DS, mitochondrial dysfunction and elevated levels of
is modulated by oxidative stress [52]. DSCRI is also known as Adapt78 oxidative stress have been reported in neuronal cell cultures, brain,
and RCAN (regulator of calcineurin). Transient overexpression of fibroblasts, erythrocytes and urine [69]. While somatic mitochondrial
DSCR1 appears to be protective but chronic upregulation is deleteri- DNA control region mutations accumulate with age in post-mitotic
ous to cells likely through the expression of SOD1 [53]. DSCR1 is also tissues including the brain, the level of these mutations is markedly
upregulated in AD [54]. DSCR1 has been found to be a regulator elevated in the brains of individuals with DS and AD in the general
of the calcineurin signaling pathway related to the reduced risk of population [70]. This increased mutation rate is also seen in peripher-
certain cancers in individuals with DS [55]. al blood and in lymphoblastoid cell lines (Fig. 2). Mitochondrial dys-
DYRK1A is a gene that is overexpressed in individuals with trisomy function in DS is not only seen in the aging-AD process but earlier
21 as well as mouse models for DS. This gene may be responsible for in life as well. In human neural progenitor cells from fetal cortex,
processes affecting synaptic plasticity and memory consolidation. Over- those genes overexpressed on chromosome 21 (such as S100B) dis-
epxression of DYRK1A in the brain of individuals with DS causes neuro- turb the redox state within mitochondria and ultimately impair neu-
fibrillary degeneration via hyperphosphorylation of tau [56]. In aged rogenesis [71]. Human skin fibroblasts from fetuses with DS show
mice overexpressing DYRK1A, marked motor abnormalities were asso- that a complex I deficit may be responsible for the oxidative defect
ciated with cell loss in the CA1–CA3 areas of the hippocampus and in in mitochondria [72]. The mitochondria from DS fibroblasts are par-
the dentate gyrus [57]. Epigallocatechin gallate — a member of a natural ticularly sensitive to okadaic acid, a toxin that may induce malforma-
polyphenols family, is found in great amount in green tea leaves. It ap- tions and interfere with the growth of cell lines [73]. In another study
pears to be a specific and safe DYRK1A inhibitor which rescues the brain evaluating the cause of mitochondrial dysfunction in DS, impairment
defects in transgenic mouse that overexpresses the gene [58]. The rela- of the methyl cycle was shown to effect mitochondrial methyl
tionship of this effect to oxidative stress has not been elucidated. availability and glutathione concentrations [74]. Mitochondrial ab-
In summary, overexpression of several chromosome 21 genes normalities in mouse models for DS have been discussed in the intro-
leads to a genetic imbalance that creates a “vicious cycle” by which ductory section to these models.
oxidative stress is exacerbated resulting in further genetic imbalance. Taken collectively, the findings of mitochondrial DNA mutations
By virtue of their role in oxidative metabolism, mitochondria may in DS and AD may be responsible for dementia in this disorder. Mito-
serve as one of the pathways for this genetic imbalance. chondrial dysfunction also occurs at early age epochs in DS and could

Fig. 2. mtDNA Regulatory Control Region (RCR) mutation frequency in the peripheral tissue samples from control and dementia patients. (A) Analysis of mtDNA RCR mutation
frequency in the frontal cortex and serum DNA (CF-DNA) from the same AD and control cases (n = 6, between the ages of 81 to 95 years). (B) Analysis of lymphoblastoid
cell lines (LCLs) from DS, DS and AD, AD, and controls (n = 5–7, between the ages of 45 to 62 years). Reprinted from Journal of Alzheimer’s Disease, 20, Coskun P, Wyrembaka
J, Derbereva O, Melkonian G, Doran E, Lott IT, Head E, Cotman CW, and Wallace D, Systemic Mitochondrial Dysfunction and the Etiology of Alzheimer’s Disease and Down Syndrome
Dementia, S293-S310, 2010, with permission from IOS press.
660 I.T. Lott / Biochimica et Biophysica Acta 1822 (2012) 657–663

provide a platform for early effects of oxidative stress. Measures of predict oxidative changes in the brain has not yet been established
mitochondrial overproliferation and deletion have been proposed conclusively. Peripheral markers carry a promise to assist in the
as a marker for AD [75]. Mitochondrial deficits have been noted in evaluation of oxidative risk throughout the lifespan with potential in-
non-neural peripheral tissues and there are a number of studies tervention through antioxidant trials. In evaluating the reliability
that have tried to capture peripheral effects of oxidative stress. of peripheral markers, of particular importance is the validation of
assays, determination of the steady state concentrations of the bio-
6. Biomarkers of oxidative stress in individuals with DS markers, utilization of multianalyte panels, and correlation with in
vivo imaging findings [88]. The relationship of peripheral marker to
In a study aimed at defining the relationship between peripheral cerebrospinal fluid (CSF) concentrations has been rarely studied, a
markers of oxidative stress and cognitive ability in adults with DS, point for consideration in future studies since CSF markers may
low ratios of SOD1 to glutathione peroxidase were associated with more closely reflect brain function than plasma. However, even with
worse memory ability, a finding which held after adjustment for CSF markers, lower levels of cytokines may reflect influx from plasma
gender, age, and the use of nutritional supplements. However, there rather than intrathecal synthesis [89].
was no relationship of the above findings to measures of F-
isoprostanes, a proxy for lipid peroxidation [76]. The authors con- 7. Antioxidant trials in the general AD population
cluded that relationship between antioxidant capacity and cognitive
phenotype in DS is complex and could be affected by the interaction In the general population, studies assessing whether the consump-
of several genes rather than a formulation of a simple association. tion of antioxidants from both dietary and supplemental sources in
Individuals with DS often have high uric acid concentration, a me- preventing AD have shown mixed results [90]. In the Chicago Health
tabolite that may have antioxidant properties. For example, children and Aging Project, increased vitamin E intake from dietary sources re-
with DS had higher level of uric acid and total antioxidant capacity duced the risk for dementia in individuals who were positive for
than the control group whereas no differences were found in adults Apoε4 allele. However, actual supplements of vitamin E were not
[77]. However, in another study of children with DS, increased uric associated with benefits. Vitamin E administration did not influence
acid concentrations were associated with an increase in oxidative stress the conversion of mild cognitive impairment to dementia [91]. Dietary
markers as measured by elevated allantoin concentrations in compari- intake of beta-carotene or vitamin C did not influence the course of
son to controls [78]. The mediator of this increase in reactive oxygen dementia. [92]. In the Rotterdam study, no effect of the ApoE genotype
species may be xanthine oxidase. Allantoin and F-isoprostanes appear was found but high vitamin C and vitamin E intake was associated with
to be related to the oxidative assault that may occur as the result of reduced risk for AD [93]. No association between disease risk and intake
chemotherapy in women with breast cancer in the general population of beta-carotene, vitamin E, and vitamin C was found in the Washington
[79]. The role of uric acid and oxidative stress in DS will require elucida- Heights-Inwood Columbia Aging Project, the Honolulu-Aging Study,
tion from additional studies. and the Cache County Study [94–96]. In another study, supplements
The antioxidant properties of melatonin as well as the neurotoxic of vitamins C and E together reduced the risk for AD [97].
factors measured by biomarkers in the kynurenine pathway were
studied in a small group of children with DS [80]. Plasma melatonin 8. The canine model for the study and treatment of oxidative
and urinary kynurenine concentrations were lower than in age- stress
matched controls but levels of kynurenic and anthranilic acids were
elevated. The authors concluded that this pattern of biomarkers con- The often contradictory results in human trials have been one of
stituted an added oxidative risk to children with DS. the factors leading to the investigation of a canine animal model of
Markers of homocysteine and folic acid were examined in 42 preg- aging which has had implications for antioxidant trials in DS. Beagle
nant mothers with a previous history of having delivered a child dogs have a median lifespan of 13.5 years and show many of the
with DS in comparison to 48 control pregnant women. The pattern changes of AD including evidence of oxidative stress [98]. Fig. 3
of metabolites suggested to the authors that mothers who delivered shows the relationship of oxidized Aβ in the brains of individuals
a child with DS had an abnormal oxidant/antioxidant ratio [81]. with DS, AD as compared to the canine model. Their neuropathology
Gelsolin is actin-binding protein that binds to Aβ and inhibits parallels many of the changes of AD including cortical atrophy, neuro-
fibrillogenesis, a pathogenetic step in AD. The active metabolite of nal loss, Aβ plaques, and amyloid angiopathy and oxidative damage.
gelsolin is a proteolytic product which contains a carboxy-terminal With aging, this particular breed of beagle dog exhibits decline in
fragment (CTF). Gelsolin-CTF is decreased in the frontal cortex of executive functioning, learning, and memory. However, the model is
adults with DS (43–63 years) but not in younger individuals (0.5– somewhat incomplete as the dogs do not develop neurofibrillary
23 years). There was a correlation in brain between the gelsolin frag- changes. Nonetheless, many of the neuropathological features in the
ment and both soluble and total concentrations of Aβ40 and Aβ42. canines correlate with the extent of cognitive decline. Topographic
This suggests a potential role for gelsolin in increasing of Aβ levels concentrations of Aβ in the dogs exhibit patterns that parallel those
in DS brain. Oxidative pathways may be involved since gelsolin is in human brain [99]. The cloning, sequencing and expression of APP
cleaved under apoptotic conditions involving oxidative stress [82]. isoforms and the enzymes related to their processing also parallel
Nitric oxide is a regulatory molecule that may play a role in the the findings in human brain [100]. The dogs also show cholinergic
pathogenesis of AD [83]. Inflammation appears to be a mediator of hypofunction similar to that seen in human AD brain, suggesting
nitric oxide effect through glial cells that produce pro-inflammatory that this model could be used for therapeutic screening as well as un-
and neurotoxic factors [84]. Neopterin is an indicator for systemic in- derstanding the links between cholinergic functioning, Aβ pathology,
flammation as well as immune activation. This compound increases and cognitive decline [101].
with age in DS and appears related to the development of dementia Of particular interest to potential clinical trials in DS was an anti-
[85]. In a large population of individuals with DS (401), there was a oxidant supplementation study in which cellular and mitochondrial
strong correlation between neopterin levels in plasma and the pro- antioxidants were given to the beagle dogs [102]. A broad based
duction of nitric oxide [86]. These findings are similar to those diet in the treated group of dogs contained dl-alpha-tocopherol ace-
found in the plasma of AD patients in the general population [87]. tate, l-carnitine, dl-alpha-lipoic acid, ascorbic acid and other dietary
Neopterin measures may reflect oxidative stress in DS and dementia. antioxidants. After one and six months of treatment, the animals
Biomarkers of oxidative and inflammatory stress in blood and were tested for spatial attention ability [103,104]. The treated aged
urine have the advantage of easy accessibility but their ability to animals performed much better than controls and better than the
I.T. Lott / Biochimica et Biophysica Acta 1822 (2012) 657–663 661

A B C

Fig. 3. Characterization of plaques containing oxidized Aβ in the brain tissues. Diffuse plaques in the prefrontal cortex contain oxidized Aβ in the brain of: (A) an aged non-
demented 86 year old individual; (B) a 31 year old non-demented individual with DS; and (C) an aged canine. Scale = 50 μm.

younger animals. The beneficial effects of the antioxidant diet were be weighed against potential risk and disease progression. Targets
enhanced further by environmental stimulation. that attempt to treat several dysfunctions simultaneously may hold
promise for future studies of alternative therapies.
9. Antioxidant trials in DS
10. Conclusions
Since individuals with DS share many of the patterns of oxidative
stress seen in the canine model, a parallel clinical trial was of interest. Oxidative stress is strongly associated with both development
We carried out a randomized, double-blind, placebo-controlled trial and aging in DS. The mouse models of DS that provide homology to
to assess whether daily antioxidant supplementation (900 IU of α- chromosome 21 have afforded a fruitful approach for the experimen-
tocopherol, 200 mg of ascorbic acid and 600 mg of alpha-lipoic acid) tal manipulation of oxidative stress through strategic interventions.
was effective, safe, and tolerable for 53 individuals with DS and AD- Several genes on chromosome 21 have been associated with
type dementia [105]. The outcome measures comprised a battery of oxidative stress and their overexpression may contribute to a genetic
neuropsychological assessments administered at baseline and every imbalance in DS which propagates the development of ROS. Mito-
6 months throughout two years of study. Compared to the placebo chondrial dysfunction is impacted by and also creates oxidative stress
group, those individuals receiving the antioxidant supplement showed in DS. Mitochondria may be a common pathway by which oxidative
neither an improvement in cognitive functioning nor a stabilization stress affects basic mechanisms underlying DS and AD.
of cognitive decline. Mean plasma levels of α-tocopherol increased However, despite strong animal evidence for oxidative stress and its
~2-fold in the treatment group and were consistently higher than the amelioration, clinical trials in both DS and AD have been disappointing
placebo group over the treatment period. Pill counts indicated good to date. Intervention, once dementia has begun in DS, is unlikely to be
compliance with the regimen. No serious adverse events attributed to therapeutic, in part because of the underlying intellectual disability
the treatment were noted. We conclude that antioxidant supplementa- and factors related to decreased synpatogenesis in the disorder. Prophy-
tion is safe, though ineffective as a treatment for dementia in individuals lactic trials may hold more encouragement in DS and it is possible
with DS and AD-type dementia. that early intervention with strategically developed antioxidants
A similar lack of antioxidant efficacy was noted in a study of 156 may positively impact intellectual functioning decades before de-
infants with DS b7 months of age. Supplementation of the diet with mentia sets in. But which antioxidants? The generation of newer
antioxidants, and folinic acid did not show efficacy in a randomized classes of antioxidants and new approaches to intervention will be
controlled trial [106]. needed to combat oxidative stress in both AD and DS.
Another approach to treating oxidative stress in AD has been
through environmental modification, particularly exercise. There is ev- Acknowledgements
idence from experimental models that exercise has time-dependant
benefits to learning and memory [107]. While definitive clinical This research was supported by NIH grants AG-21912, HD-65160,
studies are lacking, there is evidence among older people with AD AG-16573 Alzheimer's Disease Research Center. Nina Movsesyan,
in the general population that weight-bearing exercise reduces the PhD, assisted in the preparation of this manuscript.
decline in activities of daily living [108]. Neurogenesis appears to
be enhanced by environmental factors such as exercise, environ-
References
mental enrichment and dietary energy restriction [109]. In this con-
text, exercise was tested in a small study of adults with DS and lipid [1] M. Hattori, A. Fujiyama, T.D. Taylor, H. Watanabe, T. Yada, H.S. Park, A. Toyoda, K.
peroxidation, a marker of oxidative stress, was reported to be re- Ishii, Y. Totoki, D.K. Choi, Y. Groner, E. Soeda, M. Ohki, T. Takagi, Y. Sakaki, S.
Taudien, K. Blechschmidt, A. Polley, U. Menzel, J. Delabar, K. Kumpf, R. Lehmann,
duced in saliva [110]. Plasma oxidative stress markers were reduced D. Patterson, K. Reichwald, A. Rump, M. Schillhabel, A. Schudy, W. Zimmermann,
in adolescents with DS who underwent a 12 week aerobic training A. Rosenthal, J. Kudoh, K. Schibuya, K. Kawasaki, S. Asakawa, A. Shintani, T.
program [111]. The role of exercise in lowering oxidative stress in Sasaki, K. Nagamine, S. Mitsuyama, S.E. Antonarakis, S. Minoshima, N. Shimizu,
G. Nordsiek, K. Hornischer, P. Brant, M. Scharfe, O. Schon, A. Desario, J. Reichelt,
DS merits further investigation.
G. Kauer, H. Blocker, J. Ramser, A. Beck, S. Klages, S. Hennig, L. Riesselmann, E.
Many individuals with DS receive complementary and alternative Dagand, T. Haaf, S. Wehrmeyer, K. Borzym, K. Gardiner, D. Nizetic, F. Francis,
therapies, some of which are aimed at oxidative stress. The pros and H. Lehrach, R. Reinhardt, M.L. Yaspo, The DNA sequence of human chromosome
cons of pharmaceutical, nutritional, botanical and stimulatory thera- 21, Nature 405 (2000) 311–319.
[2] X. Sturgeon, K.J. Gardiner, Transcript catalogs of human chromosome 21 and
pies for AD have been reviewed [112]. The author concludes that orthologous chimpanzee and mouse regions, Mamm. Genome, 22, 2011,
potential risks of both approved and non-approved therapies should pp. 261–271.
662 I.T. Lott / Biochimica et Biophysica Acta 1822 (2012) 657–663

[3] S.E. Antonarakis, R. Lyle, E.T. Dermitzakis, A. Reymond, S. Deutsch, Chromosome [33] J. Busciglio, A. Pelsman, P. Helguera, O. Ashur-Fabian, A. Pinhasov, D.E. Brenneman,
21 and down syndrome: from genomics to pathophysiology, nature reviews, I. Gozes, NAP and ADNF-9 protect normal and Down's syndrome cortical neurons
Genetics 5 (2004) 725–738. from oxidative damage and apoptosis, Curr. Pharm. Des. 13 (2007) 1091–1098.
[4] P. Helguera, A. Pelsman, G. Pigino, E. Wolvetang, E. Head, J. Busciglio, ets-2 pro- [34] Z. Wu, Y. Zhao, B. Zhao, Superoxide anion, uncoupling proteins and Alzheimer's
motes the activation of a mitochondrial death pathway in Down's syndrome disease, J. Clin. Biochem. Nutr. 46 (2010) 187–194.
neurons, J. Neurosci. 25 (2005) 2295–2303. [35] K.J. Young, J.P. Bennett, The mitochondrial secret(ase) of Alzheimer's disease,
[5] K.T. Chang, K.T. Min, Drosophila melanogaster homolog of Down syndrome J. Alzheimer's Dis. 20 (Suppl 2) (2010) S381–S400.
critical region 1 is critical for mitochondrial function, Nat. Neurosci. 8 (2005) [36] J.R. Korenberg, H. Kawashima, S.M. Pulst, T. Ikeuchi, N. Ogasawara, K. Yamamoto,
1577–1585. S.A. Schonberg, R. West, L. Allen, E. Magenis, et al., Molecular definition of a
[6] A. Salehi, M. Faizi, P.V. Belichenko, W.C. Mobley, Using mouse models to explore region of chromosome 21 that causes features of the Down syndrome pheno-
genotype–phenotype relationship in Down syndrome, Ment. Retard. Dev. Disabil. type, Am. J. Hum. Genet. 47 (1990) 236–246.
Res. Rev. 13 (2007) 207–214. [37] S. Szymonik-Lesiuk, G. Czechowska, M. Stryjecka-Zimmer, M. Slomka, A. Madro,
[7] I. Das, R.H. Reeves, The use of mouse models to understand and improve cogni- K. Celinski, M. Wielosz, Catalase, superoxide dismutase, and glutathione perox-
tive deficits in Down syndrome, Dis. Models Mech. 4 (2011) 596–606. idase activities in various rat tissues after carbon tetrachloride intoxication,
[8] P.L. Roubertoux, M. Carlier, Mouse models of cognitive disabilities in trisomy 21 J. Hepato-Biliary-Pancreatic Surg. 10 (2003) 309–315.
(Down syndrome), American journal of medical genetics, part C, Semin. Med. [38] J.A. Badwey, M.L. Karnovsky, Active oxygen species and the functions of phago-
Genet. 154C (2010) 400–416. cytic leukocytes, Annu. Rev. Biochem. 49 (1980) 695–726.
[9] A. Salehi, J.D. Delcroix, W.C. Mobley, Traffic at the intersection of neurotrophic [39] O. Akinci, E. Mihci, S. Tacoy, F. Kardelen, I. Keser, M. Aslan, Neutrophil oxidative
factor signaling and neurodegeneration, Trends Neurosci. 26 (2003) 73–80. metabolism in Down syndrome patients with congenital heart defects, Environ.
[10] A.M. Kleschevnikov, P.V. Belichenko, A.J. Villar, C.J. Epstein, R.C. Malenka, W.C. Mol. Mutagen. 51 (2010) 57–63.
Mobley, Hippocampal long-term potentiation suppressed by increased inhibi- [40] V. Costa, L. Sommese, A. Casamassimi, R. Colicchio, C. Angelini, V. Marchesano, L.
tion in the Ts65Dn mouse, a genetic model of Down syndrome, J. Neurosci. 24 Milone, B. Farzati, A. Giovane, C. Fiorito, M. Rienzo, M. Picardi, B. Avallone, M.
(2004) 8153–8160. Marco Corsi, B. Sarubbi, R. Calabro, P. Salvatore, A. Ciccodicola, C. Napoli, Impair-
[11] M.A. Kurt, M.I. Kafa, M. Dierssen, D.C. Davies, Deficits of neuronal density in CA1 ment of circulating endothelial progenitors in Down syndrome, BMC Med.
and synaptic density in the dentate gyrus, CA3 and CA1, in a mouse model of Genomics 3 (2010) 40.
Down syndrome, Brain Res. 1022 (2004) 101–109. [41] R.X. Moldrich, L. Dauphinot, J. Laffaire, T. Vitalis, Y. Herault, P.M. Beart, J. Rossier,
[12] A.C. Costa, M.J. Grybko, Deficits in hippocampal CA1 LTP induced by TBS but not D. Vivien, C. Gehrig, S.E. Antonarakis, R. Lyle, M.C. Potier, Proliferation deficits
HFS in the Ts65Dn mouse: a model of Down syndrome, Neurosci. Lett. 382 and gene expression dysregulation in Down's syndrome (Ts1Cje) neural pro-
(2005) 317–322. genitor cells cultured from neurospheres, J. Neurosci. Res. 87 (2009) 3143–3152.
[13] L.A. Hyde, D.F. Frisone, L.S. Crnic, Ts65Dn mice, a model for Down syndrome, have [42] J. Borg, E. Chereul, Differential MRI patterns of brain atrophy in double or sin-
deficits in context discrimination learning suggesting impaired hippocampal func- gle transgenic mice for APP and/or SOD, J. Neurosci. Res. 86 (2008)
tion, Behav. Brain Res. 118 (2001) 53–60. 3275–3284.
[14] D. Patterson, Molecular genetic analysis of Down syndrome, Hum. Genet. 126 [43] K. Macleod, D. Leprince, D. Stehelin, The ets gene family, Trends Biochem. Sci. 17
(2009) 195–214. (1992) 251–256.
[15] K. Ishihara, K. Amano, E. Takaki, A.S. Ebrahim, A. Shimohata, N. Shibazaki, I. Inoue, M. [44] B. Wasylyk, S.L. Hahn, A. Giovane, The Ets family of transcription factors, Eur.
Takaki, Y. Ueda, H. Sago, C.J. Epstein, K. Yamakawa, Increased lipid peroxidation in J. Biochem. 211 (1993) 7–18 FEBS.
Down's syndrome mouse models, J. Neurochem. 110 (2009) 1965–1976. [45] A. Yang, R.H. Reeves, Increased survival following tumorigenesis in Ts65Dn mice
[16] E.A. Shukkur, A. Shimohata, T. Akagi, W. Yu, M. Yamaguchi, M. Murayama, D. that model Down syndrome, Cancer Res. 71 (2011) 3573–3581.
Chui, T. Takeuchi, K. Amano, K.H. Subramhanya, T. Hashikawa, H. Sago, C.J. [46] L.E. Reynolds, A.R. Watson, M. Baker, T.A. Jones, G. D'Amico, S.D. Robinson, C.
Epstein, A. Takashima, K. Yamakawa, Mitochondrial dysfunction and tau hyper- Joffre, S. Garrido-Urbani, J.C. Rodriguez-Manzaneque, E. Martino-Echarri, M.
phosphorylation in Ts1Cje, a mouse model for Down syndrome, Hum. Mol. Aurrand-Lions, D. Sheer, F. Dagna-Bricarelli, D. Nizetic, C.J. McCabe, A.S. Turnell,
Genet. 15 (2006) 2752–2762. S. Kermorgant, B.A. Imhof, R. Adams, E.M. Fisher, V.L. Tybulewicz, I.R. Hart, K.M.
[17] M. Shichiri, Y. Yoshida, N. Ishida, Y. Hagihara, H. Iwahashi, H. Tamai, E. Niki, Hodivala-Dilke, Tumour angiogenesis is reduced in the Tc1 mouse model of
alpha-Tocopherol suppresses lipid peroxidation and behavioral and cognitive Down's syndrome, Nature 465 (2010) 813–817.
impairments in the Ts65Dn mouse model of Down syndrome, Free Radic. Biol. [47] A. Rovelet-Lecrux, D. Hannequin, G. Raux, N. Le Meur, A. Laquerriere, A. Vital, C.
Med. 50 (2011) 1801–1811. Dumanchin, S. Feuillette, A. Brice, M. Vercelletto, F. Dubas, T. Frebourg, D. Campion,
[18] J. Lockrow, A. Prakasam, P. Huang, H. Bimonte-Nelson, K. Sambamurti, A.C. APP locus duplication causes autosomal dominant early-onset Alzheimer disease
Granholm, Cholinergic degeneration and memory loss delayed by vitamin E in with cerebral amyloid angiopathy, Nat. Genet. 38 (2006) 24–26.
a Down syndrome mouse model, Exp. Neurol. 216 (2009) 278–289. [48] V.P. Prasher, M.J. Farrer, A.M. Kessling, E.M. Fisher, R.J. West, P.C. Barber, A.C.
[19] N. Rueda, J. Florez, C. Martinez-Cue, Effects of chronic administration of SGS-111 Butler, Molecular mapping of Alzheimer-type dementia in Down's syndrome,
during adulthood and during the pre- and post-natal periods on the cognitive Ann. Neurol. 43 (1998) 380–383.
deficits of Ts65Dn mice, a model of Down syndrome, Behav. Brain Res. 188 [49] N. Schupf, B. Patel, W. Silverman, W.B. Zigman, N. Zhong, B. Tycko, P.D. Mehta, R.
(2008) 355–367. Mayeux, Elevated plasma amyloid beta-peptide 1-42 and onset of dementia in
[20] D.M. Mann, M.M. Esiri, The pattern of acquisition of plaques and tangles in the adults with Down syndrome, Neurosci. Lett. 301 (2001) 199–203.
brains of patients under 50 years of age with Down's syndrome, J. Neurol. Sci. [50] J.K. Teller, C. Russo, L.M. DeBusk, G. Angelini, D. Zaccheo, F. Dagna-Bricarelli, P.
89 (1989) 169–179. Scartezzini, S. Bertolini, D.M. Mann, M. Tabaton, P. Gambetti, Presence of soluble
[21] I.T. Lott, E. Head, Alzheimer disease and Down syndrome: factors in pathogenesis, amyloid beta-peptide precedes amyloid plaque formation in Down's syndrome,
Neurobiol. Aging 26 (2005) 383–389. Nature Med. 2 (1996) 93–95.
[22] K.E. Wisniewski, H.M. Wisniewski, G.Y. Wen, Occurrence of neuropathological [51] N.H. Zawia, D.K. Lahiri, F. Cardozo-Pelaez, Epigenetics, oxidative stress, and
changes and dementia of Alzheimer's disease in Down's syndrome, Ann. Neurol. Alzheimer disease, Free Radic. Biol. Med. 46 (2009) 1241–1249.
17 (1985) 278–282. [52] D.R. Crawford, K.P. Leahy, N. Abramova, L. Lan, Y. Wang, K.J. Davies, Hamster
[23] J. Kedziora, G. Bartosz, Down's syndrome: a pathology involving the lack of adapt78 mRNA is a Down syndrome critical region homologue that is inducible
balance of reactive oxygen species, Free Radic. Biol. Med. 4 (1988) 317–330. by oxidative stress, Arch. Biochem. Biophys. 342 (1997) 6–12.
[24] J. Busciglio, J.K. Andersen, H.M. Schipper, G.M. Gilad, R. McCarty, F. Marzatico, O. [53] G. Ermak, C. Cheadle, K.G. Becker, C.D. Harris, K.J. Davies, DSCR1(Adapt78)
Toussaint, Stress, aging, and neurodegenerative disorders, molecular mecha- modulates expression of SOD1, FASEB J. 18 (2004) 62–69.
nisms, Ann. N. Y. Acad. Sci. 851 (1998) 429–443. [54] C.D. Harris, G. Ermak, K.J. Davies, Multiple roles of the DSCR1 (Adapt78 or
[25] I.T. Lott, E. Head, E. Doran, J. Busciglio, Beta-amyloid, oxidative stress and down RCAN1) gene and its protein product calcipressin 1 (or RCAN1) in disease, cellu-
syndrome, Curr. Alzheimer Res 3 (2006) 521–528. lar and molecular life sciences, CMLS 62 (2005) 2477–2486.
[26] G.P. Reynolds, A.J. Cutts, Free radical damage in Down's syndrome brain, Bio- [55] K.H. Baek, A. Zaslavsky, R.C. Lynch, C. Britt, Y. Okada, R.J. Siarey, M.W. Lensch, I.H.
chem. Soc. Trans. 21 (1993) 221S. Park, S.S. Yoon, T. Minami, J.R. Korenberg, J. Folkman, G.Q. Daley, W.C. Aird, Z.
[27] B.W. Brooksbank, M. Martinez, R. Balazs, Altered composition of polyunsaturated Galdzicki, S. Ryeom, Down's syndrome suppression of tumour growth and the
fatty acyl groups in phosphoglycerides of Down's syndrome fetal brain, J. Neuro- role of the calcineurin inhibitor DSCR1, Nature 459 (2009) 1126–1130.
chem. 44 (1985) 869–874. [56] F. Liu, Z. Liang, J. Wegiel, Y.W. Hwang, K. Iqbal, I. Grundke-Iqbal, N. Ramakrishna,
[28] P. Odetti, G. Angelini, D. Dapino, D. Zaccheo, S. Garibaldi, F. Dagna-Bricarelli, G. C.X. Gong, Overexpression of Dyrk1A contributes to neurofibrillary degenera-
Piombo, G. Perry, M. Smith, N. Traverso, M. Tabaton, Early glycoxidation damage tion in Down syndrome, FASEB J. 22 (2008) 3224–3233.
in brains from Down's syndrome, Biochem. Biophys. Res. Commun. 243 (1998) [57] G. Arque, M.M. de Lagran, M.L. Arbones, M. Dierssen, Age-associated motor and
849–851. visuo-spatial learning phenotype in Dyrk1A heterozygous mutant mice, Neuro-
[29] J. Busciglio, B.A. Yankner, Apoptosis and increased generation of reactive oxygen biol. Dis. 36 (2009) 312–319.
species in Down's syndrome neurons in vitro, Nature 378 (1995) 776–779. [58] F. Guedj, C. Sebrie, I. Rivals, A. Ledru, E. Paly, J.C. Bizot, D. Smith, E. Rubin, B.
[30] E. Head, W. Garzon-Rodriguez, J.K. Johnson, I.T. Lott, C.W. Cotman, C. Glabe, Gillet, M. Arbones, J.M. Delabar, Green tea polyphenols rescue of brain defects
Oxidation of Abeta and plaque biogenesis in Alzheimer's disease and Down syn- induced by overexpression of DYRK1A, PLoS One 4 (2009) e4606.
drome, Neurobiol. Dis. 8 (2001) 792–806. [59] R.X. Santos, S.C. Correia, X. Wang, G. Perry, M.A. Smith, P.I. Moreira, X. Zhu,
[31] P. Mecocci, U. MacGarvey, M.F. Beal, Oxidative damage to mitochondrial DNA is Alzheimer's disease: diverse aspects of mitochondrial malfunctioning, Int.
increased in Alzheimer's disease, Ann. Neurol. 36 (1994) 747–751. J. Clin. Exp. Pathol. 3 (2010) 570–581.
[32] M.A. Smith, G. Perry, P.L. Richey, L.M. Sayre, V.E. Anderson, M.F. Beal, N. Kowall, [60] E.H. Corder, A.M. Saunders, W.J. Strittmatter, D.E. Schmechel, P.C. Gaskell, G.W.
Oxidative damage in Alzheimer's, Nature 382 (1996) 120–121. Small, A.D. Roses, J.L. Haines, M.A. Pericak-Vance, Gene dose of apolipoprotein
I.T. Lott / Biochimica et Biophysica Acta 1822 (2012) 657–663 663

E type 4 allele and the risk of Alzheimer's disease in late onset families, Science [86] A.M. Coppus, D. Fekkes, W.M. Verhoeven, S. Tuinier, C.M. van Duijn, Plasma
261 (1993) 921–923. levels of nitric oxide related amino acids in demented subjects with Down
[61] A. Patel, S.D. Rees, M.A. Kelly, S.C. Bain, A.H. Barnett, D. Thalitaya, V.P. Prasher, syndrome are related to neopterin concentrations, Amino acids 38 (2010)
Association of variants within APOE, SORL1, RUNX1, BACE1 and ALDH18A1 923–928.
with dementia in Alzheimer's disease in subjects with Down syndrome, Neu- [87] M. Hull, G.M. Pasinetti, P.S. Aisen, Elevated plasma neopterin levels in Alzheimer
rosci. Lett. 487 (2011) 144–148. disease, Alzheimer Dis. Assoc. Disord. 14 (2000) 228–230.
[62] S. Chang, T. ran Ma, R.D. Miranda, M.E. Balestra, R.W. Mahley, Huang, lipid- and [88] D. Galasko, T.J. Montine, Biomarkers of oxidative damage and inflammation in
receptor-binding regions of apolipoprotein E4 fragments act in concert to cause Alzheimer's disease, Biomarkers Med. 4 (2010) 27–36.
mitochondrial dysfunction and neurotoxicity, Proc. Natl. Acad. Sci. U. S. A. 102 [89] Y.X. Sun, L. Minthon, A. Wallmark, S. Warkentin, K. Blennow, S. Janciauskiene,
(2005) 18694–18699. Inflammatory markers in matched plasma and cerebrospinal fluid from patients
[63] H.M. Abdul, R. Sultana, D.K. St Clair, W.R. Markesbery, D.A. Butterfield, Oxidative with Alzheimer's disease, Dement. Geriatr. Cogn. Disord. 16 (2003) 136–144.
damage in brain from human mutant APP/PS-1 double knock-in mice as a func- [90] L. Craggs, R.N. Kalaria, Revisiting dietary antioxidants, neurodegeneration and
tion of age, Free Radic. Biol. Med. 45 (2008) 1420–1425. dementia, NeuroReport 22 (2011) 1–3.
[64] W.D. Parker Jr., C.M. Filley, J.K. Parks, Cytochrome oxidase deficiency in Alzheimer's [91] R.C. Petersen, R.G. Thomas, M. Grundman, D. Bennett, R. Doody, S. Ferris, D. Galasko,
disease, Neurology 40 (1990) 1302–1303. S. Jin, J. Kaye, A. Levey, E. Pfeiffer, M. Sano, C.H. van Dyck, L.J. Thal, Vitamin E and
[65] C. Caspersen, N. Wang, J. Yao, A. Sosunov, X. Chen, J.W. Lustbader, H.W. Xu, D. Stern, donepezil for the treatment of mild cognitive impairment, N. Engl. J. Med. 352
G. McKhann, S.D. Yan, Mitochondrial Abeta: a potential focal point for neuronal (2005) 2379–2388.
metabolic dysfunction in Alzheimer's disease, FASEB J. 19 (2005) 2040–2041. [92] M.C. Morris, D.A. Evans, J.L. Bienias, C.C. Tangney, D.A. Bennett, N. Aggarwal,
[66] G.E. Gibson, S.S. Karuppagounder, Q. Shi, Oxidant-induced changes in mitochon- R.S. Wilson, P.A. Scherr, Dietary intake of antioxidant nutrients and the risk
dria and calcium dynamics in the pathophysiology of Alzheimer's disease, Ann. of incident Alzheimer disease in a biracial community study, JAMA 287
N. Y. Acad. Sci. 1147 (2008) 221–232. (2002) 3230–3237.
[67] V. Rhein, X. Song, A. Wiesner, L.M. Ittner, G. Baysang, F. Meier, L. Ozmen, H. [93] M.J. Engelhart, M.I. Geerlings, A. Ruitenberg, J.C. van Swieten, A. Hofman, J.C.
Bluethmann, S. Drose, U. Brandt, E. Savaskan, C. Czech, J. Gotz, A. Eckert, Amyloid- Witteman, M.M. Breteler, Dietary intake of antioxidants and risk of Alzheimer
beta and tau synergistically impair the oxidative phosphorylation system in triple disease, JAMA 287 (2002) 3223–3229.
transgenic Alzheimer's disease mice, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) [94] J.A. Luchsinger, M.X. Tang, S. Shea, R. Mayeux, Antioxidant vitamin intake and
20057–20062. risk of Alzheimer disease, Arch. Neurol. 60 (2003) 203–208.
[68] M. Corral-Debrinski, T. Horton, M.T. Lott, J.M. Shoffner, A.C. McKee, M.F. Beal, [95] D. Laurin, K.H. Masaki, D.J. Foley, L.R. White, L.J. Launer, Midlife dietary intake of
B.H. Graham, D.C. Wallace, Marked changes in mitochondrial DNA deletion antioxidants and risk of late-life incident dementia: the Honolulu-Asia Aging
levels in Alzheimer brains, Genomics 23 (1994) 471–476. Study, Am. J. Epidemiol. 159 (2004) 959–967.
[69] S.V. Jovanovic, D. Clements, K. MacLeod, Biomarkers of oxidative stress are signifi- [96] P.P. Zandi, J.C. Anthony, A.S. Khachaturian, S.V. Stone, D. Gustafson, J.T. Tschanz,
cantly elevated in Down syndrome, Free Radic. Biol. Med. 25 (1998) 1044–1048. M.C. Norton, K.A. Welsh-Bohmer, J.C. Breitner, Reduced risk of Alzheimer dis-
[70] P.E. Coskun, J. Wyrembak, O. Derbereva, G. Melkonian, E. Doran, I.T. Lott, E. Head, ease in users of antioxidant vitamin supplements: the Cache County Study,
C.W. Cotman, D.C. Wallace, Systemic mitochondrial dysfunction and the etiology Arch. Neurol. 61 (2004) 82–88.
of Alzheimer's disease and down syndrome dementia, J. Alzheimer's Dis. 20 [97] N. Scarmeas, Y. Stern, R. Mayeux, J.A. Luchsinger, Mediterranean diet, Alzheimer
(Suppl 2) (2010) S293–S310. disease, and vascular mediation, Arch. Neurol. 63 (2006) 1709–1717.
[71] J. Lu, G. Esposito, C. Scuderi, L. Steardo, L.C. Delli-Bovi, J.L. Hecht, B.C. Dickinson, C.J. [98] C.W. Cotman, E. Head, The canine (dog) model of human aging and disease:
Chang, T. Mori, V. Sheen, S100B and APP promote a gliocentric shift and impaired dietary, environmental and immunotherapy approaches, J. Alzheimer's Dis. 15
neurogenesis in Down syndrome neural progenitors, PLoS One 6 (2011) e22126. (2008) 685–707.
[72] D. Valenti, G.A. Manente, L. Moro, E. Marra, R.A. Vacca, Deficit of complex I activity [99] E. Head, V. Pop, F. Sarsoza, R. Kayed, T.L. Beckett, C.M. Studzinski, J.L. Tomic, C.G.
in human skin fibroblasts with chromosome 21 trisomy and overproduction of Glabe, M.P. Murphy, Amyloid-beta peptide and oligomers in the brain and cere-
reactive oxygen species by mitochondria: involvement of the cAMP/PKA signalling brospinal fluid of aged canines, J. Alzheimer's Dis. 20 (2010) 637–646.
pathway, Biochem. J. 435 (2011) 679–688. [100] L. Sarasa, C. Gallego, I. Monleon, A. Olvera, J. Canudas, M. Montanes, P. Pesini, M.
[73] G. Dogliotti, E. Galliera, E. Dozio, E. Vianello, R.E. Villa, F. Licastro, I. Barajon, M.M. Sarasa, Cloning, sequencing and expression in the dog of the main amyloid pre-
Corsi, Okadaic acid induces apoptosis in Down syndrome fibroblasts, Toxicol. in cursor protein isoforms and some of the enzymes related with their processing,
Vitro 24 (2010) 815–821. Neuroscience 171 (2010) 1091–1101.
[74] V. Infantino, A. Castegna, F. Iacobazzi, I. Spera, I. Scala, G. Andria, V. Iacobazzi, [101] J.A. Araujo, N.H. Greig, D.K. Ingram, J. Sandin, C. de Rivera, N.W. Milgram, Cholin-
Impairment of methyl cycle affects mitochondrial methyl availability and gluta- esterase inhibitors improve both memory and complex learning in aged beagle
thione level in Down's syndrome, Mol. Genet. Metab. 102 (2011) 378–382. dogs, J. Alzheimer's Dis. 26 (2011) 143–155.
[75] G. Aliev, Y. Li, H.H. Palacios, M.E. Obrenovich, Oxidative stress induced mitochon- [102] E. Head, V.N. Nukala, K.A. Fenoglio, B.A. Muggenburg, C.W. Cotman, P.G. Sullivan,
drial DNA deletion as a hallmark for the drug development in the context of the Effects of age, dietary, and behavioral enrichment on brain mitochondria in a
cerebrovascular diseases, Recent patents on cardiovascular drug discovery 6 canine model of human aging, Exp. Neurol. 220 (2009) 171–176.
(2011) 222–241. [103] C.W. Cotman, E. Head, B.A. Muggenburg, S. Zicker, N.W. Milgram, Brain aging in
[76] A. Strydom, M.J. Dickinson, S. Shende, D. Pratico, Z. Walker, Oxidative stress and the canine: a diet enriched in antioxidants reduces cognitive dysfunction,
cognitive ability in adults with Down syndrome, Prog. Neuro-Psychopharmacolog. Neurobiol. Aging 23 (2002) 809–818.
Biol. Psychiatry 33 (2009) 76–80. [104] N.W. Milgram, B. Adams, H. Callahan, E. Head, B. Mackay, C. Thirlwell, C.W.
[77] C. Campos, R. Guzman, E. Lopez-Fernandez, A. Casado, Urinary uric acid and Cotman, Landmark discrimination learning in the dog, Learn. Mem. 6 (1999)
antioxidant capacity in children and adults with Down syndrome, Clin. Biochem. 54–61.
43 (2010) 228–233. [105] I.T. Lott, E. Doran, V.Q. Nguyen, A. Tournay, E. Head, D.L. Gillen, Down syndrome
[78] L. Tiano, L. Padella, P. Carnevali, O. Gabrielli, F. Bruge, F. Principi, G.P. Littarru, and dementia: a randomized, controlled trial of antioxidant supplementation,
Coenzyme Q10 and oxidative imbalance in Down syndrome: biochemical and part A, Am. J. Med. Genet. 155A (2011) 1939–1948.
clinical aspects, Biofactors 32 (2008) 161–167. [106] J.M. Ellis, H.K. Tan, R.E. Gilbert, D.P. Muller, W. Henley, R. Moy, R. Pumphrey, C.
[79] D. Il'yasova, K. Kennedy, I. Spasojevic, F. Wang, A.A. Tolun, K. Base, S.P. Young, P. Ani, S. Davies, V. Edwards, H. Green, A. Salt, S. Logan, Supplementation with
Kelly Marcom, J. Marks, D.S. Millington, M.W. Dewhirst, Individual responses to antioxidants and folinic acid for children with Down's syndrome: randomised
chemotherapy-induced oxidative stress, Breast Cancer Res. Treat. 125 (2011) controlled trial, BMJ 336 (2008) 594–597.
583–589. [107] N.C. Berchtold, N. Castello, C.W. Cotman, Exercise and time-dependent benefits
[80] J. Uberos, J. Romero, A. Molina-Carballo, A. Munoz-Hoyos, Melatonin and elimi- to learning and memory, Neuroscience 167 (2010) 588–597.
nation of kynurenines in children with Down's syndrome, J. Pediatr. Endocrinol. [108] H. Littbrand, M. Stenvall, E. Rosendahl, Applicability and effects of physical exer-
Metab. 23 (2010) 277–282. cise on physical and cognitive functions and activities of daily living among peo-
[81] N.A. Meguid, A.A. Dardir, E.M. El-Sayed, H.H. Ahmed, A.F. Hashish, A. Ezzat, Homo- ple with dementia: a systematic review, Am. J. Phys. Med. Rehabil. / Assoc. Acad.
cysteine and oxidative stress in Egyptian children with Down syndrome, Clin. Phyciatrists, 90, 2011, pp. 495–518.
Biochem. 43 (2010) 963–967. [109] O. Lazarov, M.P. Mattson, D.A. Peterson, S.W. Pimplikar, H. van Praag, When neu-
[82] L. Ji, V. Chauhan, P. Mehta, J. Wegiel, S. Mehta, A. Chauhan, Relationship between rogenesis encounters aging and disease, Trends Neurosci. 33 (2010) 569–579.
proteolytically cleaved gelsolin and levels of amyloid-beta protein in the brains [110] J.C. Zambrano, R. Marquina, N. Sulbaran, A.J. Rodriguez-Malaver, R.A. Reyes,
of Down syndrome subjects, J. Alzheimer's Dis. 22 (2010) 609–617. Aerobic exercise reduced oxidative stress in saliva of persons with Down syn-
[83] A. Akomolafe, K.L. Lunetta, P.M. Erlich, L.A. Cupples, C.T. Baldwin, M. Huyck, R.C. drome, Res. Sports Med. 17 (2009) 195–203.
Green, L.A. Farrer, Genetic association between endothelial nitric oxide synthase [111] M. Rosety-Rodriguez, I. Rosety, G. Fornieles-Gonzalez, A. Diaz, M. Rosety, F.J.
and Alzheimer disease, Clin. Genet. 70 (2006) 49–56. Ordonez, A 12-week aerobic training programme reduced plasmatic allantoin
[84] J.N. Sharma, A. Al-Omran, S.S. Parvathy, Role of nitric oxide in inflammatory in adolescents with Down syndrome, Br. J. Sports Med. 44 (2010) 685–687.
diseases, Inflammopharmacology 15 (2007) 252–259. [112] K.A. Wollen, Alzheimer's disease: the pros and cons of pharmaceutical, nutri-
[85] A.W. Coppus, D. Fekkes, W.M. Verhoeven, S. Tuinier, J.I. Egger, C.M. van Duijn, tional, botanical, and stimulatory therapies, with a discussion of treatment
Plasma amino acids and neopterin in healthy persons with Down's syndrome, strategies from the perspective of patients and practitioners, Altern. Med.
J. Neural. Transm. 114 (2007) 1041–1045. Rev. 15 (2010) 223–244.

You might also like