You are on page 1of 12

Send Orders for Reprints to reprints@benthamscience.

ae
18 Current Alzheimer Research, 2016, 13, 18-29

Aging in Down Syndrome and the Development of Alzheimer’s Disease


Neuropathology

Elizabeth Head1,*, Ira T. Lott2, Donna M. Wilcock3 and Cynthia A. Lemere4

1
Department of Pharmacology & Nutritional Sciences, Sanders Brown Center on Aging, University of
Kentucky, Lexington, KY, USA; 2Department of Pediatrics and Neurology, University of California at
Irvine School of Medicine, Orange County, CA, USA; 3Department of Physiology, Sanders Brown
Center on Aging, University of Kentucky, Lexington, KY, USA; 4Department of Neurology and the
Anne Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical
School, Boston, MA, USA

Abstract: Chromosome 21, triplicated in Down Syndrome, contains several genes that are thought to
play a critical role in the development of AD neuropathology. The overexpression of the gene for the Elizabeth Head
amyloid precursor protein (APP), on chromosome 21, leads to early onset beta-amyloid (Aβ) plaques
in DS. In addition to Aβ accumulation, middle-aged people with DS develop neurofibrillary tangles, cerebrovascular pa-
thology, white matter pathology, oxidative damage, neuroinflammation and neuron loss. There is also evidence of poten-
tial compensatory responses in DS that benefit the brain and delay the onset of dementia after there is sufficient neuropa-
thology for a diagnosis of AD. This review describes some of the existing literature and also highlights gaps in our knowl-
edge regarding AD neuropathology in DS. It will be critical in the future to develop networked brain banks with standard-
ized collection procedures to fully characterize the regional and temporal pathological events associated with aging in DS.
As more information is acquired regarding AD evolution in DS, there will be opportunities to develop interventions that
are age-appropriate to delay AD in DS.
Keywords: Beta-amyloid, neurofibrillary tangles, neuroinflammation, oxidative damage, posttranslational modifications, senile
plaques, trisomy 21, vascular pathology, white matter damage.

BACKGROUND APP PROCESSING AND DOWN SYNDROME


Down syndrome (DS) was initially described by J. Lang- Aβ is produced from a longer precursor protein, APP,
don Down in 1866 [1]. In 1959, Lejeune identified DS as a which is present in triplicate in DS [10, 11]. It is interesting
trisomy of chromosome 21 [2]. DS or Trisomy 21, is one of to note that when APP is overexpressed, there is a parallel
the most common causes of intellectual disability (ID) and increase in the release of sAPPα, which in turn can activate
recent national prevalence estimates suggest that 1 in every microglia and induce IL-1β and lead to additional increases
691 babies in the USA is born with DS leading to ~6,000 in APP [12]. This in turn may explain why there are observa-
annual DS births (www.ndss.org). DS is associated with tions of a greater than 1.5 fold increase in APP as a result of
characteristic physical features, deficits in the immune and trisomy 21 [10]. Aβ is produced by sequential cleavage of
endocrine systems and delayed cognitive development [3, 4]. APP by β-secretase and subsequently by γ-secretase [13, 14].
β-secretase has been identified as beta-site APP cleaving
Improvements in medical care, education, and support for
enzyme or BACE1 [15]. In DS, however, β-secretase activity
children and adults with DS, have led to significant exten-
sions in lifespan and enhanced quality of life [5, 6]. As a has variably been shown to increase with age [16] or shows
modest increases in protein level [17], whereas other studies
consequence, up to age 35 years, mortality rates are compa-
report little change suggesting that APP overexpression may
rable in adults with DS to individuals with intellectual dis-
be the primary driver of Aβ plaque accumulation [18, 19].
ability from other causes [7]. However, after age 35, mortal-
ity rates double every 6.4 years in DS as compared to every BACE2 can also cleave APP at the β -secretase site [20]
9.6 years for people without DS [7]. The average lifespan of Interestingly, the gene for BACE-2, which shares significant
people with DS has approximately doubled over the past 30 homology with BACE1, is also located on chromosome 21
years to 55-60 years of age (www.ndss.org; [8, 9]). One of in the DS obligate region and may contribute to increased
the consequences of aging in people with DS is increasing Aβ production. Developmentally, BACE-2 RNA levels are
accumulation of Alzheimer’s disease (AD) neuropathology significantly higher in DS fetal tissue relative to controls
primarily due to overexpression of the APP gene on chromo- [21]. In addition, in cultured fibroblasts from adults with DS,
some 21. BACE-2 mRNA (i.e. protein expression) was 2.6 fold higher
than normal controls. Increased levels of BACE-2 may also
*Address correspondence to this author at the Sanders Brown Center on
Aging, University of Kentucky, 800 South Limestone Street, Lexington, contribute to aging in DS. In 13 people with DS ranging in
KY, 40536, USA; Tel: 859-218-3172; E-mail: elizabeth.head@uky.edu age from 27 weeks to 37 years, frontal cortex BACE-2 im-

1875-5828/16 $58.00+.00 © 2016 Bentham Science Publishers


AD Neuropathology in DS Current Alzheimer Research, 2016, Vol. 13, No. 1 19

munoreactivity was observed only in neurons of adults with However when all the observations of these different re-
DS and AD. BACE-2 immunoreactivity was not observed in ports are combined they provide evidence for APP process-
younger individuals [22]. However, several studies compar- ing into heterogeneous fragments. One hypothesis specifi-
ing DS brain to similarly aged control brains do not find cally describes a “solid phase” pathway for Aβ production
higher levels of BACE2 protein [18, 23]. Similarly, no dif- from APP where nonspecific proteolytic cleavage leads to
ferences in DS as compared to controls was observed for heterogeneous Aβ (including Aβ1-40/42/43). A key compo-
BACE2 in the intracellular compartment [21]. It is possible nent of this hypothesis is that APP processing occurs within
that despite increased RNA for BACE2 in DS, there may be the endosomal/lysosomal system [43]. It is interesting to
posttranscriptional regulatory mechanisms that lead to nor- note that studies of intracellular Aβ in DS all report a possi-
mal levels of BACE 2 or that increase the degradation of this ble endosomal/lysosomal location.
enzyme [19]. A common observation in the majority of the studies of
β OLIGOMERS IN DS
SOLUBLE Aβ intracellular Aβ in DS is the early age of onset; both infants
and children with DS accumulate intracellular Aβ. In addi-
Once Aβ is cleaved from APP it may first appear in solu- tion, intracellular Aβ is consistently observed prior to the
ble form either within neurons or in the extracellular space. accumulation of extracellular Aβ deposits [38], which paral-
Changes in the levels of soluble Aβ measured biochemically lels reports in transgenic mouse models of AD [44, 45].
and from peripheral samples may allow for early detection of These findings suggest that prior to extracellular Aβ deposi-
APP processing abnormalities in people with DS [24]. tion there is an accumulation of intracellular Aβ within neu-
Higher levels of soluble Aβ are observed in DS fetal tissue rons in people with DS at a much earlier age than in the gen-
than in tissue from controls [25]. Specific soluble forms of eral population. Thus, intracellular Aβ accumulation may be
Aβ include different conformations of the peptide such as important for development and occurs prior to and contrib-
oligomers, protofibrils and Aβ-derived diffusible ligands utes to age-associated extracellular Aβ deposition. The ac-
(ADDLs) [26, 27]. Soluble species of Aβ may accumulate cumulation of neuronal Aβ may be associated with caspase
prior to extracellular Aβ in DS and may be more important cleavage products leading to increased apoptosis [46], which
than extracellular Aβ in causing neuronal dysfunction (re- in turn may partially account for observed brain atrophy and
viewed in [24]). Despite the fact that extensive extracellular neuronal loss.
Aβ accumulation is a feature of AD in both the general
population and DS, inconsistent associations between Aβ
Aβ PLAQUES
and dementia severity have been reported [28]. The hypothe-
sis that brain amyloid is the cause of dementia in sporadic While people with DS have an age-dependent increased
AD has also been questioned [29]. Transgenic mice overex- risk for developing dementia, virtually all adults with people
pressing mutant human APP show signs of cognitive dys- with DS over 40 years of age have sufficient neuritic plaques
function prior to the accumulation of insoluble and extracel- and neurofibrillary tangles for a neuropathologically based
lular Aβ [30], suggesting that earlier more soluble forms of diagnosis of AD [47-49]. Senile plaques contain the β-
Aβ may be important [31]. Aβ oligomers may be critically amyloid (Aβ) peptide that is derived from a longer precursor
important in causing neuronal dysfunction prior to overt neu- protein, β-amyloid precursor protein (APP), the gene for
ron loss [32]. Thus, there has been a shift to study earlier and which is on chromosome 21. The most common form of DS,
possibly more toxic species of Aβ. Both biochemical and trisomy 21, leads to the overexpression of APP [10]. Thus,
immunohistochemical experiments reveal significant many neurobiological studies in aged DS cases have focused
amounts of oligomeric Aβ in the AD brain [33-36]. Oli- primarily on APP processing and the temporal events in Aβ
gomeric Aβ may play a critical role in causing neuron dys- pathogenesis [24, 50] based upon the general hypothesis that
function during both development and aging in DS and is a Aβ is thought to be a causative factor in AD pathogenesis
key area requiring further study and has yet to be fully char- and that overexpression of APP may lead to the elevated
acterized in DS brain. levels of Aβ in DS [51-53].
Cerebral Aβ deposition occurs decades earlier in people
INTRACELLULAR Aβ IN DS with DS compared to aged control and AD brain [10, 50].
Although a large amount of Aβ exists in a soluble form, Extracellular Aβ accumulation in diffuse plaques appears to
insoluble deposits also begin to progressively form over consistently deposit after the age of 30 years [47], although
time. However, the subcellular location for these events is widespread diffuse Aβ42 plaques have also been observed in
less well understood, particularly in DS. Gyure et al (2001) brain sections from DS individuals in their teens and 20’s
report intracellular Aβ1-40 but not Aβ1-42 [37]. In contrast, [54] (Fig. 1, 2A). It is also apparent that diffuse plaques pre-
other studies report intracellular Aβ1-42 but not Aβ1-40 [38, cede neuritic plaques with age in the cortex. In DS, Aβ42
39], which in one study was clearly distinguished from intra- diffuse plaques precede fibrillar senile plaques containing
cellular APP immunoreactivity [38]. A report by Hirayama dystrophic neurites and the formation of neurofibrillary tan-
et al. found neither Aβ1-40 nor Aβ1-42 but observed intra- gles, and Aβ42 plaques are more prevalent than Aβ40
cellular Aβ1-43 [40]. Each length of Aβ has different prop- plaques at all ages [54, 55]. Interestingly, diffuse Aβ42 is
erties with Aβ1-40 more rapidly degraded within lysosomes deposited in DS cerebellum and striatum in the third decade
than the longer, more toxic Aβ1-42/43 [41, 42]. The reasons of life, but fibrillar plaques are rarely observed in these brain
for observations of different length Aβ species in intracellu- areas in older DS brain [56], suggesting regional effects on
lar deposits in each study may be due in part to technical plaque maturation. Overall, senile plaques progress in the
differences. same brain regions and cortical layers as in AD, however,
20 Current Alzheimer Research, 2016, Vol. 13, No. 1 Head et al.

Fig. (1). Aβ42 immunostaining in the frontal cortex of DS autopsy cases ranging in age from (A, B) 12 years, (C, D) 15 years, (E, F) 21 years
and (G, H) 61 years. Note the extensive diffuse plaque deposition in younger cases compared with compared with cored and compacted
plaques, subpial plaques and vascular amyloid in the older DS brain. (C42 pAb kindly provided by Dr. Takaomi Saido at the RIKEN Brain
Institute, Japan and described in [38]). Plaques denoted by an asterisk at low magnification on the left (A, C, E, G) are shown at high magni-
fication on the right (B, D, F, H). Scale bars, 100 microns.

plaque density is higher in DS brain [57]. In addition, the Between the ages of 30 and 40 years, neuropathology
presence of an Apo E ε4 allele doubles the cerebral amyloid rapidly accumulates until it reaches levels sufficient for a
plaque burden and shortens life span in DS [58]. Apo E pro- diagnosis of AD in people with DS by 40 years [49]. In fact,
tein, a possible chaperone for Aβ deposition, is detected there is an exponential rise in AD pathology, and specifically
early in plaques in DS brain [54]. Aβ measured biochemically after the age of 40 years [16],
AD Neuropathology in DS Current Alzheimer Research, 2016, Vol. 13, No. 1 21

suggesting an acceleration phase to disease development. appears to consistently occur in younger adults with DS be-
There are reports of younger individuals with AD pathology, tween 13-42 years [81]. Other studies also report much lower
including plaques and tangles, although typically, not of suf- frequency of atheroma [82, 83]. Cerebral amyloid angiopa-
ficient extent for a neuropathology diagnosis of the disease thy (CAA) is the common term used to define the deposition
[54, 59, 60]. The source of extracellular Aβ is thought to be of amyloid in the walls of medium- and small-size leptomen-
mainly neuronal and in autopsy studies of younger people ingeal and cortical arteries, arterioles and, less frequently,
with DS shows that as young as 3 years, intracellular Aβ can capillaries and veins. CAA can lead to micro and macro
be observed [38, 61]. Intracellular Aβ is localized to en- hemorrhages [84]. CAA is consistently observed in older
dosomes, intracellular organelles responsible for degrading individuals with DS (>55 yrs - [85-87] and also contains
and turning over proteins within cells [61, 62]. post-translationally modified Aβ [68]. Interestingly, there are
reports of CAA in DS that is associated with extensive
POST-TRANSLATIONALLY MODIFIED Aβ hemorrhages [87-91] in some studies but not in others [85,
86]. Brain Aβ40 (typically associated with CAA – Fig. 2B)
Over time, extracellular Aβ with the N-terminus starting rises exponentially with age in DS [36]. Thus, adults with
at Asp 1 in DS can be post-translationally modified by iso- DS represent an important cohort to study CVD co-
merization [63], racemization [64] and oxidation [65]. In morbidities because of their unique characteristics:
addition, an N-terminally truncated form of Aβ is generated atheroma-free model and lower blood pressure but with
by degradation of the first 2 amino acids followed by cycli- significant cerebral amyloid angiopathy (CAA).
zation of the newly formed N-terminus by glutaminyl cy-
clase, resulting in degradation-resistant, highly toxic aggre- There are currently no systematic reports of CVD as a
gates of pyroglutamate-3 Aβ that deposit into plaques and function of aging, cognition, dementia or WM integrity in
blood vessel walls in AD and DS brain [54, 66-68]. Mehta DS. It is known, based upon autopsy studies, that there
and colleagues recently reported increased levels of pyroglu- should be significant WMH, CAA and CVD that could be
tamate-3 Aβ in plasma of older DS people compared to non- captured and quantified by the appropriate MR imaging pro-
DS people with and without developmental disabilities [69]. tocols. In terms of designing future clinical trials, character-
Pyroglutamate-11 Aβ, a minor species, is also detected in izing the age of onset and extent of CVD in adults with DS
some plaque cores and vascular amyloid in DS brain [70, will be critical given that CVD is mediated to large extent by
71]. Unmodified Aβ11-40/42 is also elevated in DS brain, lifestyle factors that are amenable to intervention. Individu-
possibly due to the overexpression of BACE 2, a gene en- als with hypertension, high cholesterol, obesity, type II dia-
coded on chromosome 21 [71]. In addition, the APP P3 pep- betes, show a higher risk of CVD (reviewed in: [92]).
tide starting at the non-amyloidogenic α-secretase site
(Aβ17leu) can be observed readily in DS brain extracts [72] ENZYMES INVOLVED WITH Aβ β DEGRADATION
and cerebellar plaques [73]. These modifications may reflect AND CLEARANCE IN THE BRAIN
mechanisms of Aβ production and could potentially serve as
Once Aβ begins to accumulate either in soluble or in-
possible chronobiological age markers for individual depos-
soluble form, several enzymes in the brain may be involved
its and Alzheimer’s disease progression.
with subsequent degradation and clearance. These Aβ clear-
ing enzymes include insulin degrading enzyme (IDE), ne-
CEREBROVASCULAR Aβ PATHOLOGY IN DS prilysin, and tissue plasminogen activator [93-95]. In DS,
A key contributor to sporadic AD, the role of cerebrovas- two components leading to increased production of Aβ are
cular disease (CVD) has been virtually unexplored in DS. present in triplicate and include APP and BACE2. However,
The CVD contribution to AD is increasingly being recog- despite life-long overexpression of these two proteins, full
nized as a critical comorbidity that accelerates the age of blown AD neuropathology is not consistently observed until
onset of dementia and also leads to a faster progression of after age 40 years. Thus, Aβ may be cleared or degraded in
the disease [74]. Estimates of a mixed etiology of AD with the DS brain by the activity of Aβ degrading enzymes. Ne-
CVD range from 5.7-45% in autopsy cases from the general prilysin protein concentration is increased in DS and corre-
population [75]. CVD can serve as a “second hit” necessary lates with levels of insoluble Aβ [17]. Considering the thera-
for clinical signs of dementia, particularly when significant peutic potential of enhancing Aβ degradation and clearance,
Aβ is present in the brain [76]. In addition, evidence from this represents a major gap in our knowledge in the study of
Rotterdam Scan Study suggests a link between WM integrity development and aging in people with DS.
and the number of cerebral microbleeds in older nonde-
mented individuals (>60 years)[77]): individuals with higher CSF Aβ
numbers of microbleeds had lower fractional anistropy.
There are few studies of CSF Aβ in DS. In children with
Thus, CVD may be a second major contributor to cognitive
DS ranging in age from 6 months to 56 months no differ-
decline and WM integrity, with each potentially being addi-
ences were noted in CSF Aβ when analyzed in a cross sec-
tive [78, 79].
tional study [96]. However, in 2 children followed over 4
DS represents a unique opportunity to study the cere- years, progressively increasing CSF Aβ 1-42 was observed
brovascular features of aging and AD in a setting of more over a 4 year span in 2 of these individuals and in particular
limited systemic vascular risk factors. In a previous study of with CSF Aβ 1-37 and Aβ 1-38, which contrasts with studies
70 adults with DS ranging in age from 40-66 years, there in older individuals and may reflect basal rates of Aβ due to
was an absence of atheroma and lower blood pressure than APP overexpression. In a study of 5 adults with DS with an
similarly aged adults without DS [80]. Lower blood pressure average age of 55.3 years, when AD pathology may be pre-
22 Current Alzheimer Research, 2016, Vol. 13, No. 1 Head et al.

sent, Aβ1-40 did not distinguish DS from “diseased con- 4 years [112]. In people with DS with dementia over 4 years,
trols” that did not have AD. Interestingly, Aβ1-42 was sig- plasma Aβ1-40 was lower and the ratio of Aβ1-42/1-40 was
nificantly decreased in CSF from DS patients [97]. A subse- higher compared to people with dementia lasting less than 4
quent study in 12 persons with DS (mean age 41 years) with years. Schupf and colleagues next reported that increasing
3 showing signs of dementia [98] showed a significant de- levels of plasma Aβ 1-40 and decreased Aβ 1-42 was a good
crease in CSF Aβ1-42 as a function of age in DS (25-60 predictor of conversion to dementia in DS [109]. Indeed,
years). Further, CSF Aβ 1-42 was significantly lower in adults with DS with decreasing plasma Aβ 1-42 over time
older adults with DS (> 40 years) relative to younger adults were 5 times more likely to become demented within 4
with DS(<40 years). years. Similarly, in a separate prospective study of 405 per-
sons with DS, those adults in the highest levels of plasma Aβ
A more recent study of 12 people with DS (mean age 41
years) and 20 healthy controls tested for a battery of CSF 1-42 and Aβ 1-40 also had the highest risk of developing
dementia over, on average, a 4.7 year period of time [113].
peptides that may distinguish the two groups [99]. Although
CSF Aβ 1-42 was not different in DS from controls, it was
correlated with age in DS and decreased with increasing age. TAU PHOSPHORYLATION AND NEUROFIBRIL-
CSF Aβ X-40 was increased in DS compared with controls, LARY TANGLES IN DS
however this may have been due to one individual with DS AD-associated changes such as tau phosphorylation and
who had a high level of AβX-40. Further, increases in aggregation are evident in DS brain, especially in plaque-
sAPPα and sAPPβ were observed in people with DS relative associated dystrophic neurites and intraneuronal neurofibril-
to controls but there was also significant overlap with control lary tangles (NFTs) comprised of the hyperphosphorylated
samples. In a second follow up study, CSF Aβ 1-42 was sig- microtubule-associated tau protein (reviewed in [114, 115]
nificantly decreased in DS with shorter Aβ peptides (includ- (Fig. 2C). One potential contributing factor may be the over-
ing 1-34, 1-38, 1-39) also tending to be lower [100]. Further, expression of the dual-specificity tyrosine phosphorylated
there was a significant negative correlation between CSF Aβ and regulated kinase 1A gene (DYRK1A), which is encoded
1-42 and age (from 21 to 60 years). Overall, there appears to on chromosome 21. DYRK1A phosphorylates tau protein
be a relatively consistent decrease in CSF Aβ 1-42 in older making it a better substrate for GSK3β phosphorylation, and
adults with DS that is consistent with reports in sporadic AD DYRK1A phosphorylates alternate splicing factors leading
[101]. These results suggest that CSF Aβ42 is reduced as to an increase ratio of 3R:4R tau, which is associated with
plaques are formed in the brain parenchyma. neurodegeneration [116]. Consistent with this finding, there
is an increase in the number of DYRK1A-positive and 3R-
PLASMA Aβ positive NFTs in middle-aged and older DS brain compared
to sporadic AD [116]. Another possible contributing factor is
Studies of plasma Aβ have provided variable results with
the overexpression of the regulator of calcineurin-1
respect to distinguishing demented from nondemented indi-
(RCAN1) or calcipressin1 gene, also encoded on chromo-
viduals with DS and aging in some cohorts but more consis-
some 21. RCAN1 is elevated in hippocampus and cerebral
tent outcomes appear in larger cohorts, particularly in pro-
cortex in AD [117]. RCAN1 inhibits calcineurin, and may
spective studies. In cross-sectional studies, plasma Aβ1-42
and Aβ 1-40 are consistently elevated in DS relative to con- enhance tau phosphorylation by lowering calcineurin phos-
phatase activity and increasing GSK3β levels [115]. APP,
trols [102-107] but not in fetal plasma samples [108]. Aging
which is encoded on chromosome 21 and overexpressed in
in DS is associated with increasing levels of plasma Aβ1-40
DS, may also play a role in tau phosphorylation in DS brain
[103, 109] but not in other studies [105, 107]. Plasma Aβ 1-
as Aβ42 upregulates both DYRK1A [118] and RCAN1
42 has been reported to be increased with age in adults over
[119]. Thus, overexpression of all 3 genes in DS may coop-
50 years with DS in one study [106] but this was not repli-
cated in subsequent studies [107, 109-111]. As mentioned erate to drive tau pathology and neurodegeneration.
earlier, post-translationally truncated and modified pyroglu- Early tau pathological changes have been observed in the
tamate-3 Aβ is elevated in plasma from older DS individuals outer molecular layer of hippocampus in middle-aged (30-40
(Mehta et al., 2014). Further analysis of this pathogenic Aβ year-old) DS people, followed later by NFTs in the hippo-
species may reveal a potential biomarker for AD conversion campal CA1 region and subiculum, and neuron loss in the
in DS. entorhinal cortex [120]. In general, NFTs follow a similar
distribution in DS as AD, starting in entorhinal cortex and
In cross sectional studies comparing demented adults
spreading to hippocampus and later neocortex, but at a
with DS to those without dementia, most studies report no
higher density in DS compared to AD brain [57].
differences in plasma Aβ1-40 [102, 105, 107, 111, 112] ex-
cept in one study [113]. However, increased plasma Aβ 1-42
can distinguish demented adults with DS [105] and may im- CSF TAU IN DS
prove predictive models for dementia when included [107]. There are four published reports of CSF tau in DS to our
However, ApoE genotype can influence the outcomes [105, knowledge. The first was in 1999 that described a single
107, 110, 113]. person with DS who was 73 years of age who showed high
Prospective studies of aging and conversion to dementia tau levels relative to controls and to sporadic AD [121]. A
in adults with DS have yielded exciting outcomes. Prasher, second paper published in 2001 by Tapiola and colleagues
Schupf and colleagues reported on a study of 83 nonde- observed increased tau as a function of age in people with
mented and 44 demented adults with DS and compared those DS (21-56 years, average age 41 years) that was significant
with dementia duration of over 4 years compared with under [98]. In children with DS under the age of 56 months, CSF
AD Neuropathology in DS Current Alzheimer Research, 2016, Vol. 13, No. 1 23

tau did not discriminate children between 6-10 months, 19- the brain are the microglial cells. For many years it has been
44 months or 55-56 months of age [96]. Last, in a study of known that microglial cells respond to the amyloid plaques
12 DS individuals ranging in age from 21-60 years and an and neurofibrillary tangles by becoming activated and ex-
average age of 41 years, total tau and phosphorylated tau pressing various cytokines and chemokines [133]. Recently
increased with age [99] in DS although these markers were several genome wide array studies have shown that several
not different in DS when compared with controls. Further, genetic risk factors are associated with inflammation genes
older DS participants (>40 years) had significantly higher [134], most recently the TREM2 gene and the CD33 gene
total tau when compared to younger individuals with DS. [135].
Thus, although there are few studies of CSF tau in DS, the
There are many inflammation genes on chromosome 21,
consensus appears to be increasing levels of total tau.
and that are therefore overexpressed in DS [136]. Neuroin-
flammation is a relatively understudied aspect of DS. Al-
OXIDATIVE DAMAGE AND AGING IN DS though whether inflammation is a life-long phenomenon in
In addition to Aβ and NFT neuropathology, several other DS and/or whether it is exacerbated with age and AD has yet
types of pathology have been reported at different ages that to be fully explored [136]. For example, complement (C1q)
may contribute to the development of dementia either early activation has been reported in a 15 year old person with DS
in the disease or possibly exacerbate cognitive symptoms [60] (although this person also had evidence of neuritic
later in disease. People with DS show higher levels of oxida- plaques) but C1q activation is more consistently observed
tive stress at all ages than people in the general population. after 29 years of age [60, 137] in parallel with the deposition
For example, oxidative damage is increased in prenatal DS of fibrillar Aβ. Griffin and colleagues showed that DS brain
brains compared to non-DS controls [122, 123] and is higher is associated with increased S100β expression and IL-1β
in adult DS brain compared to similarly aged individuals expression [138]. Given that IL-1β is a regulator of many
without DS [36]. Early increases in lipid peroxidation meas- pro-inflammatory processes, upregulation in people with DS
ured by 8,12-iso-iPF2α-VI, is observed in the urine of young could indicate an exacerbated pro-inflammatory response.
subjects (1-15 years) with DS, as compared to age-matched Clearly, based on the triplication of many inflammation as-
controls [124]. Interestingly, the extent of protein carbonyl sociated genes in DS neuroinflammation should be a focus
accumulation, 3-nitrotyrosine and 4-hyroxy-2-trans-noneal of future studies.
(HNE) (all indicators of oxidative damage) does not appear Microglial cells, key mediators of inflammation in the
to increase with age per se in DS but HNE levels were brain, also show morphological and pathological changes in
higher overall in DS [36]. Other types of oxidative damage people over the age of 40 years with DS (Fig. 2D) and may
to proteins, detected using proteomics approaches, show be tightly linked to NFT accumulation [139]. Interestingly, a
select proteins are modified that can contribute to several key lack of microglial activation was also reported in this study
pathogenic pathways [125-127]. Thus, oxidative damage suggesting that the neuroinflammation theory be reconsid-
may be a critical contributor to the development of AD neu- ered both in DS and in AD [139]. It should be noted, how-
ropathology in DS. ever, that the classical markers of microglial activation used
Mitochondria in DS, responsible for producing cellular for immunohistochemical studies do not inform the func-
energy but also for producing reactive oxygen species tional state of the microglial cell. Markers such as MHC-II
(ROS), are dysfunctional and in turn lead to abnormalities in and CD45 are associated more with an anti-inflammatory /
APP processing and enhanced Aβ production [128-130]. wound repair type of microglial response that includes ex-
Indeed mitochondrial dysfunction has been observed in fetal pression of matrix remodeling proteins, IL-4 and IL-10 (re-
DS cells [128]. Mutations in mitochondrial DNA have been viewed in [140]).
related to AD changes in DS as well as the general popula-
tion [131]. Oxidized DNA/RNA is higher in DS and in- NEURON LOSS IN DS
creases in the teens and twenties and is further exacerbated
Neuron loss is seen early in subcortical areas of DS brain
by the presence of Aβ [132]. Thus, some types of oxidative
such as locus coeruleus [141, 142] and basal forebrain nuclei
damage increase with age in DS prior to the development of
[143], as well as entorhinal cortex [144], followed later by
AD neuropathology. These studies suggest that mitochon-
neuron loss in hippocampus and temporal cortex and other
drial dysfunction may be a critical event driving oxidative cortical areas [145]. As in AD, noadrenergic neuron loss in
damage with age in people with DS.
people with DS occurs in the rostral area of the locus coer-
uleus in which neurons project to the cerebral cortex, hypo-
NEUROINFLAMMATION AND AGING IN DS thalamus and forebrain, but not in the caudal region [142].
Neuroinflammation is a now recognized as a key compo- Cholinergic neuron loss in basal forebrain nuclei that project
nent of neurodegenerative disorders and the innate immune to hippocampus and cerebral cortex, such as the nucleus ba-
response in the brain is sophisticated and robust. In AD, neu- salis of Meynert, increases with age and AD pathology in DS
roinflammation has been linked to both the exacerbation of [146]. Thus, it is possible that early synapse loss in AD and
amyloid plaques and neurofibrillary tangles, as well as the DS cortex may reflect retrograde neurodegeneration in sub-
clearance of amyloid plaques. It is thought that such di- cortical nuclei. In addition, while overall entorhinal cortex
chotomous findings can be accounted for by differences in volume was reduced 42% in older DS brain, the number of
the types of neuroinflammation, whether this is due to the intact, NFT-free neurons was reduced by 90% compared to
stimulus or the profile of inflammatory mediators expressed. controls, suggesting that neurofibrillary degeneration was the
The primary mediators of the neuroinflammatory response in main cause for neuron loss in people with DS [144]. How-
24 Current Alzheimer Research, 2016, Vol. 13, No. 1 Head et al.

Fig. (2). Neuropathological features of DS brain. (A) Diffuse plaque labeling using an antibody against Aβ (R1282, gift from Dr. D. Selkoe,
Boston, MA) in the temporal cortex of an individual with DS aged 21 years. (B) Significant cerebral amyloid angiopathy in a 46 year old
male with DS and AD detected by immunostaining of the frontal cortex with Aβ1-40 antibody (arrows), which can be distinguished from
extracellular plaques (arrowheads). (C) Neurofibrillary tangles labeled using the PHF-1 antibody (provided by Dr. Peter Davies- arrows) in
the frontal cortex of a 46 year old male with DS and AD. (D) Immunostaining using IBA-1 for microglial cells shows significant hypertrophy
(arrows) and association with the cerebrovasculature (arrowheads) suggesting neuroinflammation.

ever, Mann and colleagues reported proportionally less over- DS [149]. Interestingly, this pattern was virtually identical to
all neuron loss in older DS brain than sporadic AD brain due non-DS autopsy cases. Corpora amylacea, thought to reflect
to the lower number of neurons in these brain regions in swollen glial processes has also been observed in the frontal
young DS people compared to young controls, and thereby WM of a 32 year old DS case [150]. The most recent study
suggesting that those with DS have less neuronal reserve of WM pathology in DS showed that αB-crystallin, which is
[145]. a member of the heat shock protein family, was increased as
a function of age up to 23 years in DS (older ages were not
WHITE MATTER DEGENERATION IN DS included in the study) [151]. Given the role of αB-crystallin
in acting as a molecular chaperone to prevent aggregation of
In addition to AD cortical pathology, there is some evi- proteins and maintaining proteins in a folding-competent
dence of white matter (WM) degeneration that is more ex- state under stress conditions, this suggests that the DS brain
tensive in DS relative to non-DS autopsy cases or that in- is under potentially chronic levels of stress and that WM is
crease with age and disease [50]. Further, diffusion tensor particularly vulnerable. Indeed, DRYK1a (a gene on chro-
imaging (DTI) studies have shown white matter degradation mosome 21), is expressed at higher levels in people with DS
in non-DS women with a family history of dementia and at compared to controls when WM of the corpus callosum was
least one apolipoprotein E4 allele. While structural imaging examined [152] and may be a contributor to WM vulnerabil-
studies have suggested a greater AD risk in non-DS indi- ity to stress. Thus, in combination, there are several studies
viduals with WM degeneration [147], there are few system- over the past two decades suggesting frontal cortex WM
atic studies of large collections of DS autopsy samples or pathology and degeneration with age in DS.
DTI. Therefore, there are many gaps in our knowledge re-
garding the role of WM pathology and dementia. WM degeneration may be caused by the development of
AD or the aging process but is also strongly associated with
Aβ deposits and APP accumulation has been observed in cerebrovascular disease and associated risks. Although the
the WM of the frontal cortex in DS [85, 148]. Ubiquitin risk of cerebrovascular accident in aging persons with intel-
positive punctuate deposits, that correspond to WM degen- lectual disability is not different from the general population,
eration in the frontal cortex, increases after 21 years of age in
AD Neuropathology in DS Current Alzheimer Research, 2016, Vol. 13, No. 1 25

the number of people with DS included in this study was CONFLICT OF INTEREST
small [153, 154]. In several ways, people with DS may be
The author(s) confirm that this article content has no con-
protected from cerebrovascular incidents given reports of
flict of interest.
generally lower blood pressure [155] and lower incidence of
atherosclerosis [80, 154, 156]. However, Aβ is deposited
along blood vessel walls in people with DS in older ages and ACKNOWLEDGEMENTS
this can lead to cerebrovascular compromise and hemorrhage Funding that supported the authors in the preparation of
[88]. this manuscript includes NIH NICHD R01HD064993 to EH,
NIH grant NS079637 to DMW and Alzheimer’s Association
POSSIBLE SYNAPTIC COMPENSATORY RE- Grant DSADNIP-13-282631 to DMW and EH, NIH
SPONSES IN DS BRAIN R01HD065160, NIH R01AG16573 to ITL and NIH/NIA R01
AG040092 to CAL. The authors thank Dr. Krystyna Wis-
There are over 200 genes on chromosome 21 that may
not only contribute to pathological events observed in the niewski (in memoriam; NY State Institute for Basic Re-
search in Developmental Disabilities, Staten Island, NY) and
aging DS brain, but also may be protective [157]. For exam-
her son, Dr. Thomas Wisniewski (NYU School of Medicine)
ple, in vivo imaging studies of glucose utilization shows in-
for providing archived brain samples from young Down syn-
creased activity in the DS entorhinal cortex prior to the de-
drome individuals to CAL for pathological analyses.
velopment of dementia [158]. Consistent with this observa-
tion are the results of an autopsy study showing a possible
sprouting phenotype in the hippocampus of similarly aged REFERENCES
DS brains [120]. More recently, in a study measuring the [1] Down JLH. Observations on ethnic classification of idiots. Lond
protein levels of synaptophysin and synaptojanin 1, the latter Hosp Rep 3: 259-62 (1866).
is on chromosome 21 and overexpressed in DS, interesting [2] Lejeune J, Gautier M, Turpin R. Etude des chromosomes
somatiques de neuf enfants mongoliens. Comptes Rendus
dissociations from sporadic AD was observed. In DS, despite Hebdomadaires des Seances de L'Academie des Sciences 248:
a reduction of synaptophysin protein levels with AD, similar 1721-22 (1959).
to that seen in sporadic AD, synaptojanin 1 was increased in [3] Roizen NJ, Patterson D. Down's syndrome. Lancet 361(9365):
DS with AD [159]. Further, higher levels of synaptojanin 1 1281-9 (2003).
[4] Lott IT, Dierssen M. Cognitive deficits and associated neurological
were correlated with several measures of Aβ suggesting the complications in individuals with Down's syndrome. Lancet Neurol
two events may be related. It will be interesting in future 9(6): 623-33 (2010).
studies to characterize other proteins with genes on chromo- [5] Glasson EJ, Sullivan SG, Hussain R, Petterson BA, Montgomery
some 21 to determine of other possible compensatory events PD, Bittles AH. The changing survival profile of people with
occur with or precede the development of AD neuropathol- Down's syndrome: implications for genetic counselling. Clin Ge-
netx 62(5): 390-3 (2002).
ogy. Given that there is approximately a decade delay be- [6] Bittles AH, Bower C, Hussain R, Glasson EJ. The four ages of
tween the development of AD pathology and clinical signs Down syndrome. Eur J Public Health 17(2): 221-5 (2007).
of dementia, protective genes may delay impaired neuronal [7] Strauss D, Eyman RK Mortality of people with mental retardation
function in DS. in California with and without Down syndrome, 1986-1991. Am J
Ment Retard 100(6): 643-53 (1996).
[8] Zigman WB. Atypical aging in down syndrome. Dev Disabil Res
FUTURE DIRECTIONS Rev 18(1): 51-67 (2013).
[9] Hartley D, lumenthal T, Carrillo M, DiPaolo G, Esralew L,
Although this review is not exhaustive, there are gaps in Gardiner K, et al. Down syndrome and Alzheimer's disease:
our knowledge regarding the progression of AD neuropa- Common pathways, common goals. Alzheimers Dement 11(6):
thology as a function of age in DS. For example, it is unclear 700-9 (2015).
if the regional and temporal progression of AD neuropathol- [10] Rumble B, Retallack R, Hilbich C, Simms G, Multhaup G, Martins
R, et al. Amyloid A4 and its precursor in Down's syndrome and
ogy in DS is similar or different from sporadic AD. Further, Alzheimer's disease. New Engl J Med 320: 1446-62 (1989).
a detailed examination and systematic description of the dis- [11] Tanzi RE, McClatchey AI, Lamperti ED, Villa-komaroff L,
tribution of neuritic and diffuse plaques and CAA within the Gusella JF, Neve RL. Protease inhibitor domain encoded by an
hippocampus and cortex as well as other brain regions such amyloid protein precursor mRNA associated with Alzheimer's
diase. Nature 333: 528-530 (1988).
as striatum and cerebellum, and how this varies with age and [12] Barger SW, Harmon AD. Microglial activation by Alzheimer
dementia in people with DS, would be helpful in the future. amyloid precursor protein and modulation by apolipoprotein E.
The development of networked collections of autopsy cases, Nature 388(6645): 878-81 (1997).
with standardized collection procedures and including bio- [13] Nunan J, Small DH. Regulation of APP cleavage by a-, b- and g-
chemical analyses will provide significant new advances. It secretases. FEBS Lett 483: 6-10 (2000).
[14] Selkoe DJ. Normal and abnormal biology of the beta-amyloid
will be critical to determine how various pathologies as we precursor protein. Annu Rev Neurosci 17: 489-517 (1994).
discussed here may interact over time to lead to dementia. In [15] Vassar R, Bennett BD, Babu-Khan S, Kahn S, Mendiaz EA, Denis
turn, understanding the progression of events and more fully P, Teplow DB, et al. Beta-secretase cleavage of Alzheimer's
characterizing which pathologies evolve at which ages in amyloid precursor protein by the transmembrane aspartic protease
BACE. Science 286(5440): 735-41 (1999).
people with DS will lead to new therapeutic opportunities, [16] Nistor M, Don M, Parekh M, Sarsoza F, Goodus M, Lopez GE, et
which may include preventative approaches. al. Alpha- and beta-secretase activity as a function of age and beta-
amyloid in Down syndrome and normal brain. Neurobiol Aging
DISCLOSURE 28(10): 1493-506 (2007).
[17] Miners JS, Morris S, Love S, Kehoe PG. Accumulation of
Part of this article has been previously published in Eur J insoluble amyloid-beta in down's syndrome is associated with
Neurodegener Dis 2012; 1(3): 353-364.
26 Current Alzheimer Research, 2016, Vol. 13, No. 1 Head et al.

increased BACE-1 and neprilysin activities. J Alzheimers Dis [40] Hirayama A, Horikoshi Y, Maeda M, Ito M, Takashima S.
23(1): 101-8 (2011). Characteristic developmental expression of amyloid b40,42 and 43
[18] Holler CJ, Webb RL, Laux AL, Beckett TL, Niedowicz DM, in patients with Down syndrome. Brain Develop 25: 180-5 (2003).
Ahmed RR, et al. BACE2 expression increases in human [41] Knauer MF, Soreghan B, Burdick D, Kosmoski J, Glabe CG..
neurodegenerative disease. Am J Pathol 180(1): 337-50 (2012). Intracellular accumulation and resistance to degradation of the
[19] Webb RL, Murphy MP. beta-Secretases, Alzheimer's Disease, and Alzheimer amyloid A4/beta protein. Proc Natl Acad Sci USA
Down Syndrome. Curr Gerontol Geriatr Res 2012: 362839 (2012). 89(16): 7437-41 (1992).
[20] Hussain I, Powell DJ, Howlett DR, Chapman GA, Gilmour L, [42] Yang AJ, Chandswangbhuvana D, Shu T, Henschen A, Glabe CG.
Murdock PR, et al. ASP1 (BACE2) cleaves the amyloid precursor Intracellular accumulation of insoluble, newly synthesized abetan-
protein at the beta-secretase site. Mol Cell Neurosci 16(5): 609-19 42 in amyloid precursor protein-transfected cells that have been
(2000). treated with Abeta1-42. J Biol Chem 274(29): 20650-6 (1999).
[21] Barbiero L, Benussi L, Ghidoni R, Alberici A, Russo C, Schettin [43] Glabe, C. Intracellular mechanisms of amyloid accumulation and
G, et al. BACE-2 is overexpressed in Down's syndrome. Exp pathogenesis in Alzheimer's Disease. J Mol Neurosci 17: 137-145
Neurol 182: 335-45 (2003). (2001).
[22] Motonaga K, Itoh, M, Becker LE, Goto Y, Takashima S. Elevated [44] Oddo S, Caccamo A, Shepherd JD, Murphy MP, Golde TE, Kayed
expression of beta-site amyloid precursor protein cleaving enzyme R, et al. Triple-transgenic model of Alzheimer's disease with
2 in brains of patients with Down syndrome. Neurosci Lett 326: 64- plaques and tangles: Intracellular Ab and synaptic dysfunction.
6 (2002). Neuron 39: 409-421 (2003).
[23] Cheon MS, Dierssen M, Kim SH, Lubec G. Protein expression of [45] Wirth O, Multhaup G, Czech C, Blanchard V, Moussaoui S, Tremp
BACE1, BACE2 and APP in Down syndrome brains. Amino Acids G, et al. Intraneuronal Ab accumulation precedes plaque formation
35(2): 339-43 (2008). in b-amyloid precursor protein and presenilin-1 double-transgenic
[24] Head E, Lott IT. Down syndrome and beta-amyloid deposition. mice. Neurosci Lett 306: 116-120 (2001).
Curr Opin Neurol 17(2): 95-100 (2004). [46] Head E, Lott IT, Cribbs DH, Cotman CW, Rohn TT. Beta-amyloid
[25] Teller JK, Russo C, DeBusk LM, Angelini G, Zacceo D, Dagna- deposition and neurofibrillary tangle association with caspase
Bricarelli F, et al. Presence of soluble amyloid b-peptide precedes activation in Down syndrome. Neurosci Lett 330(1): 99-103
amyloid plaque formation in Down's syndrome. Nat Med 2(1): 93- (2002).
5 (1996). [47] Mann DMA, Esiri MM. The pattern of acquisition of plaques and
[26] Walsh DM, Klyubin I, Fadeeva JV, Rowan MJ, Selkoe DJ, et al., tangles in the brains of patients under 50 years of age with Down's
Amyloid-beta oligomers: their production, toxicity and therapeutic syndrome. J Neurol Sci 89: 169-179 (1989).
inhibition. Biochem Soc Trans 30(4): 552-7 (2002). [48] Wisniewski K, Howe J, Williams G, Wisniewski HM. Precocious
[27] Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, aging and dementia in patients with Down's syndrome. Biological
Liosatos M, et al., Diffusable, nonfibrillar ligands derived from Psychiatry 13(5): 619-627 (1978).
Ab1-42 are potent central nervous system neurotoxins. Proc Natl [49] Wisniewski K, Wisniewski H, Wen G. Occurrence of
Acad Sci 95: 6448-53 (1998). neuropathological changes and dementia of Alzheimer's disease in
[28] Cummings BJ. Plaques and tangles: searching for primary events in Down's syndrome. Ann Neurol 17: 278-282 (1985).
a forest of data. Neurobiol Aging 18(4): 358-62 (1997). [50] Head E, Powell D, Gold BT, Schmitt FA.. Alzheimer's disease in
[29] Struble RG, Ala T, Patrylo PR, Brewer GJ, Yan XX, et al. Is brain Down syndrome. European Journal of Neurodegenerative Dis 1(3):
amyloid production a cause or a result of dementia of the 353-364 (2012).
Alzheimer's type? J Alzheimers Dis 22(2): 393-9 (2010). [51] Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer's
[30] Van Dam D, D'Hooge R, Staufenbiel M, Van Ginneken C, Van disease: progress and problems on the road to therapeutics. Science
Meir F, De Deyn PP. Age-dependent cognitive decline in APP23 297(5580): 353-6 (2002).
model precedes amyloid deposition. Eur J Neurosci 17: 388-96 [52] Rovelet-Lecrux A, Hannequin D, Raux G, Le Meur N, Laquerrière
(2003). A, Vital A, Dumanchin C, et al. APP locus duplication causes
[31] Westerman MA, Cooper-Blacketer D, Mariash A, Kotilinek L, autosomal dominant early-onset Alzheimer disease with cerebral
Kawarabayashi T, Younkin LH, et al. The relationship between Ab amyloid angiopathy. Nat Genet 38(1): 24-6 (2006).
and memory in the Tg2576 mouse model of Alzheimer's disease. J [53] Theuns, J., et al., Promoter mutations that increase amyloid
Neurosci 22(5): 1858-67 (2002). precursor-protein expression are associated with Alzheimer
[32] Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe disease. Am J Hum Genet 78(6): 936-46 (2006).
MS, et al. Naturally secreted oligomers of amyloid beta protein [54] Lemere CA, Blusztajn JK, Yamaguchi H, Wisniewski T, Saido TC,
potently inhibit hippocampal long-term potentiation in vivo. Nature Selkoe DJ, et al. Sequence of deposition of heterogeneous amyloid
416(6880): 535-9 (2002). beta-peptides and APOE in Down Syndrome: Implications for
[33] Kuo YM, Emmerling MR, Vigo-Pelfrey C, Kasunic TC, initial events in amyloid plaque formation. Neurobiol Dis 3: 16-32
Kirkpatrick JB, Murdoch GH, et al. Water-soluble Ab(N-40, N-42) (1996).
oligomers in normal and Alzheimer disease brains. J Biol Chem [55] Iwatsubo T, Mann DM, Odaka A, Suzuki N, Ihara Y. Amyloid beta
271: 4077-4081 (1996). protein (A beta) deposition: A beta 42(43) precedes A beta 40 in
[34] Kayed R, Head E, Thompson JL, McIntire TM, Milton SC, Cotman Down syndrome. Ann Neurol 37(3): 294-9 (1995).
CW, et al. Common structure of soluble amyloid oligomers implies [56] Mann DM, Iwatsubo T. Diffuse plaques in the cerebellum and
common mechanism of pathogenesis. Science 300: 486-489 corpus striatum in Down's syndrome contain amyloid beta protein
(2003). (A beta) only in the form of A beta 42(43). Neurodegeneration
[35] Gong Y, Chang L, Viola KL, Lacor PN, Lambert MP, Finch CE, 5(2): 115-20 (1996).
et al. Alzheimer's disease-affected brain: Presence of oligomeric [57] Hof PR, Bouras C, Perl DP, Sparks DL, Mehta N, Morrison JH.
Ab ligands (ADDLs) suggests a molecular basis for reversible Age-related distribution of neuropathologic changes in the cerebral
memory loss. Proc Natl Acad Sci 100(18): 10417-10422 (2003). cortex of patients with Down's syndrome. Arch Neurol, 52: 379-
[36] Cenini, G., et al. Association between frontal cortex oxidative 391 (1995).
damage and beta-amyloid as a function of age in Down syndrome. [58] Hyman BT, West HL, Rebeck GW, Lai F, Mann DM.
Biochim Biophys Acta 1822(2): 130-8 (2012). Neuropathological changes in Down's syndrome hippocampal
[37] Gyure KA, Durham R, Stewart WF, Smialek JE, Troncoso JC. formation. Effect of age and apolipoprotein E genotype. Arch
Intraneuronal Ab -Amyloid precedes development of amyloid Neurol 52: 373-378 (1995 ).
plaques in Down syndrome. Arch Pathol Lab Med 125: 489-492 [59] Leverenz JB, Raskind MA. Early amyloid deposition in the medial
(2001). temporal lobe of young Down syndrome patients: A regional
[38] Mori C, Spooner ET, Wisniewsk KE, Wisniewski TM, Yamaguch quantitative analysis. Experimental Neurology 150: 296-304
H, Saido TC, Tolan DR, et al., Intraneuronal Abeta42 accumulation (1998).
in Down syndrome brain. Amyloid 9(2): 88-102 (2002). [60] Stoltzner SE, Grenfell TJ, Mori C, Wisniewski KE, Wisniewski
[39] Gouras GK, Tsai J, Naslund J, Vincent B, Edgar M, Checler F, et TM, Selkoe DJ, et al. Temporal accrual of complement proteins in
al., Intraneuronal Abeta42 accumulation in human brain. Am J amyloid plaques in Down's syndrome with Alzheimer's disease.
Pathol 156(1): 15-20 (2000). Am J Patho 156(2): 489-499 (2000).
AD Neuropathology in DS Current Alzheimer Research, 2016, Vol. 13, No. 1 27

[61] Cataldo AM, Petanceska S, Terio NB, Peterhoff CM, Durham R, [81] Rodrigues, A.N., et al., Stiffness of the large arteries in individuals
Mercken M, et al. Abeta localization in abnormal endosomes: with and without Down syndrome. Vasc Health Risk Manag 7: p.
association with earliest Abeta elevations in AD and Down 375-81 (2011).
syndrome. Neurobiol Aging 25(10): 1263-72 (2004). [82] Brattstrom L, Englund E, Brun A. Does Down syndrome support
[62] Cataldo AM, Peterhoff CM, Troncoso JC, Gomez-Isla T, Hyman homocysteine theory of arteriosclerosis. Lancet 14(8529): 391-392
BT, Nixon RA. Endocytic pathway abnormalities precede amyloid (1987).
beta deposition in sporadic Alzheimer's disease and Down [83] Yla-Herttuala S, Luoma J, Nikkari T, Kivimaki T. Down's
syndrome: differential effects of APOE genotype and presenilin syndrome and atherosclerosis. Atherosclerosis 76(2-3): 269-272
mutations. Am J Pathol 157: 277-286 (2000). (1989).
[63] Fonseca MI, Head E, Velazquez P, Cotman CW, Tenner AJ. The [84] Vinters HV. Cerebral amyloid angiopathy. A critical review. Stroke
presence of isoaspartic acid in beta-amyloid plaques indicates 18(2): 311-24 (1987).
plaque age. Exp Neurol 157(2): 277-88(1999). [85] Ikeda S, Tokuda T, Yanagisawa N, Kametani F, Ohshima T, Allsop
[64] Azizeh BY, Head E, Ibrahim MA, Torp R, Tenner AJ, Kim RC, D. et al., Variability of beta-amyloid protein deposited lesions in
Lott IT, et al. Molecular dating of senile plaques in the brains of Down's syndrome brains. Tohoku J Exp Med 1174(3): 189-98
individuals with Down syndrome and in aged dogs. Exp Neurol (1994).
163(1): 111-22 (2000). [86] Lai F, Williams MD. A prospective study of Alzheimer Disease in
[65] Head E, Garzon-Rodriguez W, Johnson JK, Lott IT, Cotman CW, Down Syndrome. Arch Neurol 46: 849-853 (1989).
Glabe C. Oxidation of Abeta and plaque biogenesis in Alzheimer's [87] Belza MG, Urich H. Cerebral amyloid angiopathy in Down's
disease and Down syndrome. Neurobiol Dis 8(5): 792-806 (2001). syndrome. Clin Neuropathol 5(6): 257-60 (1986).
[66] Saido TC, Iwatsubo T, Mann DM, Shimada H, Ihara Y, [88] Mendel T, Bertrand E, Szpak GM, Stępień T, Wierzba-Bobrowicz
Kawashima S. Dominant and differential deposition of distinct T. Cerebral amyloid angiopathy as a cause of an extensive brain
beta-amyloid peptide species, A beta N3 (pE), in senile plaques. hemorrhage in adult patient with Down's syndrome - a case report.
Neuron 14: 457 (1995). Folia Neuropathol 48(3): 206-11 (2010).
[67] Schilling S, Hoffmann T, Manhart S, Hoffmann M, Demuth HU. [89] Naito KS, Sekijima Y, Ikeda S. Cerebral amyloid angiopathy-
Glutaminyl cyclases unfold glutamyl cyclase activity under mild related hemorrhage in a middle-aged patient with Down's
acid conditions. FEBS Lett 563(1-3): 191-6 (2004). syndrome. Amyloid 15(4): 275-7 (2008).
[68] Frost JL, Le KX, Cynis H, Ekpo E, Kleinschmidt M, Palmour RM, [90] McCarron MO, Nicoll JA, Graham DI. A quartet of Down's
et al. Pyroglutamate-3 amyloid-beta deposition in the brains of syndrome, Alzheimer's disease, cerebral amyloid angiopathy, and
humans, non-human primates, canines, and Alzheimer disease-like cerebral haemorrhage: interacting genetic risk factors. J Neurol
transgenic mouse models. Am J Pathol 183(2): 369-81 (2013). Neurosurg Psychiatry 65(3): 405-6 (1998).
[69] Mehta, P., et al. Incrased Plasma Pyroglutamate-3 Amyloid Beta [91] Donahue JE, Khurana JS, Adelma L S. Intracerebral hemorrhage in
Protein Levels in Older Persons with Down Syndrome (DS). two patients with Down's syndrome and cerebral amyloid
Neurology 82(10): S38.004 (2014). angiopathy. Acta Neuropathol 95(2): 213-6 (1998).
[70] Iwatsubo T, Saido TC, Mann DM, Lee VM, Trojanowski JQ. Full- [92] Reitz C, Mayeux R. Alzheimer disease: epidemiology, diagnostic
length amyloid-beta (1-42(43)) and amino-terminally modified and criteria, risk factors and biomarkers. Biochem Pharmacol 88(4):
truncated amyloid-beta 42(43) deposit in diffuse plaques. Am J 640-51 (2014).
Pathol 149(6): 1823-30 (1996). [93] Carson JA, Turner AJ. Beta-amyloid catabolism: roles for
[71] Liu K, Solano I, Mann D, Lemere C, Mercken M, Trojanowski JQ, neprilysin (NEP) and other metallopeptidases? J Neurochem 81(1):
Lee VM. Characterization of Abeta11-40/42 peptide deposition in 1-8 (2002).
Alzheimer's disease and young Down's syndrome brains: [94] Melchor JP, Pawlak R, Stickland S. The tissue plasminogen
implication of N-terminally truncated Abeta species in the activator-plasminogen proteolytic cascade accelerates amyloid-b
pathogenesis of Alzheimer's disease. Acta Neuropathol 112(2): (Ab) degradation and inhibits Ab-induced neurodegeneration. J
163-74 (2006). Neurosci 23(26): 8867-8871 (2003).
[72] Saido TC, Yamao-Harigaya W, Iwatsubo T, Kawashima S. Amino- [95] Selkoe DJ. Clearing the brain's amyloid cobwebs. Neuron 32: 177-
and carboxyl-terminal heterogeneity of b-amyloid peptides 180 (2001).
deposited in human brain. Neurosci Lett 215: 173-176 (1996). [96] Englund H, Annerén G, Gustafsson J, Wester U, Wiltfang J,
[73] Lalowski M, Golabek A, Lemere CA, Selkoe DJ, Wisniewski HM, Lannfelt L, et al. Increase in beta-amyloid levels in cerebrospinal
Beavis RC, et al. The "nonamyloidogenic" p3 fragment (amyloid fluid of children with Down syndrome. Dement Geriatr Cogn
beta17-42) is a major constituent of Down's syndrome cerebellar Disord 24(5): 369-74 (2007).
preamyloid. J Biol Chem 271(52): 33623-31 (1996). [97] Tamaoka A, Sekijima Y, Matsuno S, Tokuda T, Shoji S, Ikeda
[74] Snyder HM, Corriveau RA, Craft S, Faber JE, Greenberg SM, SI.et al. Amyloid beta protein species in cerebrospinal fluid and in
Knopman D, et al. Vascular contributions to cognitive impairment brain from patients with Down's syndrome. Ann Neurol 46(6): 933
and dementia including Alzheimer's disease. Alzheimers Dement (1999).
11(6): 710-7 (2015) [98] Tapiola T, Soininen H, Pirttila T. CSF tau and Ab42 levels in
[75] Jellinger KA. Pathology and pathogenesis of vascular cognitive patients with Down's syndrome. Neurology 56: 979-980 (2001).
impairment-a critical update. Front Aging Neurosci 5: 17 (2013). [99] Portelius E, Soininen H, Andreasson U, Zetterberg H, Persson R,
[76] Provenzano FA Muraskin J, Tosto G, Narkhede A, Wasserman BT, Karlsson G, Blennow K, et al, Exploring Alzheimer molecular
Griffith EY, Narkhede A, et al. White matter hyperintensities and pathology in Down's syndrome cerebrospinal fluid. Neurodegener
cerebral amyloidosis: necessary and sufficient for clinical Dis 14(2): 98-106 (2014).
expression of Alzheimer disease? JAMA Neurol 70(4): 455-61 [100] Portelius E, Hölttä M, Soininen H, Bjerke M, Zetterberg H,
(2013). Westerlund A, Herukka SK, et al. Altered cerebrospinal fluid levels
[77] Akoudad S, de Groot M, Koudstaal PJ, van der Lugt A, Niessen of amyloid beta and amyloid precursor-like protein 1 peptides in
WJ, Hofman A, et al. Cerebral microbleeds are related to loss of Down's syndrome. Neuromolecular Med 16(2): 510-6 (2014).
white matter structural integrity. Neurology 81(22): 1930-7 (2013). [101] Blennow K, Zetterberg H. The application of cerebrospinal fluid
[78] Reed BR, Mungas DM, Kramer JH, Ellis W, Vinters HV, Zarow C, biomarkers in early diagnosis of Alzheimer disease. Med Clin
et al. Profiles of neuropsychological impairment in autopsy-defined North Am 97(3): 369-76 (2013).
Alzheimer's disease and cerebrovascular disease. Brain 130(Pt 3): [102] Tokuda T, Fukushima T, Ikeda S, Sekijima Y, Shoji S, Yanagisawa
731-9 (2007). N, Tamaoka A, et al. Abeta1-42(43) are elevated in Down's
[79] Lo RY, JagustWJ, I. Alzheimer's Disease Neuroimaging, Vascular syndrome. Ann Neurol 41(2): 271-3 (1997).
burden and Alzheimer disease pathologic progression. Neurology [103] Mehta PD, Dalton AJ, Mehta SP, Kim KS, Sersen EA, Wisniewski
79(13): 1349-55 (2012). HM. Increased plasma amyloid b protein 1-42 levels in Down
[80] Murdoch JC, Rodger JC, Rao SS, Fletcher CD. Dunningham, syndrome. Neuroscience Lett 241: 13-16 (1998).
M.G., Down's syndrome: an atheroma-free model? . Br Med J [104] Cavani S, Tamaoka A, Moretti A, Marinelli L, Angelini G, Di
2(6081): 226-228 (1977). Stefano S, et al. Plasma levels of amyloid b 40 and 42 are
independent from ApoE genotype and mental retardation in Down
syndrome. Am J Med Genet 95: 224-228 (2000).
28 Current Alzheimer Research, 2016, Vol. 13, No. 1 Head et al.

[105] Schupf, N., et al., Elevated plasma amyloid beta-peptide 1-42 and [130] Helguera P, Seiglie J, Rodriguez J, Hanna M, Helguera G,
onset of dementia in adults with Down syndrome. Neurosci Lett Busciglio J. Adaptive downregulation of mitochondrial function in
301(3): 199-203 (2001). down syndrome. Cell Metab 17(1): 132-40 (2013).
[106] Mehta PD, et al. Plasma amyloid beta protein 1-42 levels are [131] Coskun PE, Wyrembak J, Derbereva O, Melkonian G, Doran E,
increased in old Down Syndrome but not in young Down Lott IT, Head E, et al. Systemic mitochondrial dysfunction and the
Syndrome. Neurosci Lett 342(3): 155-8 (2003). etiology of Alzheimer's disease and down syndrome dementia. J
[107] Head E, et al. Plasma amyloid-beta as a function of age, level of Alzheimers Dis 20(2): S293-310 (2010).
intellectual disability, and presence of dementia in Down [132] Nunomura A, Perry G, Pappolla M, Friedland RP, Hirai K, Chiba
syndrome. J Alzheimers Dis 23(3): 399-409 (2011). S, et al. Neuronal oxidative stress precedes amyloid-b deposition in
[108] Bartha JL, Soothill PW. Plasma amyloid beta protein 1-42 levels in Down syndrome. J Neuropath Exp Neurol 59(11): 1011-1017
fetuses with Down syndrome. Early Hum Dev 81(4): 351-4 (2005). (2000).
[109] Schupf, N., et al., Change in plasma Ass peptides and onset of [133] Akiyama, H., Barger S, Barnum S, Bradt B, Bauer J, Cole GM,
dementia in adults with Down syndrome. Neurology 75(18): 1639- Cooper NR, et al. Inflammation and Alzheimer's disease.
44 (2010). Neurobiol Aging 21(3): 383-421 (2000).
[110] Matsuoka Y, et al. The relationship of plasma Abeta levels to [134] Di Bona D, Plaia A, Vasto S, Cavallone L, Lescai F, Franceschi C,
dementia in aging individuals with down syndrome. Alzheimer Dis et al. Association between the interleukin-1beta polymorphisms
Assoc Disord 23(4): 315-8 (2009). and Alzheimer's disease: a systematic review and meta-analysis.
[111] Jones EL, et al. Amyloid beta concentrations in older people with Brain Res Rev 59(1): 155-63 (2008).
Down syndrome and dementia. Neurosci Lett 451(2): 162-4 (2009). [135] Karch CM, Goate AM. Alzheimer's disease risk genes and
[112] Prasher VP, et al. Plasma beta-amyloid and duration of Alzheimer's mechanisms of disease pathogenesis. Biol Psychiatry 77(1): 43-51
disease in adults with Down syndrome. Int J Geriatr Psychiatry (2015).
(2009). [136] Wilcock DM. Neuroinflammation in the aging down syndrome
[113] Coppus AM, et al., Plasma beta amyloid and the risk of brain; lessons from Alzheimer's disease. Curr Gerontol Geriatr Res
Alzheimer's disease in Down syndrome. Neurobiol Aging 33(9): 2012: 170276 (2012).
1988-94 (2012. [137] Head E, Azizeh BY, Lott IT, Tenner AJ, Cotman CW, Cribbs DH,
[114] Iqbal K, et al., Mechanisms of tau-induced neurodegeneration. et al. Complement association with neurons and beta-amyloid
Acta Neuropathol 118(1): 53-69 (2009). deposition in the brains of aged individuals with Down Syndrome.
[115] Cardenas AM, et al. Role of tau protein in neuronal damage in Neurobiol Dis 8(2): 252-65 (2001).
Alzheimer's disease and Down syndrome. Arch Med Res 43(8): [138] Griffin WS, Stanley LC, Ling C, White L, MacLeod V, Perrot LJ,
645-54 (2012). et al. Brain interleukin I and S-100 immunoreactivity are elevated
[116] Wegiel, J., et al., Link between DYRK1A overexpression and in Down syndrome and Alzheimer's disease. Proc Natl Acad Sci
several-fold enhancement of neurofibrillary degeneration with 3- USA 86: 7611-7615 (1989).
repeat tau protein in Down syndrome. J Neuropathol Exp Neurol [139] Xue QS, Streit WJ. Microglial pathology in Down syndrome. Acta
70(1): 36-50 (2011). Neuropathol 122(4): 455-66 (2011).
[117] Harris CD, Ermak G, Davies KJ. RCAN1-1L is overexpressed in [140] Colton C, Wilcock DM. Assessing activation states in microglia.
neurons of Alzheimer's disease patients. FEBS J 274(7): 1715-24 CNS Neurol Disord Drug Targets 9(2): 174-91 (2010).
(2007). [141] Mann DM. The locus coeruleus and its possible role in ageing and
[118] Kimura R, et al., The DYRK1A gene, encoded in chromosome 21 degenerative disease of the human central nervous system. Mech
Down syndrome critical region, bridges between beta-amyloid Ageing Dev 23(1): 73-94 (1983).
production and tau phosphorylation in Alzheimer disease. Hum [142] German, DC, Manaye KF, White CL 3rd, Woodward DJ, McIntire
Mol Genet 16(1): 15-23 (2007). DD, Smith WK, Kalaria RN, et al. Disease-specific patterns of
[119] Lloret A, et al., Amyloid-beta toxicity and tau locus coeruleus cell loss. Ann Neurol 32(5): 667-76 (1992).
hyperphosphorylation are linked via RCAN1 in Alzheimer's [143] Schliebs R, Arendt T. The cholinergic system in aging and
disease. J Alzheimers Dis 27(4): 701-9 (2011). neuronal degeneration. Behav Brain Res 221(2): 555-63 (2011).
[120] Head E, et al. Parallel Compensatory and Pathological events [144] Sadowski M, Wisniewski HM, Tarnawski M, Kozlowski PB, Lach
Associated with Tau Pathology in Middle Aged Individuals with B, Wegiel J. Entorhinal cortex of aged subjects with Down's
Down Syndrome. J Neuropath Exp Neurol 62(9): 917-926 (2003). syndrome shows severe neuronal loss caused by neurofibrillary
[121] Molina L, et al., Tau and apo E in CSF: potential aid for pathology. Acta Neuropathol (Berl) 97(2): 156-64 (1999) .
discriminating Alzheimer's disease from other dementias. [145] Mann DM, Yates PO, Marcyniuk B, Ravindra CR. Loss of
Neuroreport 10(17): 3491-5 (1999). neurones from cortical and subcortical areas in Down's syndrome
[122] Brooksbank BW, Martinez M, Balazs R. Altered composition of patients at middle age. Quantitative comparisons with younger
polyunsaturated fatty acyl groups in phosphoglycerides of Down's Down's patients and patients with Alzheimer's disease. J Neurol Sci
syndrome fetal brain. J Neurochem 44(3): 869-74 (1985). 80(1): 79-89 (1987).
[123] Odetti P, et al. Early glycoxidation damage in brains from Down's [146] Casanova MF, Walker LC, Whitehouse PJ, Price DL.
syndrome. Biochem Biophys Res Commun 243(3): 849-51 (1998). Abnormalities of the nucleus basalis in Down's syndrome. Ann
[124] Pratico D, et al. Down's syndrome is associated with increased Neurol 18(3): 310-3 (1985).
8,12-iso-iPF2alpha-VI levels: evidence for enhanced lipid [147] Gold BT, Johnson NF, Powell DK, Smith CD. White matter
peroxidation in vivo. Ann Neurol 48(5): 795-8 (2000). integrity and vulnerability to Alzheimer's disease: preliminary
[125] Di Domenico F, et al. Redox proteomics analysis of HNE-modified findings and future directions. Biochim Biophys Acta 1822(3):
proteins in Down syndrome brain: clues for understanding the 416-22 (2012).
development of Alzheimer disease. Free Radic Biol Med 71C: 270- [148] Tokuda T, Tanaka K, Kametani F, Ikeda S, Yanagisawa N.
280 (2014). Secretory form of beta-amyloid precursor protein is much
[126] Di Domenico F, et al. Impairment of proteostasis network in Down abundantly contained in the cerebral white matter in human brain.
syndrome prior to the development of Alzheimer's disease Neurosci Lett 175(1-2): 33-6 (1994).
neuropathology: redox proteomics analysis of human brain. [149] Mattiace LA, Kress Y, Davies P, Ksiezak-Reding H, Yen SH,
Biochim Biophys Acta 1832(8): 1249-59 (2013). Dickson DW. Ubiquitin-immunoreactive dystrophic neurites in
[127] Butterfield DA, et al. Redox proteomics analysis to decipher the Down's syndrome brains. J Neuropathol Exp Neurol 50(5): 547-59
neurobiology of Alzheimer-like neurodegeneration: overlaps in (1991).
Down's syndrome and Alzheimer's disease brain. Biochem J [150] Nishimura A, Sawada S, Ushiyama I, Yamamoto Y, Nakagawa T,
463(2): 177-89 (2014). Tanegashima A, Lectin-histochemical detection of degenerative
[128] Busciglio J, et al. Altered metabolism of the amyloid b precursor glycoconjugate deposits in human brain. Forensic Sci Int 113(1-3):
protein is associated with mitochondrial dysfunction in Down's 265-9 (2000).
syndrome. Neuron 33: 677-688 (2002). [151] Palminiello S, Jarzabek K, Kaur K, Walus M, Rabe A, Albertini G,
[129] Tiano L, Busciglio J. Mitochondrial dysfunction and Down's et al. Upregulation of phosphorylated alphaB-crystallin in the brain
syndrome: is there a role for coenzyme Q(10) ? Biofactors 37(5): of children and young adults with Down syndrome. Brain Res
386-92 (2011). 1268: 162-73 (2009).
AD Neuropathology in DS Current Alzheimer Research, 2016, Vol. 13, No. 1 29

[152] Dowjat WK, Adayev T, Kuchna I, Nowicki K, Palminiello S, [156] Pueschel SM, Craig WY, Haddow JE. Lipids and lipoproteins in
Hwang YW, et al. Trisomy-driven overexpression of DYRK1A persons with Down's syndrome. J Intellect Disabil Res 36(Pt 4):
kinase in the brain of subjects with Down syndrome. Neurosci Lett 365-369 (1992).
413(1): 77-81 (2007). [157] Head E, Lott IT, Patterson D, Doran E, Haier RJ. Possible
[153] Jansen J, Rozeboom W, Penning C, Evenhuis HM. Prevalence and compensatory events in adult Down syndrome brain prior to the
incidence of myocardial infarction and cerebrovascular accident in development of Alzheimer disease neuropathology: targets for
ageing persons with intellectual disability. J Intellect Disabil Res nonpharmacological intervention. J Alzheimers Dis 11(1): 61-76
57(7): 681-5 (2013). (2007).
[154] Draheim CC, Geijer JR, Dengel DR. Comparison of intima-media [158] Haier RJ, Alkire MT, White NS, Uncapher MR, Head E, Lott IT, et
thickness of the carotid artery and cardiovascular disease risk al. Temporal cortex hypermetabolism in Down syndrome prior to
factors in adults with versus without the Down syndrome. Am J the onset of dementia. Neurology 61(12): 1673-9 (2003).
Cardiol 106(10): 1512-6 (2010). [159] Martin SB, Dowling AL, Lianekhammy J, Lott IT, Doran E,
[155] Morrison RA, McGrath A, Davidson G, Brown JJ, Murray GD, Murphy MP, et al. Synaptophysin and Synaptojanin-1 in Down
Lever AF. Low blood pressure in Down's syndrome, A link with Syndrome are Differentially Affected by Alzheimer's Disease. J
Alzheimer's disease? Hypertension 28(4): 569-75 (1996). Alzheimers Dis 42(3): 767-75 (2014).

Received: March 12, 2015 Revised: May 18, 2015 Accepted: September 01, 2015

You might also like