You are on page 1of 11

PERSPECTIVES

proposed to modulate the course of AD‑DS,


OPINION
but further work is required to determine
their role and relative importance.
A genetic cause of Alzheimer The aim of this Opinion article is to pre-
sent an overview of clinical and pathologi-
disease: mechanistic insights cal features of AD‑DS and, by comparing
these to other forms of AD (particularly AD
from Down syndrome induced by Dup-APP), to highlight shared
genetic, pathogenic and protective mecha-
nisms and to discuss key future research
Frances K. Wiseman, Tamara Al-Janabi, John Hardy, Annette Karmiloff-Smith, areas. Similarities in the aetiologies of
Dean Nizetic, Victor L. J. Tybulewicz, Elizabeth M. C. Fisher and André Strydom AD‑DS and other forms of AD may high-
light common disease mechanisms, whereas
Abstract | Down syndrome, which arises in individuals carrying an extra copy of differences between these forms of AD may
chromosome 21, is associated with a greatly increased risk of early-onset Alzheimer help to identify novel genes and pathways
disease. It is thought that this risk is conferred by the presence of three copies of that are important in particular aspects of
the gene encoding amyloid precursor protein (APP) — an Alzheimer disease risk AD. Recent advances in genetic, cellular and
neuroimaging technologies have provided the
factor — although the possession of extra copies of other chromosome 21 genes
means to comprehensively explore the link
may also play a part. Further study of the mechanisms underlying the development between AD and DS, and recent improve-
of Alzheimer disease in people with Down syndrome could provide insights into ments in the life expectancy of people who
the mechanisms that cause dementia in the general population. have DS mean that more individuals than
ever before are developing AD‑DS. The grow-
Down syndrome (DS) is a complex, highly universally causes the development of ing interest in AD‑DS is long overdue, given
variable disorder that arises from trisomy amyloid plaques and neuro­fibrillary tangles the high AD burden in the DS population,
of chromosome 21. It was one of the first (NFTs), which are typical characteristics of and it is likely that research into AD‑DS may
chromosomal disorders to be identified1 and AD brain pathology, by the age of 40, and also lead to a better understanding of AD in
occurs with an incidence of approximately 1 approximately two-thirds of individuals the general population.
in 800 births2. Its prevalence within a given with DS develop dementia by the age of 60
population is influenced by infant mortality (REFS 6,7). However, rates of dementia do not Prevalence of AD‑DS
rates, access to health care, termination rates, reach 100%, even in older individuals, sug- A loss of cognitive function in middle-aged
average maternal age3 and life expectancy. gesting that some individuals with DS are adults with DS was described soon after
Indeed, despite the increased availability of protected from the onset of AD (FIG. 1). the identification of the syndrome18, and it
prenatal diagnosis and access to the option All of the features of DS arise because was later shown that this loss resulted from
of termination, the global prevalence of DS is of aberrant dosages of coding and/or non- the onset of AD dementia. As indicated
rising because of improvements in life expec- coding sequences present on chromosome 21. above, AD is now common in adults with
tancy: the number of adults with DS aged Among these sequences, the gene encoding DS who are over the age of 40 years and,
over 40 years has doubled in northern Europe amyloid precursor protein (APP) is thought like other genetic forms of EOAD, develops
since 1990 and, in the United Kingdom, one- to have a key role in the pathology of AD. two to three decades earlier in individu-
third of the estimated 40,000 people with DS The additional copy of APP may drive the als with DS than in the general population.
are thought to be over 40 years of age4. development of AD in individuals with DS Data describing the prevalence of AD‑DS
DS is the most common form of intellec- (AD‑DS) by increasing the levels of amyloid-β vary between studies because of diagnostic
tual disability. In addition to the features that (Aβ), a cleavage product of APP that misfolds issues, such as the presence of variable pre-
are found in everyone with the disorder, such and accumulates in the brain in people with morbid deficits, and survey methodology 19.
as the characteristic facial dysmorphology, AD. Consistent with this hypothesis, indi- However, the prevalence of AD in people
there are many DS‑associated phenotypes viduals with small internal chromosome 21 who have DS is <5% under the age of 40
that have variable penetrance and severity. duplications that result in three copies of APP (REF. 20) and then roughly doubles with each
For example, approximately 40% of indi- — a rare familial trait known as duplication 5‑year interval up to the age of 60. Hence,
viduals with DS have heart malformations of APP (Dup-APP) — also develop EOAD8–15. approximately 5–15% of individuals with DS
(usually atrioventricular septal defects)5. A Conversely, partial trisomy of chromo- aged 40–49 years and >30% of those aged
key feature of DS is a striking propensity to some 21 that does not result in the presence of 50–59 years experience significant cognitive
develop early-onset Alzheimer disease (EOAD). an extra APP does not lead to AD16,17. Several decline, indicating dementia (FIG. 1). Thus, as
Complete trisomy of chromosome 21 additional genes on chromosome 21 are with AD in the general population, age is a

564 | SEPTEMBER 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

AD-DS Dup-APP-associated AD Thus, people who have DS are at a greatly


100 100
increased risk of developing dementia, with
approximately 70% of individuals develop-
80 80
ing the condition by the age of 65. However,
Cumulative frequency

Cumulative frequency
unlike the situation for LOAD, gender and
60 60
cognitive reserve do not seem to influence
AD‑DS onset.
40 40

Clinical features of AD‑DS


20 20
The early symptoms of AD‑DS include fea-
tures that are typical of other forms of AD,
0 0
0 20 40 60 80 0 20 40 60 80 such as a decline in memory and language
Age (years) Age (years) skills that may be present several years before
Aβ-amyloid NFT PiB positive Dementia dementia is diagnosed35–37. However, changes
in personality and behaviour are more com-
Figure 1 | Development of pathology and dementia in AD‑DS and Nature Dup-APP.  The graphs
Reviews show the
| Neuroscience mon in the early stages of AD‑DS than they
cumulative frequency of amyloid plaque deposition (measured using histological methods and posi-
are in other forms of AD: individuals typi-
tron emission tomography with Pittsburgh compound B (PiB), a radioactive analogue of thioflavin that
binds to amyloid) and neurofibrillary tangle (NFT) development (measured using histological methods), cally display either apathy, lack of motivation
and the cumulative frequency of dementia in people with Alzheimer disease and Down syndrome and stubbornness, or increasing behavioural
(AD‑DS)6,33 and in individuals with familial AD induced by duplication of amyloid precursor protein excesses and impulsivity. These ‘non-cogni-
(Dup‑APP). As shown, people who have DS can live for many years with substantial amyloid deposition tive’ changes (also referred to as behavioural
before the development of dementia. Solid lines are based on the data described in Supplementary and psychological symptoms of dementia
information S1–S3 (tables). Dashed lines indicate hypothesized development of pathology for which (BPSDs))38–42 are associated with deficits in
there are currently no data available. Further pathological and clinical studies directly comparing executive functioning and with the frontal atro-
these two populations are required to verify the apparent differences in clinical dementia onset and phy that is visible on MRI scans, which may
to determine whether the development of pathology differs from that proposed here. Aβ, amyloid-β. indicate frontal lobe dysfunction40,43. These
changes may be related to pre-existing deficits
strong independent risk factor for AD-DS21. rates of other cardiovascular risk factors in the integrity of the frontal tracts that have
By the age of 65, 68–80% of individuals with — including hypertension, atherosclerosis been observed in individuals with DS44 and
DS have been shown to have developed and smoking 29,30 — that are thought to con- that may be worsened by Aβ deposition in
dementia6,7 (FIG. 1; Supplementary informa- tribute to the development of dementia in the frontal lobes45. Although BPSDs are very
tion S1 (table)), and some studies of institu- the general population31. Further studies are prominent in early AD‑DS, this presentation
tionalized people with DS suggest that rates required to understand how trisomy alters is not unique to these individuals — it also
are even higher 6,20,22. However, not all older the biology of the cardiovascular system and occurs, albeit at lower rates, during the early
individuals with DS develop dementia, with what impact this has on neurodegeneration stages of LOAD46 and EOAD47, particularly
some reaching their 70s without significant in people who have DS. in cases arising from mutations in the AD
symptoms of AD despite having full trisomy The brain reserve hypothesis is based risk gene presenilin 1 (PSEN1; which maps
of chromosome 21 (REF. 23). After the age of on the observation that, in the general to chromosome 14). Further studies are
60, prevalence rates decrease, probably owing population, individuals with higher levels required to determine the earliest changes
to the high mortality rate that is associated of education and/or more-active social associated with the development of dementia
with dementia21. and intellectual lifestyles have a lower risk in people who have DS, and to delineate other
The average age at which menopause of developing dementia32. The hypothesis clinical differences between AD‑DS, LOAD
begins in women with DS correlates with the predicts that individuals with more-severe and familial forms of EOAD, such as the
age of onset of dementia24–26; however, unlike premorbid cognitive impairment will have frequencies of co‑morbidities that may affect
the incidence of AD in euploid individu- an increased risk of developing demen- the onset and progression of dementia (for
als, gender does not affect the incidence of tia. However, no convincing relationship example, cardiovascular disease and systemic
AD-DS20,21. The reasons for this difference between severity of intellectual disability infections).
between the two populations are unknown, (or intelligence quotient (IQ) score) and Another feature of AD‑DS is the more-
although it is possible that trisomy may risk of AD has been found in people with frequent and earlier appearance of neuro-
cause changes in hormonal or cardiovascular DS33, possibly because of diagnostic dif- logical symptoms such as gait disturbance
biology that alter AD risk. The influence of ficulties in those with severe impairments. and seizures19 when compared with LOAD.
gender on dementia development is complex Survival time for AD‑DS does not differ Although heterogeneous, seizures associ-
in both the DS and euploid populations, much from that for late‑onset AD (LOAD), ated with AD‑DS often initially present with
and warrants more-extensive, longitudinal, with estimates varying between 3.5 years myoclonic jerks before progressing to tonic–
population-based study. (s.d 2.2)34 and 6.24 years (s.d. 4.1)6. However, clonic seizures and later to non-epileptic
Although increased levels of triglycerides individuals with severe intellectual disability myoclonus with cerebellar signs; electro-
and total body fat and low rates of exercise and dementia were found to have a longer encephalograms show diffuse slowing and
are reported in adults with DS27, and higher survival time after diagnosis than those with spike-and-wave patterns48–50. In individuals
cholesterol levels have been associated with milder intellectual disability 6, further sug- with LOAD, both complex partial and
the risk of developing dementia in this gesting that reduced brain reserve does not tonic–clonic seizures have been reported
group28, individuals with DS have lower accelerate disease progression in AD‑DS. to be the predominant type51,52. Although

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | SEPTEMBER 2015 | 565

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

seizures are reported to occur in 0.5–64% of well as symptoms of parkinsonism22. In sum- or synaptic loss, whereas dense-core plaques
people with LOAD51, more-recent popula- mary, BPSDs may be an important early fea- are often associated with dystrophic neurites
tion studies have suggested that seizure inci- ture of AD‑DS, and seizures are commonly and activated astroglia and microglia89. Also,
dence in LOAD is relatively low, occurring associated with AD‑DS. However, further Aβ42 — a form of Aβ that has a high tendency
in <5% of cases of the disease53. By contrast, comparative and mechanistic studies are to aggregate — accumulates before deposition
most people with AD‑DS eventually develop required to unravel the importance of these of Aβ40 in AD‑DS73,74,80, which is consistent
seizures, and a sudden onset of seizures in clinical observations. with the higher abundance of Aβ42 reported
older adults with DS is highly suggestive of in plaques in other forms of AD89. Cerebral
AD. Co‑morbid seizures are associated with Neuropathological changes in AD-DS amyloid angiopathy (CAA) — the deposition
a more-aggressive course of AD-DS54 and a The similarity between the neuropathological of Aβ within cerebral blood vessels — is also
greater dementia-associated mortality rate6. changes that occur in AD‑DS and those that observed in older individuals with DS74,80,87,90.
The mechanism underlying this striking characterize AD in other individuals was first However, unlike in LOAD, infarcts64 and vas-
clinical feature of AD‑DS is not understood, noted in 1929 (REF. 57) and was important for cular dementia seem to be rare in AD-DS91,
and the study of this may provide significant the widespread recognition of dementia in although cases of CAA-associated cerebral
insights into neurodegeneration, in particu- people who have DS. This discovery also had haemorrhage have been described92–95.
lar how changes in neuronal structure and a key role in the identification of Aβ as the In contrast to the findings of the histolog-
organization affect disease progression. major constituent of amyloid plaques58, the ical studies described above, in vivo amyloid
Similarly to other forms of AD, the identification of the first AD gene, APP59, and imaging by positron emission tomography
decline through middle-stage AD‑DS the subsequent development of the amyloid (PET) indicates that the earliest site of Aβ
dementia progressively affects more cogni- cascade hypothesis60. accumulation in AD‑DS, as in EOAD, could
tive domains and results in symptoms such as The overall distribution and biochemical be the striatum96 and that enhanced deposi-
dyspraxia, increasing incontinence and patho- composition of plaques (largely composed tion may occur in the frontal and parietal
logical grasping and sucking reflexes55,56, as of Aβ) and NFTs (largely composed of tau cortex 97. This discrepancy may be because
protein (encoded by microtubule-associated amyloid imaging recognizes only a subset of
protein tau (MAPT))) in people who have DS, Aβ aggregates, thus not all deposition may
Glossary EOAD and LOAD are similar 58,61–63. However, be detected98. Nonetheless, most individuals
a greater deposition of plaques and tangles with DS have amyloid-positive PET scans
Dyspraxia
Disrupted fine or gross motor coordination. occurs in the hippocampus in AD‑DS than in by the age of 50 (REFS 45,96,99,100). Amyloid
EOAD64 and, consistent with this, histological load, as measured by PET, does not correlate
Early-onset Alzheimer disease studies suggest that the earliest Aβ deposi- well with cognitive function in adults who
(EOAD). Occurrence of Alzheimer disease before the
tion in AD-DS occurs in the hippocampus65, have DS in cross-sectional studies45,99, high-
age of 65 years.
whereas in LOAD the earliest deposition lighting the importance of factors other than
Euploid occurs in the basal cortex 66. Furthermore, a amyloid in the development of dementia.
Having a normal chromosome number (46 chromosomes lower density of Aβ plaques has been reported However, longitudinal imaging studies in
in 23 pairs in humans). in the cortex in AD‑DS than in LOAD64,67,68. this population have yet to be undertaken
These differences may relate to amyloid and may be highly informative45,99.
Executive functioning
Mental processing skills involving the frontal cortex; used plaques in AD‑DS having a more amorphous No NFTs have been reported in AD‑DS in
for planning, attention focusing, working memory, mental morphology and a larger average size than the absence of dense-core plaque pathology,
flexibility and self-control. those present in LOAD69,70, resulting in a which is consistent with the predictions of
lower density caused by the presence of fewer the amyloid cascade hypothesis. The density
Incidence
The rate of new occurrences of a disorder within a
but larger plaques. In addition, the aggrega- of NFTs triples between the fourth and fifth
specified period of time. tion kinetics of Aβ may differ in people with decade of life in AD-DS77, mirroring the onset
DS because of a higher concentration of the of dementia, and NFT formation rather than
Lewy bodies peptide resulting from their additional copy amyloid deposition correlates best with cogni-
Protein aggregates typically containing α-synuclein.
of APP. Alternatively, differences in plaque tive decline34, which is consistent with similar
Myoclonic jerks load may result from the neurodevelopmental findings in LOAD. Thus, changes in tau may
Brief involuntary muscle twitches that are a medical sign differences that occur in people who have result in neuronal dysfunction in both AD‑DS
of various neurological disorders. DS, resulting in changes in synaptic activity, and LOAD. Interestingly, smaller relative
which is known to regulate Aβ production71. changes in nucleolar volume and a trend of
Parkinsonism
A clinical syndrome including bradykinesia (slow
In AD‑DS, intracellular accumulation of reduced cell loss have been reported in the
movements), muscle rigidity and tremor, often due to the Aβ precedes extracellular plaque accumu- cortex and locus coeruleus in AD‑DS com-
neurodegenerative condition Parkinson disease but also lation72–75 but becomes less prominent in pared with LOAD, despite comparable NFT
associated with other neurological conditions, toxins and older individuals with extensive pathology, loads, although similar cell loss was observed
medications.
as also observed in LOAD76. Additionally, in other brain areas68. This may reflect a dif-
Prevalence in AD‑DS, diffuse plaques composed of ferential response of the trisomic CNS to
The number of cases of a disorder at one time within non-fibrillary deposits of Aβ develop before accumulation of aggregated tau — suggest-
a population. those with dense cores that are composed ing, intriguingly, that chromosome 21 could
of amyloid (Supplementary information S2 encode a gene (or genes) that is neuroprotec-
Tonic–clonic seizures
A common type of epileptic seizure with a tonic phase
(table))34,64,65,73,74,77–88. Diffuse plaques are tive when triplicated. Further study is required
(stiffening of muscles and loss of consciousness) followed by typically not associated with other forms of to determine whether trisomy 21 may provide
a clonic phase (rapid, rhythmic jerking of arms and legs). neuropathology, such as activated glial cells protection from neurodegeneration.

566 | SEPTEMBER 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

As with people in the euploid popula- ubiquitylated and aggregated transactive genome-wide association studies have high-
tion, people who have DS may have exten- response DNA-binding protein 43 (TDP43; lighted the importance of the endosomal sys-
sive amyloid deposition but no clinical also known as TARDBP) in the cytoplasm tem in LOAD106, indicating that this system
signs of dementia (FIG. 1). Understanding and neurites is similar in AD‑DS (7–14% of may be of particular significance to disease.
how pathological changes due to AD relate cases) and familial AD (10–14% of cases),
to cognitive dysfunction is therefore a key whereas TDP43 neuropathology occurs more AD‑DS versus Dup-APP-associated AD
research challenge. Identifying the pro- frequently in LOAD (29–79% of cases), per- Dup-APP is a rare cause of familial EOAD,
cesses that cause an amyloid-laden brain to haps because of the later disease onset101,102. and comparison with AD‑DS yields pathoge-
convert from cognitively intact to impaired Lewy bodies, particularly in the amygdala, netic insights, as an additional copy of APP is
is crucial for understanding and success- occur at a similar frequency in AD-DS and present in both diseases. They therefore differ
fully treating AD. As people who have DS LOAD103, but dementia with Lewy bodies from other forms of familial AD that are the
develop amyloid deposition and NFTs by (DLB), which is characterized by cognitive result of mutations in APP, PSEN1 or PSEN2
the age of 40, study of this group of indi- decline with hallucinations and parkinson- that modulate the processing of APP and
viduals is likely to provide an important ism features, is rare in DS104. Granulovacuolar the generation of Aβ. In Dup-APP, regions
insight into the factors that cause dementia. degeneration, the formation of electron-dense of chromosome 21 triplication vary in size8–
Indeed, observations of AD‑DS neuropa- granules in double-membrane-bound cyto- 15,47,107,108
(FIG. 2); the smallest known duplica-
thology already underpin our mechanistic plasmic vacuoles, associated with plaque and tion contains only an additional copy of APP
understanding of AD, providing a detailed NFT pathology occurs at a similar frequency and no other coding genes8. By contrast, in
sequence of pathological changes and how in AD-DS and AD64. How this pathology AD‑DS, triplication of any chromosome 21
these may relate to changes in cognition. relates to the very early endosomal abnormal- gene in addition to APP may modulate the
Pathological features other than plaques ities that are reported to occur before birth development of dementia. Studying these
and NFTs also develop in both AD‑DS in individuals with DS105 is unclear and war- genes may therefore provide novel insights
and LOAD. Neuronal accumulation of rants further investigation. Recent AD‑related into AD mechanisms.

N6AMT1
POTED
LIPI LTN1
RBM11 MRPL39 RWDD2B, USP16,
HSPA13 CXADR JAM2 CCT8
SAMSN1 BTG3 ATP5J, GABPA MAP3K7CL
AF165138.7 C21orf91 NCAM2 APP BACH1
NRIP1 CHODL CYYR1 GRIK1
Country Sample USP25 TMPRSS15 ADAMTS1 CLDN17
Family 028 0.58 Mb
Family 037 0.78 Mb
France Family 009 1.98 Mb
Family 019 3.96 Mb
Family 229 6.37 Mb
The Family 1104 0.7 Mb
Netherlands Family 1043 0.7 Mb
Finland Proband 0.55 Mb
III-2, PED 3281 4.2 Mb
Japan
III-3, PED 2945 0.7 Mb
Sweden Proband 1.01 Mb
Proband 1 6.35 Mb
Proband 2 6.49 Mb
UK Proband 3 15.5 Mb
Proband 4 4.96 Mb
Proband 5 2.77 Mb
Family VI 3.4 Mb
USA 0.38 Mb
Family BRB
USA Proband 0.5 Mb
Spain Proband 14.5 Mb

Figure 2 | Regions of chromosome 21 duplicated in Dup-APP EOAD repeat‑binding protein‑alpha; GRIK1, glutamate receptor ionotropic,
and ICH.  Schematic illustrating the genetic regions affected in reported kainate 1; HSPA13, heat shock protein 70 kDa 13;
NatureICH, intracerebral
Reviews haem-
| Neuroscience
cases of early-onset Alzheimer disease (EOAD) accompanied by duplication orrhage; JAM2, junction adhesion molecule 2; LIPI, lipase member I; LTN1,
of amyloid precursor protein (Dup‑APP)8–15,108. The minimal duplicated listerin E3 ubiquitin protein ligase 1; MAP3K7CL, MAP3K7 carboxy‑terminal
region is shown in blue: the only gene duplicated in all cases is APP. like; MRPL39, mitochondrial ribosomal protein L39; N6AMT1, N-6 adenine-
ADAMTS1, a disintegrin and metalloproteinase with thrombospondin specific DNA methyltransferase 1; NCAM2, neural cell adhesion molecule 2;
motifs 1; ATP5J, ATP synthase‑coupling factor 6; BACH1, BTB and CNC NRIP1, nuclear receptor-interacting protein 1; PED, pedigree; POTED, POTE
homologue 1; BTG3, BTG family member 3; C21orf91, chromosome 21 open ankyrin domain family member D; RBM11, RNA‑binding motif protein 11;
reading frame 91; CCT8, chaperonin containing TCP1 8; CHODL, chondro­ RWDD2B, RWD domain-containing 2B; SAMSN1, SAM domain, SH3 domain
lectin; CLDN17, claudin 17; CXADR, coxsackie virus and adenovirus and nuclear localization signals 1; TMPRSS15, transmembrane protease
receptor homologue; CYYR1, cysteine- and tyrosine-rich 1; GABPA, GA serine 15; USP, ubiquitin‑specific peptidase.

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | SEPTEMBER 2015 | 567

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

The age of onset of dementia in individu- Box 1 | Identifying risk and protective factors for AD in young children
als with Dup-APP ranges from 39 to 64 years
(mean age ~52 years), and dementia shows It may seem counterintuitive to study infants and young children to understand a disease that
virtually complete penetrance by the age of presents only in adulthood. However, Alzheimer disease (AD) does not have an abrupt onset but
65 years. By contrast, there is a broad vari- emerges from a lengthy developmental trajectory in which precursors (for example, prodromal
ation in age of onset in AD-DS, and many changes) surface well before overt dementia symptoms. Several genes involved in
neurodevelopment have been suggested to have an important role in AD (including components
individuals present with significant cogni-
of the WNT and reelin signalling pathway201,202). Additionally, cultures of cells derived from infants
tive decline only after the age of 55 years,
with Down syndrome (DS) show clear overexpression of amyloid precursor protein (APP)119–122,
or even escape it altogether. This is remark- and amyloid‑β (Aβ) plaques have been found in the brains of children with DS who are as young as
able given the usual co‑morbid health 8 years of age65. Thus, the syndrome offers a longitudinal perspective on the multilevel effects of
issues and relative lack of brain reserve in Aβ and tau pathology during development.
individuals with DS. Thus, a possible pro- DS is diagnosed prenatally or at birth, and all infants with DS are at a significantly increased risk
tective mechanism (or mechanisms) from of subsequently developing AD, although not all will present with dementia, even as ageing adults.
triplication of an unknown gene (or genes) It is possible that in adults with DS, patterns of individual differences between those with AD and
on chromosome 21 may be important for those without AD are already rooted in their individual differences when they are just infants, at
resistance to dementia in people with DS. the genetic, cellular, neural, cognitive, behavioural, sleep and/or environmental levels. The
Moreover, intracerebral haemorrhage (ICH) challenge is to identify individual differences in childhood that pinpoint risk and protective factors
is common in individuals with Dup‑APP for subsequent AD outcome in adulthood. We can then identify biomarkers and devise early
intervention strategies, initially for individuals with DS and subsequently for members of the
(occurring in 20–50% of cases)9–14,47,108,
euploid population, revolutionizing our understanding of the pathways that lead to AD. Thus, a
whereas individuals with DS are generally
developmental approach is essential, especially as it has already been shown that differences that
protected from this pathology, with only can be observed in infancy in individuals with DS (for example, in the simple planning of saccadic
occasional reports. Thus, triplication of a eye movements) have cascading effects on cognitive outcomes in childhood and adulthood
chromosome 21 gene (or genes) may protect (for example, on numerical processing, language and face processing)203. Therefore, to fully
against some AD co‑morbidity, and further comprehend AD in adults, it is crucial to study its full developmental trajectory, and understanding
comparative study of AD‑DS and Dup‑APP DS makes this possible.
is required to understand the mechanisms
underpinning this observation.
The few histopathological Dup-APP must have a key role in AD. The pheno- gene is sufficient to cause EOAD in the
studies that have been carried out report type resulting from a dosage-sensitive gene absence of trisomy of the rest of chromo-
diffuse atrophy with associated neu- depends on the number of copies of the some 21 (REFS 8–15,47,107). The additional
ronal loss, deposition of plaques, CAA gene in the genome. However, not all genes copy of APP in DS does not typically cause
and accumulation of intraneuronal are dosage sensitive, as homeostasis often substantial Aβ accumulation until the
Aβ40 and NFTs11,109, and this pathology prevents a gene from being overexpressed second or third decade of life, although
seems to be similar to AD‑DS pathology and the regulation of expression is often amyloid pathology has been demonstrated
(Supplementary information S3 (table)). dependent on environmental context 112. in a few childhood post-mortem exami-
However, further studies are needed75,109. Furthermore, trisomy 21 causes widespread nations (BOX 1; FIG. 1). This lack of early
Clinical DLB and cortical Lewy bodies have transcriptional dysregulation112,113, which Aβ accumulation may be due to APP not
been observed in a few cases11,13,109, but cur- may be the result of aneuploidy rather than becoming dosage sensitive until adult-
rently there are insufficient data on these of triplication of a specific gene. The impor- hood, as suggested by both mouse and
phenotypes to compare Dup-APP with tance of this in AD‑DS remains unclear. human studies115–117. However, increased
AD‑DS or LOAD. As in AD‑DS, there is a Finally, acceleration of the epigenetic levels of soluble Aβ42 are found in ~50% of
greatly increased risk of dementia-associ- changes associated with ageing occurs in trisomy 21 fetal brains118, suggesting that
ated seizures in Dup-APP10–13,47, in contrast the DS brain114 — whether this alters gene APP may be dosage sensitive during fetal
to LOAD, in which seizures are relatively expression or modulates the development development of individuals with DS but that
rare. This suggests that duplication of of AD is an important area for future study. this change may not be sufficient to cause
APP, and possibly of other genes located The development of neuropathology extensive Aβ deposition in the developing
nearby, could be epileptogenic; however, and dementia varies significantly between brain — perhaps because of efficient clear-
as late‑onset seizures often follow onset of individuals with DS, and understanding ance. Consistent with this, overexpression
dementia, they may also be related to syn- the factors (genetic or environmental) that of APP and/or increased levels of Aβ have
aptic deterioration that results in abnormal cause this variation is likely to provide key been reported in trisomy 21 human cell
synchronization of neuronal networks and insights into disease mechanisms. Below, we models, including in induced pluripotent
hyperexcitability 110. describe the genes that are currently impli- stem cells (iPSCs) derived from infants or
cated in the development of AD‑DS and young adults with DS119–122. Although trip-
Genes and mechanisms in AD‑DS highlight the importance of further study of lication of APP does not necessarily lead
The presence of three copies of a dosage- the genetics of AD‑DS to understand how to enhanced expression of APP and subse-
sensitive gene (or genes) on chromosome 21 variation in the whole genome influences quent increase in Aβ accumulation in all
results in greatly enhanced risk of AD. the development of disease. contexts, overexpression of APP is strongly
Chromosome 21 carries 233 coding genes, linked to Aβ deposition in adult life. Thus,
299 long non-coding genes (Ensembl Triplication of APP. The key dosage- elucidating the factors that control the reg-
release 78) and 29 microRNAs (miRBase sensitive gene for AD‑DS is likely to be ulation of APP expression will considerably
release 21)111; thus, one or more of these APP, as an additional normal copy of this aid our understanding of AD.

568 | SEPTEMBER 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Box 2 | Modelling AD‑DS in mice and in human iPSCs


are central to Aβ generation. Moreover, an
extra copy of APP is sufficient to cause endo-
Amyloid precursor protein (APP) overexpression in mouse models causes dysfunction of basal somal enlargement and intracellular traf-
forebrain cholinergic neurons and synaptic and behavioural changes141,204–206. However, increased ficking defects141,142 via an Aβ‑independent
expression of wild‑type APP, even at levels in excess of those present in Down syndrome (DS), is mechanism143. Enlargement of endosomes
insufficient to cause extensive Alzheimer disease (AD) neuropathology207. Only mice expressing
in trisomic neurons may cause axonal traf-
mutant APP and/or other AD‑associated genes recapitulate aspects of AD neuropathology and/ or
cognitive change207. Similarly, although altered expression of many chromosome 21 genes modifies
ficking defects that contribute to neuronal
mouse models of familial AD, whether a single extra copy of these genes is sufficient to affect degeneration141.
pathology and behaviour remains unclear. However, chromosome engineering, which enables the Triplication of chromosome 21 genes
generation of mouse models with large genomic duplications, may help to elucidate the effects of other than APP may also affect the secretory–
trisomy on neurodegeneration208. endosome system, thereby affecting synaptic
Reprogramming human somatic cells into induced pluripotent stem cells (iPSCs; which are in an function, Aβ production and Aβ clearance.
embryonic stem cell-like state) is revolutionizing AD modelling, and advances in three-dimensional Small segmental duplications of the chromo-
differentiation now permit the development of extensive amyloid‑β (Aβ) and tau pathology in vitro. some 21 endosome-to-Golgi‑trafficking gene
Comparisons have been made between euploid and trisomy 21 iPSCs derived from multiple DOPEY2 (REF. 144) have been associated with
sources, including different individuals (non-isogenic)122,209; isogenic lines generated in cell culture,
LOAD and mild cognitive impairment14,145,
spontaneously or by selection154,210; lines in which one of the three copies of chromosome 21 has
been silenced211; monozygotic twins that were discordant for trisomy 21 (REF. 169); and non-inte‑
although this was not replicated in an inde-
gration-reprogrammed isogenic lines from an adult with mosaic DS (a condition in which only a pendent study 146. A reduction in the dose of
percentage of an individual’s cells carry an extra copy of chromosome 21)121. Neurons derived from the chromosome 21 gene cystatin B (CSTB),
iPSCs show cellular phenotypes underpinning AD pathology, such as increased Aβ production, which encodes an endogenous inhibitor of
abnormal subcellular distribution of phosphorylated tau, mitochondrial abnormalities and lysosomal cathepsins, decreases the accu-
accelerated cellular ageing121,122,154,212. DS iPSC models can be used to dissect the effect of trisomy mulation of Aβ and associated cognitive
of individual chromosome 21 genes (for example, by genome editing using clustered regularly deficits147. Overexpression of another chro-
interspaced short palindromic repeat–CRISPR‑associated protein 9 (CRISPR–Cas9) technology), mosome 21 gene, synaptojanin 1 (SYNJ1),
to develop high-throughput screening assays for phenotype-correcting compounds and to which encodes a phosphoinositide phos-
investigate cellular phenotypes in iPSCs generated from individuals with DS with very early
phatase that regulates levels of membrane
versus very late ages of onset of dementia.
phosphatidylinositol‑4,5‑bisphosphate, has
been associated with endosomal enlarge-
Interaction of other chromosome 21 genes Whether triplication of any chromosome 21 ment148, whereas reduced expression of
with APP. Several proteins encoded by gene alters APP biology sufficiently to modu- SYNJ1 lowers Aβ accumulation, as well
other chromosome 21 genes have been sug- late the development of AD remains to be as neuronal dysfunction and cognitive
gested to modulate APP processing and Aβ determined. deficits149,150. How endosomal enlargement
generation (BOX 2; FIG. 3). For example, the caused by trisomy contributes to neuronal
transcription factor ETS2 is thought to trans- Genes involved in LOAD. Polymorphisms dysfunction and degeneration is another
activate the APP promoter, leading to over- in genes with important functions in LOAD important area for future research.
expression123. The chromosome 21‑encoded have similar roles in the development of
proteins small ubiquitin-related modifier 3 AD‑DS; for example, the apolipoprotein E Mitochondria and ROS. Mitochondrial
(SUMO3) and dual-specificity tyrosine- (APOE) ε4 allele is associated with greater dysfunction and enhanced production of
phosphorylation-regulated kinase 1A Aβ deposition, as well as with earlier onset reactive oxygen species (ROS) occur in peo-
(DYRK1A) modify APP post-translationally, and increased risk of AD‑DS, whereas the ple with DS and in trisomy 21 models151–154,
which may alter Aβ generation124–126. APOE ε2 allele leads to reduced Aβ deposi- and may contribute to the accelerated age-
Additionally, the chromosome 21 microRNA tion and a lower risk of disease132–138. Similarly, ing reported in people who have DS155.
miR‑155 has been suggested to modulate variants of phosphatidylinositol-binding Mitochondrial impairment may directly
γ‑secretase activity and hence the processing clathrin assembly protein (PICALM) and affect energy-hungry synapses, contributing
of APP, through its effect on the expression sortilin-related receptor 1 (SORL1) influ- to cognitive deficits156. Moreover, increased
of sorting nexin 27 (REF. 127). Moreover, the ence age of onset in AD‑DS, as they do in levels of ROS make trisomic neurons more
β‑secretase responsible for processing APP, LOAD132,139,140, further supporting the theory prone to undergoing apoptosis, potentially
β‑site APP-cleaving enzyme 1 (BACE1), has that common mechanisms underlie both dis- making them more likely to degenerate151.
a homologue, BACE2, encoded on chromo- eases. Whether variation in other genes with Trisomy 21‑associated increases in ROS
some 21, which may influence the onset of a role in LOAD is also important for AD‑DS levels may alter APP processing, promot-
dementia in people with DS128. BACE2 does remains to be determined and is an important ing intracellular accumulation of Aβ119,151.
not have β‑secretase activity, and in fact area for future study. Large-scale study of the Thus, protecting the trisomic brain from
cleaves APP on the carboxy‑terminal side of genetic variants that contribute to the onset of ROS may be of therapeutic value, although
the β‑secretase cut site within the Aβ region, dementia in AD‑DS will provide an opportu- antioxidant supplementation has failed to
preventing generation of the peptide. Thus, nity to gain insights into the mechanisms that show efficacy in preventing dementia in
enhancing BACE2 expression may be protec- underpin variation in the onset of dementia. this population157. Interestingly, superoxide
tive against accumulation of Aβ129. However, dismutase 1 (SOD1), which has a key role
BACE2 overexpression does not alter Aβ Disruption of secretory and endosomal sys- in processing ROS, lies on chromosome 21,
accumulation in a mouse model130, and the tems. The earliest site of Aβ accumulation and upregulation of SOD1 seems to protect
protein does not seem to have enhanced in AD‑DS is within the neuron72–75, indicat- against APP and Aβ neurotoxicity 158, per-
expression in the adult DS brain115,131. ing that secretory and endosomal systems haps by modulating Aβ oligomerization159.

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | SEPTEMBER 2015 | 569

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Consistent with this, higher SOD1 enzymatic APOE


SOD1 ABCG1
activity correlates with better memory in
adults with DS160. However, increased SOD1 Oxidative Cholesterol
activity has also been suggested to cause DYRK1A stress metabolism
accelerated cell senescence by increasing the SUMO3
levels of hydrogen peroxide, a form of ROS161. ETS2
CSTB
? APOE
SORL1 ? APOE ?
Neuronal development and function. Several
BACE2 Processing Clearance PICALM ?
processes are likely to contribute to the PICALM ? CSTB
intellectual disability associated with DS. DOPEY2 SYNJ1 mir-155
ITSN1
These include a reduction in the numbers

of neurons and dendritic spines, dendritic APP
arborization, an alteration in the excita-
tory–inhibitory balance and a global impair-
ment in network connectivity 68,162–166. These Aggregation
perturbations in the structure, function and Aβ plaques
organization of the CNS may profoundly Deposition
affect its degeneration in AD‑DS (BOX 1). Oligomeric Aβ
Triplication of several chromosome 21 genes
contributes to changes in neurodevelopment
and/or neuronal function. For example, Tau
ubiquitin‑specific peptidase 16 (USP16) Microglial
or DYRK1A upregulation alters stem cell cell
fate167–169, which may in turn alter neuronal S100B • Splicing RCAN1
• Aggregation DYRK1A
differentiation. Additionally, overexpres- mir-155 APOE ? APOE ?
• Phosphorylation
sion of several chromosome 21 genes (for Inflammation P P
P P
example, the microRNA gene mir‑155 and Activated
microglial P P P
the protein-coding genes SYNJ1, regulator of P P P NFT
cell PP P P
calcineurin 1 (RCAN1), intersectin 1 (ITSN1) P
P P
and DS cell adhesion molecule (DSCAM)) P P
P
DOPEY2 mir-155
has been implicated in deficits in synaptic
structure and function148,170,171. These genes ITSN1 SYNJ1
may also play a part in AD‑DS, perhaps via RCAN1
Vascular
an effect on APP processing or on cognitive Synaptic function APOE ?
dysfunction
reserve. APP overexpression may also affect DYRK1A
CNS function independently of the produc- Oxidative Nervous system
SOD1 DSCAM
tion and accumulation of Aβ, because the stress development
expression level of full-length APP influ- USP16
ences neurogenesis, neuronal migration, Neurodegeneration
axonal growth and the maintenance of the
excitatory–inhibitory balance172,173. How the
Dementia
changes in CNS function caused by trisomy
of chromosome 21 affect neurodegeneration Figure 3 | Schematic of suggested mechanisms that are important Naturein AD‑DS| Neuroscience
Reviews and their related
in AD‑DS is little understood and is a crucial genes.  Several genes may modulate processes that are relevant to the development of Alzheimer
area of future research. disease in people with Down syndrome (AD‑DS); these include non-chromosome 21 genes, such as
apolipoprotein E (APOE; which could alter disease by influencing cholesterol metabolism and possibly
many other pathways), phosphatidylinositol‑binding clathrin assembly protein (PICALM), sortilin-
Intracellular signalling and tau. Perturbations related receptor 1 (SORL1; which may influence disease via the endocytosis system and amyloid
in intracellular signalling associated with tri- precursor protein (APP) processing) and microtubule-associated protein tau (MAPT). Tau aggregates
somy 21 (REF. 174) may affect the response of to form neurofibrillary tangles (NFTs). Numerous chromosome 21 genes have also been suggested to
the CNS to pathological changes. For exam- influence the development of AD‑DS, including genes that may influence APP processing and synaptic
ple, overexpression of the chromosome 21 function via their role in the secretory–endosome system (including cystatin B (CSTB), DOPEY2, syn-
genes RCAN1 and DYRK1A promotes aber- aptojanin 1 (SYNJ1), intersectin 1 (ITSN1) and the microRNA gene mir‑155), APP processing (including
rant phosphorylation of tau152,175–177. DYRK1A small ubiquitin-like modifier 3 (SUMO3), ETS2 and beta-site APP-cleaving enzyme 2 (BACE2)),
is dosage sensitive in the adult brain178, and cholesterol metabolism (including ATP-binding cassette G1 (ABCG1)), cellular signalling and tau
overexpression of this gene modulates tau phosphorylation (including dual-specificity tyrosine-phosphorylation-regulated kinase 1A (DYRK1A)
and regulator of calcineurin 1 (RCAN1)), inflammation (including mir‑155 and S100 calcium‑binding
splicing, altering the relative abundance of
protein beta (S100B)), synaptic function (including DOPEY2, SYNJ1, ITSN1, RCAN1 and mir‑155),
tau with three or four microtubule-binding neurodevelopment (including ubiquitin‑specific peptidase 16 (USP16), DYRK1A and DS cell adhesion
domains (3‑repeat (3R) and 4R tau, respec- molecule (DSCAM)) and oxidative stress (superoxide dismutase 1 (SOD1)). The relative importance of
tively), which may affect the formation of these processes to the development of dementia in AD‑DS remains unclear and constitutes an area
NFTs179,180. Consistent with this, an increase for future study. Chromosome 21 genes and gene products are shown in purple; non-chromosome 21
in the ratio of 3R/4R tau has been reported genes and gene products are shown in green. Aβ, amyloid-β.

570 | SEPTEMBER 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

to occur in AD‑DS, as compared with LOAD to AD‑DS has yet to be fully explored. The remarkable set of tools for studying AD‑DS,
or age-matched euploid individuals without chromosome 21 gene S100 calcium‑binding ranging from new model systems to genomics
dementia179,180. Additionally, an increase in protein beta (S100B) is expressed in astro- studies. Although there are undoubtedly spe-
the total amount of tau has been reported cytes and is upregulated in both AD196 and cific problems in both analysing and treating
in the cortex in AD-DS as compared with AD-DS192, and it may contribute to neuro- people who have DS for AD, such as issues of
that in age-matched euploid individuals degeneration by promoting Aβ deposition197 informed consent, trisomy 21 is an extremely
without dementia, and in DS iPSC‑derived and tau phosphorylation198 and by creating a important disorder for learning about the
neurons122,179; this upregulation may be the neurotoxic environment through the release development of neurodegeneration and for
result of increased APP levels181. DYRK1A of extracellular signals199. testing potential therapeutic strategies to the
also downregulates the levels of neural restric- benefit of everyone at risk of AD.
tive silencing factor (NRSF; also known as Translational research
REST), a neuroprotective protein168,169, which The life expectancy of people with DS is Frances K. Wiseman is a member of the LonDownS
has reduced expression in people with AD182. increasing because of better health care and Consortium and is at the Department of
Neurodegenerative Disease, Institute of Neurology,
Variants in DYRK1A have been associated improved social inclusion. However, as with
University College London, Queen Square, London
with risk of LOAD183, further indicating the euploid population, ageing brings new WC1N 3BG, UK.
a possible role in disease pathogenesis, issues; in people with DS, a major ageing- Tamara Al-Janabi is a member of the LonDownS
although this association was not replicated related issue is a vastly increased risk of Consortium and is at the Division of Psychiatry,
in an independent study 184. EOAD. People who have DS develop amyloid University College London, Maple House,
plaques and NFTs by the age of 40 years, 149 Tottenham Court Road, London W1T 7NF, UK.
Cholesterol metabolism. Alterations in cho- and many individuals subsequently go on John Hardy is a member of the LonDownS Consortium
lesterol metabolism may contribute to the to develop dementia. Despite genetic and and is at the Department of Molecular Neuroscience,
Institute of Neurology, University College London,
development of dementia31. Total cholesterol Aβ differences between the various forms Queen Square, London WC1N 3BG, UK.
levels have been suggested to predict the of EOAD and LOAD, many similarities in
Annette Karmiloff-Smith is a member of the LonDownS
onset of dementia in people with DS, par- disease process are observed such that AD Consortium and is at the Centre for Brain and Cognitive
ticularly in those individuals who have an seems to converge on common mechanisms Development, Birkbeck, University of London, Malet
APOE ε4 allele28. Clinical trials are therefore of pathology. Thus, in the AD‑DS patient Street, London WC1E 7HX, UK.
underway to determine whether statins can population, it is feasible both to determine Dean Nizetic is a member of the LonDownS Consortium
prevent decline in older adults with DS, which the factors (genetic and/or environmental) and is at the Lee Kong Chian School of
Medicine, Nanyang Technological University, Novena
may provide both clinical and mechanistic that cause conversion from pathological
Campus, 11 Mandalay Road, Singapore 308232;
insights185. The chromosome 21 lipid trans- disease to cognitive decline and to undertake and the Blizard Institute, Barts and the London School
porter ATP-binding cassette G1 (ABCG1) intervention trials to halt the development of of Medicine, Queen Mary University of London,
has been suggested to regulate cholesterol dementia. 4 Newark Street, London E1 2AT, UK.
efflux and may alter cholesterol metabolism As APP gene dosage is the major determi- Victor L. J. Tybulewicz is a member of the LonDownS
in people with DS186. Whether trisomy of this nant of AD‑DS, it follows that therapies aimed Consortium and is at the Francis Crick Institute,
Mill Hill Laboratory, London NW7 1AA, UK.
gene is related to the development of AD‑DS at reducing Aβ (such as BACE inhibition or
Elizabeth M. C. Fisher is a member of the LonDownS
remains unclear, as ABCG1 overexpression Aβ immunization) might have a beneficial
Consortium and is at the Department of
has been reported both to increase and to effect in the DS population. Such approaches Neurodegenerative Disease, Institute of Neurology,
decrease Aβ generation in vitro187,188 and are being trialled for people with familial AD University College London, Queen Square,
does not change Aβ accumulation in vivo189, arising from APP or PSEN1 mutations200, London WC1N 3BG, UK.
suggesting that this gene may not be asso- and similar clinical trials in AD‑DS could André Strydom is a member of the LonDownS
ciated with the development of AD‑DS. provide valuable additional insight, given the Consortium and is at the Division of Psychiatry,
University College London, Maple House,
Further study is required to understand the predictable conversion to AD neuropathology
149 Tottenham Court Road, London W1T 7NF, UK.
mechanisms that underlie the link between and subsequent dementia in this population.
Correspondence to: E.M.C.F.
increased cholesterol levels and the onset of Other treatment options that require further e-mail: e.fisher@prion.ucl.ac.uk
dementia in individuals with DS. development include DYRK1A inhibitors and
doi:10.1038/nrn3983
ROS modulators. Notably, treatment safety Published online 5 August 2015
Immune system dysfunction. Growing is of particular importance because many
evidence shows that the immune system individuals with DS are unable to consent to 1. Lejeune, J., Gautier, M. & Turpin, R. Etude des
plays an important part in the development their own participation in clinical trials and chromosomes somatiques de neuf enfants mongoliens.
C. R. Hebd. Seances Acad. Sci. 248, 1721–1722 (in
of AD106,190. Individuals with DS are at an because they will probably need to undergo French) (1959) .
increased risk of immune system dysfunction: treatment for many years. 2. de Graaf, G., Buckley, F. & Skotko, B. G. Estimates of
the live births, natural losses, and elective
these individuals have a higher incidence of terminations with Down syndrome in the United
both autoimmune and infectious disease191, Conclusions States. Am. J. Med. Genet. A 167A, 756–767
(2015).
and show upregulation of pro-inflammatory Many questions remain to be answered in 3. Wu, J. H. & Morris, J. K. Trends in maternal age
makers, including interleukin‑1, in the AD‑DS, including, most importantly, the distribution and the live birth prevalence of Down’s
syndrome in England and Wales: 1938–2010. Eur.
brain192,193. This dysregulation may contrib- mechanisms underlying the later onset of J. Hum. Genet. 21, 943–947 (2013).
ute to AD‑DS through alterations in micro- dementia as compared with Dup‑APP, how 4. Wu, J. H. & Morris, J. K. The population prevalence of
Down’s syndrome in England and Wales in 2011. Eur.
glial activation190. Microglia in AD‑DS have neurodevelopmental perturbations affect J. Hum. Genet. 21, 1016–1019 (2013).
been reported to be associated with both neurodegeneration and the identity of any 5. Wiseman, F. K., Alford, K. A., Tybulewicz, V. L. J. &
Fisher, E. M. C. Down syndrome — recent progress
mature Aβ plaques194 and NFTs195, although chromosome 21 gene (or genes) that may and future prospects. Hum. Mol. Genet. 18, R75–R83
the contribution of the immune response protect against dementia. We now have a (2009).

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | SEPTEMBER 2015 | 571

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

6. McCarron, M., McCallion, P., Reilly, E. & Mulryan, N. 30. van de Louw, J., Vorstenbosch, R., Vinck, L., 54. Lott, I. T. et al. Down syndrome and dementia:
A prospective 14‑year longitudinal follow‑up of Penning, C. & Evenhuis, H. Prevalence of hypertension seizures and cognitive decline. J. Alzheimers Dis. 29,
dementia in persons with Down syndrome. J. Intellect. in adults with intellectual disability in the Netherlands. 177–185 (2012).
Disabil. Res. 58, 61–70 (2014). J. Intellect. Disabil. Res. 53, 78–84 (2009). 55. Crayton, L., Oliver, C., Holland, A., Bradbury, J. &
7. Zigman, W. B., Schupf, N., Urv, T., Zigman, A. & 31. Stampfer, M. J. Cardiovascular disease and Hall, S. The neuropsychological assessment of age
Silverman, W. Incidence and temporal patterns of adaptive Alzheimer’s disease: common links. J. Internal Med. related cognitive deficits in adults with Down’s
behavior change in adults with mental retardation. Am. 260, 211–223 (2006). syndrome. J. Appl. Res. Intellect. Disabil. 11,
J. Mental Retard. 107, 161–174 (2002). 32. Stern, Y. Cognitive reserve in ageing and Alzheimer’s 255–272 (1998).
8. Hooli, B. V. et al. Role of common and rare APP DNA disease. Lancet Neurol. 11, 1006–1012 (2012). 56. Dalton, A. J., Mehta, P. D., Fedor, B. L. & Patti, P. J.
sequence variants in Alzheimer disease. Neurology 33. Zigman, W. B. et al. Alzheimer’s disease in adults with Cognitive changes in memory precede those in praxis
78, 1250–1257 (2012). Down syndrome. Int. Rev. Res. Ment. Retard. 36, in aging persons with Down syndrome. J. Intellect.
9. Kasuga, K. et al. Identification of independent APP 103–145 (2008). Dev. Disabil. 24, 169–187 (1999).
locus duplication in Japanese patients with early-onset 34. Margallo-Lana, M. L. et al. Fifteen-year follow‑up of 92 57. Struwe, F. Histopathologische Untersuchungen über
Alzheimer disease. J. Neurol. Neurosurg. Psychiatry hospitalized adults with Down’s syndrome: incidence Entstehung und Wesen der senile Plaques. Z. Gesamte
80, 1050–1052 (2009). of cognitive decline, its relationship to age and Neurol. Psy. 122, 291–307 (in German) (1929) .
10. McNaughton, D. et al. Duplication of amyloid neuropathology. J. Intellect. Disabil. Res. 51, 58. Glenner, G. G. & Wong, C. W. Alzheimer’s disease and
precursor protein (APP), but not prion protein (PRNP) 463–477 (2007). Downs syndrome — sharing of a unique
gene is a significant cause of early onset dementia in a 35. Devenny, D. A., Krinsky-McHale, S. J., Sersen, G. & cerebrovascular amyloid fibril protein. Biochem.
large UK series. Neurobiol. Aging 33, 426.e13–426. Silverman, W. P. Sequence of cognitive decline in Biophys. Res. Commun. 122, 1131–1135 (1984).
e21 (2010). dementia in adults with Down’s syndrome. J. Intellect. 59. Goate, A. et al. Segregation of a missense mutation
11. Rovelet-Lecrux, A. et al. APP locus duplication causes Disabil. Res. 44, 654–665 (2000). in the amyloid precursor protein gene with familial
autosomal dominant early-onset Alzheimer disease 36. Devenny, D. A., Zimmerli, E. J., Kittler, P. & Krinsky- Alzheimer’s disease. Nature 349, 704–706 (1991).
with cerebral amyloid angiopathy. Nat. Genet. 38, McHale, S. J. Cued recall in early-stage dementia in 60. Hardy, J. A. & Higgins, G. A. Alzheimer’s disease — the
24–26 (2006). adults with Down’s syndrome. J. Intellect. Disabil. Res. amyloid cascade hypothesis. Science 256, 184–185
12. Rovelet-Lecrux, A. et al. APP locus duplication in a 46, 472–483 (2002). (1992).
Finnish family with dementia and intracerebral 37. Krinsky-McHale, S. J., Devenny, D. A. & 61. Goedert, M., Spillantini, M. G., Cairns, N. J. &
haemorrhage. J. Neurol. Neurosurg. Psychiatry 78, Silverman, W. P. Changes in explicit memory Crowther, R. A. Tau proteins of Alzheimer paired
1158–1159 (2007). associated with early dementia in adults with Down’s helical filaments — abnormal phosphorylation of all 6
13. Sleegers, K. et al. APP duplication is sufficient to syndrome. J. Intellect. Disabil. Res. 46, 198–208 brain isoforms. Neuron 8, 159–168 (1992).
cause early onset Alzheimer’s dementia with (2002). 62. Mann, D. M. A. Alzheimer’s disease and Down’s
cerebral amyloid angiopathy. Brain 129, 38. Adams, D. & Oliver, C. The relationship between syndrome. Histopathology 13, 125–137 (1988).
2977–2983 (2006). acquired impairments of executive function and 63. Wisniewski, H. M. & Rabe, A. Discrepancy between
14. Swaminathan, S. et al. Analysis of copy number behaviour change in adults with Down syndrome. Alzheimer-type neuropathology and dementia in
variation in Alzheimer’s disease in a cohort of J. Intellect. Disabil. Res. 54, 393–405 (2010). persons with Down’s syndrome. Ann. NY Acad. Sci.
clinically characterized and neuropathologically 39. Ball, S. L. et al. Personality and behaviour changes 477, 247–260 (1986).
verified individuals. PLoS ONE 7 e50640 (2012). mark the early stages of Alzheimer’s disease in adults 64. Mann, D. M. A. The pathological association between
15. Thonberg, H. et al. Mutation screening of patients with Down’s syndrome: findings from a prospective Down syndrome and Alzheimer’s disease. Mech.
with Alzheimer disease identifies APP locus population-based study. Int. J. Geriatr. Psychiatry 21, Ageing Dev. 43, 99–136 (1988).
duplication in a Swedish patient. BMC Res. Notes 4, 661–673 (2006). 65. Leverenz, J. B. & Raskind, M. A. Early amyloid
476 (2011). 40. Ball, S. L., Holland, A. J., Treppner, P., Watson, P. C. & deposition in the medial temporal lobe of young Down
16. Prasher, V. P. et al. Molecular mapping of Alzheimer- Huppert, F. A. Executive dysfunction and its syndrome patients: a regional quantitative analysis.
type dementia in Down’s syndrome. Ann. Neurol. 43, association with personality and behaviour changes in Exp. Neurol. 150, 296–304 (1998).
380–383 (1998). the development of Alzheimer’s disease in adults with 66. Braak, H. & Braak, E. Neuropathological staging of
17. Korbel, J. O. et al. The genetic architecture of Down Down syndrome and mild to moderate learning Alzheimer-related changes. Acta Neuropathol. 82,
syndrome phenotypes revealed by high-resolution disabilities. Br. J. Clin. Psychol. 47, 1–29 (2008). 239–259 (1991).
analysis of human segmental trisomies. Proc. Natl 41. Holland, A. J., Hon, J., Huppert, F. A. & Stevens, F. 67. Egensperger, R. et al. Reverse relationship between
Acad. Sci. USA 106, 12031–12036 (2009). Incidence and course of dementia in people with β-amyloid precursor protein and beta-amyloid peptide
18. Fraser, J. & Mitchell, A. Kalmuc idiocy: report of a case Down’s syndrome: findings from a population-based plaques in Down’s syndrome versus sporadic/familial
with autopsy with notes on 62 cases. J. Mental Sci. study. J. Intellect. Disabil. Res. 44, 138–146 (2000). Alzheimer’s disease. Acta Neuropathol. 97, 113–118
22, 161–169 (1876). 42. Oliver, C., Kalsy, S., McQuillan, S. & Hall, S. (1999).
19. Strydom, A. et al. Dementia in older adults with Behavioural excesses and deficits associated with 68. Mann, D. M. A., Yates, P. O., Marcyniuk, B. &
intellectual disabilities — epidemiology, dementia in adults who have Down syndrome. J. Appl. Ravindra, C. R. Loss of neurons from cortical and
presentation, and diagnosis. J. Intellect. Disabil. 7, Res. Intellect. Disabil. 24, 208–216 (2011). subcortical areas in Down’s syndrome patients at
96–110 (2010). 43. Nelson, L. D., Orme, D., Osann, K. & Lott, I. T. middle-age — quantitative comparisons with younger
20. Tyrrell, J. et al. Dementia in people with Down’s Neurological changes and emotional functioning in Down’s patients and patients with Alzheimer’s disease.
syndrome. Int. J. Geriatr. Psychiatry 16, 1168–1174 adults with Down Syndrome. J. Intellect. Disabil. Res. J. Neurol. Sci. 80, 79–89 (1987).
(2001). 45, 450–456 (2001). 69. Allsop, D., Kidd, M., Landon, M. & Tomlinson, A.
21. Coppus, A. et al. Dementia and mortality in persons 44. Powell, D. et al. Frontal white matter integrity in Isolated senile plaque cores in Alzheimer’s disease and
with Down’s syndrome. J. Intellect. Disabil. Res. 50, adults with Down syndrome with and without Down’s syndrome show differences in morphology.
768–777 (2006). dementia. Neurobiol. Aging 35, 1562–1569 (2014). J. Neurol. Neurosurg. Psychiatry 49, 886–892
22. Visser, F. E., Aldenkamp, A. P., vanHuffelen, A. C., 45. Jennings, D. et al. Age dependence of brain ß-amyloid (1986).
Kuilman, M. & Overweg, J. Prospective study of the deposition in Down syndrome: an [18F]florbetaben PET 70. Armstrong, R. A. Size frequency distributions of
prevalence of Alzheimer-type dementia in study. Neurology 84, 500–507 (2015). β-amyloid (4β) deposits: a comparative study of four
institutionalized individuals with Down syndrome. 46. Masters, M. C., Morris, J. C. & Roe, C. M. neurodegenerative disorders. Folia Neuropathol. 50,
Am. J. Mental Retard. 101, 400–412 (1997). ‘Noncognitive’ symptoms of early Alzheimer disease: a 240–249 (2012).
23. Krinsky-McHale, S. J. et al. Successful aging in longitudinal analysis. Neurology 84, 617–622 71. Bero, A. W. et al. Neuronal activity regulates the
a 70‑year-old man with Down syndrome: a case (2015). regional vulnerability to amyloid-β deposition. Nat.
study. Intellect. Dev. Disabil. 46, 215–228 47. Wallon, D. et al. The French series of autosomal Neurosci. 14, 750–756 (2011).
(2008). dominant early onset Alzheimer’s disease cases: 72. Gyure, K. A., Durham, R., Stewart, W. F.,
24. Coppus, A. M. W. et al. Early age at menopause is mutation spectrum and cerebrospinal fluid Smialek, J. E. & Troncoso, J. C. Intraneuronal
associated with increased risk of Dementia and biomarkers. J. Alzheimers Dis. 30, 847–856 (2012). Aβ-amyloid precedes development of amyloid
mortality in women with Down syndrome. 48. De Simone, R., Puig, X. S., Gélisse, P., Crespel, A. & plaques in Down syndrome. Arch. Pathol. Lab. Med.
J. Alzheimers Dis. 19, 545–550 (2010). Genton, P. Senile myoclonic epilepsy: delineation of a 125, 489–492 (2001).
25. Schupf, N. et al. Onset of dementia is associated with common condition associated with Alzheimer’s 73. Hirayama, A., Horikoshi, Y., Maeda, M., Ito, M. &
age at menopause in women with Down’s syndrome. disease in Down syndrome. Seizure 19, 383–389 Takashima, S. Characteristic developmental
Ann. Neurol. 54, 433–438 (2003). (2010). expression of amyloid β40, 42 and 43 in patients with
26. Cosgrave, M. P., Tyrrell, J., McCarron, M., Gill, M. 49. Moller, J. C., Hamer, H. M., Oertel, W. H. & Down syndrome. Brain Dev. 25, 180–185 (2003).
& Lawlor, B. A. Age at onset of dementia and age Rosenow, F. Late-onset myoclonic epilepsy in Down’s 74. Iwatsubo, T., Mann, D. M. A., Odaka, A., Suzuki, N. &
of menopause in women with Down’s syndrome. syndrome (LOMEDS). Seizure 10, 303–305 (2001). Ihara, Y. Amyloid β protein (Aβ) deposition: Aβ42(43)
J. Intellect. Disabil. Res. 43, 461–465 (1999). 50. d’Orsi, G. & Specchio, L. M. Progressive myoclonus precedes Aβ40 in Down syndrome. Ann. Neurol. 37,
27. Draheim, C. C., Geijer, J. R. & Dengel, D. R. epilepsy in Down syndrome patients with dementia. 294–299 (1995).
Comparison of intima-media thickness of the J. Neurol. 261, 1584–1597 (2014). 75. Mori, C. et al. Intraneuronal Aβ42 accumulation in
carotid artery and cardiovascular disease risk factors 51. Friedman, D., Honig, L. S. & Scarmeas, N. Seizures Down syndrome brain. Amyloid 9, 88–102 (2002).
in adults with versus without the Down syndrome. and epilepsy in Alzheimer’s disease. CNS Neurosci. 76. Wegiel, J. et al. Intraneuronal Aβ immunoreactivity
Am. J. Cardiol. 106, 1512–1516 (2010). Ther. 18, 285–294 (2012). is not a predictor of brain amyloidosis-β or
28. Zigman, W. B. et al. Cholesterol level, statin use and 52. Vossel, K. A. et al. Seizures and epileptiform activity in neurofibrillary degeneration. Acta Neuropathol.
Alzheimer’s disease in adults with Down syndrome. the early stages of Alzheimer disease. JAMA Neurol. 113, 389–402 (2007).
Neurosci. Lett. 416, 279–284 (2007). 70, 1158–1166 (2013). 77. Wisniewski, K. E., Wisniewski, H. M. & Wen, G. Y.
29. Ylaherttuala, S., Luoma, J., Nikkari, T. & Kivimaki, T. 53. Irizarry, M. C. et al. Incidence of new-onset seizures in Occurrence of neuropathological changes and
Downs-syndrome and atherosclerosis. Atherosclerosis mild to moderate Alzheimer disease. Arch. Neurol. 69, dementia of Alzheimer’s disease in Down’s syndrome.
76, 269–272 (1989). 368–372 (2012). Ann. Neurol. 17, 278–282 (1985).

572 | SEPTEMBER 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

78. Wisniewski, K. E., Dalton, A. J., Mclachlan, D. R. C., 101. Lippa, C. F. et al. Transactive response DNA-binding 126. Ryoo, S. R. et al. Dual-specificity tyrosine(Y)-
Wen, G. Y. & Wisniewski, H. M. Alzheimer’s disease in protein 43 burden in familial Alzheimer disease and phosphorylation regulated kinase 1A‑mediated
Down’s syndrome: clinicopathologic studies. Down syndrome. Arch. Neurol. 66, 1483–1488 (2009). phosphorylation of amyloid precursor protein:
Neurology 35, 957–961 (1985). 102. Davidson, Y. S. et al. TDP‑43 pathological changes in evidence for a functional link between Down syndrome
79. Burger, P. C. & Vogel, F. S. The development of early onset familial and sporadic Alzheimer’s disease, and Alzheimer’s disease. J. Neurochem. 104,
pathologic changes of Alzheimer’s disease and senile late onset Alzheimer’s disease and Down’s syndrome: 1333–1344 (2008).
dementia in patients with Down’s syndrome. Am. association with age, hippocampal sclerosis and 127. Wang, X. et al. Sorting nexin 27 regulates Aβ
J. Pathol. 73, 457–476 (1973). clinical phenotype. Acta Neuropathol. 122, 703–713 production through modulating γ-secretase activity.
80. Lemere, C. A. et al. Sequence of deposition of (2011). Cell Rep. 9, 1023–1033 (2014).
heterogeneous amyloid β-peptides and APO E in Down 103. Gibb, W. R. G., Mountjoy, C. Q., Mann, D. M. A. & 128. Mok, K. Y. et al. Polymorphisms in BACE2 may affect
syndrome: implications for initial events in amyloid Lees, A. J. A pathological study of the association the age of onset Alzheimer’s dementia in Down
plaque formation. Neurobiol. Dis. 3, 16–32 (1996). between Lewy body disease and Alzheimer’s disease. syndrome. Neurobiol. Aging 35, 1513.e1–1513.e5
81. Ball, M. J. & Nuttall, K. Neurofibrillary tangles, J. Neurol. Neurosurg. Psychiatry 52, 701–708 (2014).
granulovacuolar degeneration, and neuron loss in Down (1989). 129. Sun, X. L., He, G. Q. & Song, W. H. BACE2, as a novel
syndrome: quantitative comparison with Alzheimer 104. Prasher, V. P., Airuehia, E. & Carey, M. The first APP θ-secretase, is not responsible for the
dementia. Ann. Neurol. 7, 462–465 (1980). confirmed case of Down syndrome with dementia with pathogenesis of Alzheimer’s disease in Down
82. Mann, D. M. A. & Esiri, M. M. The pattern of Lewy bodies. J. Appl. Res. Intellect. Disabil. 23, syndrome. FASEB J. 20, 1369–1376 (2006).
acquisition of plaques and tangles in the brains of 296–300 (2010). 130. Azkona, G., Levannon, D., Groner, Y. & Dierssen, M.
patients under 50 years of age with Down’s syndrome. 105. Cataldo, A. M. et al. Aβ localization in abnormal In vivo effects of APP are not exacerbated by BACE2
J. Neurol. Sci. 89, 169–179 (1989). endosomes: association with earliest Aβ elevations in co‑overexpression: behavioural characterization of a
83. Whalley, L. J. The dementia of Down’s syndrome and AD and Down syndrome. Neurobiol. Aging 25, double transgenic mouse model. Amino Acids 39,
its relevance to etiological studies of Alzheimer’s 1263–1272 (2004). 1571–1580 (2010).
disease. Ann. NY Acad. Sci. 396, 39–53 (1982). 106. Lambert, J. C. et al. Meta-analysis of 74,046 individuals 131. Holler, C. J. et al. BACE2 expression increases in
84. Ropper, A. H. & Williams, R. S. Relationship between identifies 11 new susceptibility loci for Alzheimer’s human neurodegenerative disease. Am. J. Pathol.
plaques, tangles, and dementia in Down syndrome. disease. Nat. Genet. 45, 1452–1458 (2013). 180, 337–350 (2012).
Neurology 30, 639–644 (1980). 107. Hooli, B. V. et al. Rare autosomal copy number 132. Patel, A. et al. Association of variants within APOE,
85. Godridge, H., Reynolds, G. P., Czudek, C., variations in early-onset familial Alzheimer’s disease. SORL1, RUNX1, BACE1 and ALDH18A1 with
Calcutt, N. A. & Benton, M. Alzheimer-like Mol. Psychiatry 19, 676–681 (2014). dementia in Alzheimer’s disease in subjects with Down
neurotransmitter deficits in adult Down’s syndrome 108. Llado, A. et al. Large APP locus duplication in a syndrome. Neurosci. Lett. 487, 144–148 (2011).
brain tissue. J. Neurol. Neurosurg. Psychiatry 50, sporadic case of cerebral haemorrhage. Neurogenetics 133. Prasher, V. P. et al. Significant effect of APOE epsilon 4
775–778 (1987). 15, 145–149 (2014). genotype on the risk of dementia in Alzheimer’s
86. Murphy, G. M. et al. Antigenic profile of plaques and 109. Cabrejo, L. et al. Phenotype associated with APP disease and mortality in persons with Down syndrome.
neurofibrillary tangles in the amygdala in Down’s duplication in five families. Brain 129, 2966–2976 Int. J. Geriatr. Psychiatry 23, 1134–1140 (2008).
syndrome: a comparison with Alzheimer’s disease. (2006). 134. Deb, S. et al. APOE epsilon 4 influences the manifestation
Brain Res. 537, 102–108 (1990). 110. Noebels, J. A perfect storm: converging paths of of Alzheimer’s disease in adults with Down’s syndrome.
87. Motte, J. & Williams, R. S. Age-related-changes in the epilepsy and Alzheimer’s dementia intersect in the Br. J. Psychiatry 176, 468–472 (2000).
density and morphology of plaques and neurofibrillary hippocampal formation. Epilepsia 52, 39–46 135. Coppus, A. M. W. et al. The impact of apolipoprotein E
tangles in Down syndrome brain. Acta Neuropathol. (2011). on dementia in persons with Down’s syndrome.
77, 535–546 (1989). 111. Griffiths-Jones, S. The microRNA registry. Nucleic Neurobiol. Aging 29, 828–835 (2008).
88. Head, E. et al. Parallel compensatory and pathological Acids Res. 32, D109–D111 (2004). 136. Schupf, N. et al. Onset of dementia is associated with
events associated with Tau pathology in middle aged 112. Vilardell, M. et al. Meta-analysis of heterogeneous apolipoprotein E epsilon 4 in Down’s syndrome.
individuals with Down syndrome. J. Neuropathol. Exp. Down syndrome data reveals consistent genome-wide Ann. Neurol. 40, 799–801 (1996).
Neurol. 62, 917–926 (2003). dosage effects related to neurological processes. BMC 137. Hyman, B. T. et al. Quantitative analysis of senile
89. Serrano-Pozo, A., Frosch, M. P., Masliah, E. & Genomics 12, 229 (2011). plaques in Alzheimer disease: observation of log-
Hyman, B. T. Neuropathological alterations in 113. Letourneau, A. et al. Domains of genome-wide gene normal size distribution and molecular epidemiology
Alzheimer disease. Cold Spring Harb. Perspect. Med. expression dysregulation in Down’s syndrome. Nature of differences associated with apolipoprotein E
1, a006189 (2011). 508, 345–350 (2014). genotype and trisomy 21 (Down syndrome). Proc. Natl
90. Mann, D. M. A. Cerebral amyloidosis, aging and 114. Horvath, S. et al. Accelerated epigenetic aging in Acad. Sci. USA 92, 3586–3590 (1995).
Alzheimer’s disease: a contribution from studies on Down syndrome. Aging Cell 14, 491–495 (2015). 138. Royston, M. C. et al. Apolipoprotein‑e epsilon‑2 allele
Down’s syndrome. Neurobiol. Aging 10, 397–399 115. Cheon, M. S., Dierssen, M., Kim, S. H. & Lubec, G. promotes longevity and protects patients with Down’s
(1989). Protein expression of BACE1, BACE2 and APP in syndrome from dementia. Neuroreport 5,
91. Evenhuis, H. M. The natural history of dementia in Down syndrome brains. Amino Acids 35, 339–343 2583–2585 (1994).
Down’s syndrome. Arch. Neurol. 47, 263–267 (2008). 139. Jones, E. L. et al. Evidence that PICALM affects age at
(1990). 116. Choi, J. H. K. et al. Age-dependent dysregulation of onset of Alzheimer’s dementia in Down syndrome.
92. McCarron, M. O., Nicoll, J. A. R. & Graham, D. I. A brain amyloid precursor protein in the Ts65Dn Down Neurobiol. Aging 34, 2441.e1–2441.e5 (2013).
quartet of Down’s syndrome, Alzheimer’s disease, syndrome mouse model. J. Neurochem. 110, 140. Lee, J. H. et al. Association between genetic variants
cerebral amyloid angiopathy, and cerebral 1818–1827 (2009). in sortilin-related receptor 1 (SORL1) and Alzheimer’s
haemorrhage: interacting genetic risk factors. J. Neurol. 117. Seo, H. & Isacson, O. Abnormal APP, cholinergic and disease in adults with Down syndrome. Neurosci. Lett.
Neurosurg. Psychiatry 65, 405–406 (1998). cognitive function in Ts65Dn Down’s model mice. Exp. 425, 105–109 (2007).
93. Mendel, T., Bertrand, E., Szpak, G. M., Stepien, T. & Neurol. 193, 469–480 (2005). 141. Salehi, A. et al. Increased App expression in a mouse
Wierzba-Bobrowicz, T. Cerebral amyloid angiopathy as 118. Teller, J. K. et al. Presence of soluble amyloid model of Down’s syndrome disrupts NGF transport
a cause of an extensive brain hemorrhage in adult β-peptide precedes amyloid plaque formation in and causes cholinergic neuron degeneration. Neuron
patient with Down’s syndrome — a case report. Down’s syndrome. Nat. Med. 2, 93–95 (1996). 51, 29–42 (2006).
Folia Neuropathol. 48, 206–211 (2010). 119. Busciglio, J. et al. Altered metabolism of the amyloid β 142. Torroja, L., Chu, H., Kotovsky, I. & White, K. Neuronal
94. Naito, K. S., Sekijima, Y. & Ikeda, S. I. Cerebral precursor protein is associated with mitochondrial overexpression of APPL, the Drosophila homologue of
amyloid angiopathy-related hemorrhage in a middle- dysfunction in Down’s syndrome. Neuron 33, the amyloid precursor protein (APP), disrupts axonal
aged patient with Down’s syndrome. Amyloid 15, 677–688 (2002). transport. Curr. Biol. 9, 489–492 (1999).
275–277 (2008). 120. Govoni, S. et al. Fibroblasts of patients affected by 143. Jiang, Y. et al. Alzheimer’s‑related endosome
95. Donahue, J. E., Khurana, J. S. & Adelman, L. S. Down’s syndrome oversecrete amyloid precursor dysfunction in Down syndrome is Aβ-independent but
Intracerebral hemorrhage in two patients with Down’s protein and are hyporesponsive to protein kinase C requires APP and is reversed by BACE‑1 inhibition.
syndrome and cerebral amyloid angiopathy. Acta stimulation. Neurology 47, 1069–1075 (1996). Proc. Natl Acad. Sci. USA 107, 1630–1635 (2010).
Neuropathol. 95, 213–216 (1998). 121. Murray, A., Letourneau, A. & Canzonetta, C. Isogenic 144. Barbosa, S., Pratte, D., Schwarz, H., Pipkorn, R. &
96. Handen, B. L. et al. Imaging brain amyloid in induced pluripotent stem cell lines from an adult with Singer-Kruger, B. Oligomeric Dop1p is part of the
nondemented young adults with Down syndrome mosaic Down syndrome model accelerated neuronal endosomal Neo1p–Ysl2p–Arl1p membrane
using Pittsburgh compound B. Alzheimers Dement. 8, ageing and neurodegeneration. Stem Cells 33, remodeling complex. Traffic 11, 1092–1106 (2010).
496–501 (2012). 2077–2084 (2015). 145. Swaminathan, S. et al. Analysis of copy number
97. Nelson, L. D. et al. Positron emission tomography of 122. Shi, Y. C. et al. A human stem cell model of early variation in Alzheimer’s disease: the NIA-LOAD/
brain β-amyloid and Tau levels in adults with Down Alzheimer’s disease pathology in Down syndrome. Sci. NCRAD family study. Curr. Alzheimer Res. 9,
syndrome. Arch. Neurol. 68, 768–774 (2011). Transl. Med. 4, 124ra29 (2012). 801–814 (2012).
98. Matveev, S. V. et al. A distinct subfraction of Aβ is 123. Wolvetang, E. W. et al. The chromosome 21 146. Chapman, J. et al. A genome-wide study shows a
responsible for the high-affinity Pittsburgh compound transcription factor ETS2 transactivates the β-APP limited contribution of rare copy number variants to
B‑binding site in Alzheimer’s disease brain. promoter: implications for Down syndrome. Biochim. Alzheimer’s disease risk. Hum. Mol. Genet. 22,
J. Neurochem. 131, 356–368 (2014). Biophys. Acta 1628, 105–110 (2003). 816–824 (2013).
99. Hartley, S. L. et al. Cognitive functioning in relation to 124. Dorval, V., Mazzella, M. J., Mathews, P. M., Hay, R. T. 147. Yang, D. S. et al. Reversal of autophagy dysfunction in
brain amyloid-β in healthy adults with Down syndrome. & Fraser, P. E. Modulation of Aβ generation by small the TgCRND8 mouse model of Alzheimer’s disease
Brain 137, 2556–2563 (2014). ubiquitin-like modifiers does not require conjugation ameliorates amyloid pathologies and memory deficits.
100. Landt, J. et al. Using positron emission tomography to target proteins. Biochem. J. 404, 309–316 Brain 134, 258–277 (2011).
and carbon 11‑labeled Pittsburgh compound B to (2007). 148. Cossec, J. C. et al. Trisomy for synaptojanin1 in Down
image brain fibrillar β-amyloid in adults with Down 125. Li, Y. H. et al. Positive and negative regulation of APP syndrome is functionally linked to the enlargement of
syndrome safety, acceptability, and feasibility. Arch. amyloidogenesis by sumoylation. Proc. Natl Acad. Sci. early endosomes. Hum. Mol. Genet. 21, 3156–3172
Neurol. 68, 890–896 (2011). USA 100, 259–264 (2003). (2012).

NATURE REVIEWS | NEUROSCIENCE VOLUME 16 | SEPTEMBER 2015 | 573

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

149. McIntire, L. B. J. et al. Reduction of synaptojanin 1 172. Wang, B. P. et al. The amyloid precursor protein 196. Sheng, J. G., Mrak, R. E. & Griffin, W. S. T. S100β
ameliorates synaptic and behavioral impairments in a controls adult hippocampal neurogenesis through protein expression in Alzheimer disease: potential role
mouse model of Alzheimer’s disease. J. Neurosci. 32, GABAergic interneurons. J. Neurosci. 34, in the pathogenesis of neuritic plaques. J. Neurosci.
15271–15276 (2012). 13314–13325 (2014). Res. 39, 398–404 (1994).
150. Zhu, L. et al. Reduction of synaptojanin 1 accelerates 173. Nicolas, M. & Hassan, B. A. Amyloid precursor protein 197. Mori, T. et al. Overexpression of human S100B
Aβ clearance and attenuates cognitive deterioration in and neural development. Development 141, exacerbates cerebral amyloidosis and gliosis in the
an Alzheimer mouse model. J. Biol. Chem. 288, 2543–2548 (2014). Tg2576 mouse model of Alzheimer’s disease.
32050–32063 (2013). 174. Ma’ayan, A., Gardiner, K. J. & Iyengar, R. The cognitive Glia 58, 300–314 (2010).
151. Busciglio, J. & Yankner, B. A. Apoptosis and increased phenotype of Down syndrome: insights from 198. Esposito, G. et al. S100B induces tau protein
generation of reactive oxygen species in Down’s intracellular network analysis. NeuroRx 3, 396–406 hyperphosphorylation via Dickopff‑1 up‑regulation
syndrome neurons in vitro. Nature 378, 776–779 (2006). and disrupts the Wnt pathway in human neural
(1995). 175. Drewes, G. et al. Dephosphorylation of Tau-protein stem cells. J. Cell. Mol. Med. 12, 914–927
152. Shukkur, E. A. et al. Mitochondrial dysfunction and tau and Alzheimer paired helical filaments by calcineurin (2008).
hyperphosphorylation in Ts1Cje, a mouse model for and phosphatase‑2A. Febs Lett. 336, 425–432 199. Chen, C. et al. Role of astroglia in Down’s syndrome
Down syndrome. Hum. Mol. Genet. 15, 2752–2762 (1993). revealed by patient-derived human-induced
(2006). 176. Liu, F. et al. Overexpression of Dyrk1A contributes to pluripotent stem cells. Nat. Commun. 5, 4430
153. Phillips, A. C. et al. Defective mitochondrial function neurofibrillary degeneration in Down syndrome. (2014).
in vivo in skeletal muscle in adults with Down’s syndrome: FASEB J. 22, 3224–3233 (2008). 200. Mills, S. M. et al. Preclinical trials in autosomal
a 31P‑MRS study. PLoS ONE 8, e84031 (2013). 177. Woods, Y. L. et al. The kinase DYRK phosphorylates dominant AD: implementation of the DIAN‑TU trial.
154. Weick, J. P. et al. Deficits in human trisomy 21 iPSCs protein-synthesis initiation factor eIF2Bepsilon at Rev. Neurol. 169, 737–743 (2013).
and neurons. Proc. Natl Acad. Sci. USA 110, Ser539 and the microtubule-associated protein tau at 201. Bothwell, M. & Giniger, E. Alzheimer’s disease:
9962–9967 (2013). Thr212: potential role for DYRK as a glycogen synthase neurodevelopment converges with neurodegeneration.
155. Zigman, W. B. Atypical aging in Down syndrome. kinase 3‑priming kinase. Biochem. J. 355, 609–615 Cell 102, 271–273 (2000).
Dev. Disabil. Res. Rev. 18, 51–67 (2013). (2001). 202. Purro, S. A., Galli, S. & Salinas, P. C. Dysfunction of
156. Picard, M. & Mcewen, B. S. Mitochondria impact brain 178. Dowjat, W. K. et al. Trisomy-driven overexpression of Wnt signaling and synaptic disassembly in
function and cognition. Proc. Natl Acad. Sci. USA 111, DYRK1A kinase in the brain of subjects with Down neurodegenerative diseases. J. Mol. Cell Biol. 6,
7–8 (2014). syndrome. Neurosci. Lett. 413, 77–81 (2007). 75–80 (2014).
157. Lott, I. T. et al. Down syndrome and dementia: a 179. Shi, J. et al. Increased dosage of Dyrk1A alters 203. Karmiloff-Smith, A. et al. Genetic and environmental
randomized, controlled trial of antioxidant alternative splicing factor (ASF)-regulated alternative vulnerabilities in children with neurodevelopmental
supplementation. Am. J. Med. Genet. A 155A, splicing of Tau in Down syndrome. J. Biol. Chem. 283, disorders. Proc. Natl Acad. Sci. USA 109,
1939–1948 (2011). 28660–28669 (2008). 17261–17265 (2012).
158. Carlson, G. A. et al. Genetic modification of the 180. Wegiel, J. et al. Link between DYRK1A overexpression 204. Moran, P. M., Higgins, L. S., Cordell, B. & Moser, P. C.
phenotypes produced by amyloid precursor protein and several-fold enhancement of neurofibrillary Age-related learning-deficits in transgenic mice
overexpression in transgenic mice. Hum. Mol. Genet. degeneration with 3‑repeat Tau protein in Down expressing the 751‑amino acid isoform of human
6, 1951–1959 (1997). syndrome. J. Neuropathol. Exp. Neurol. 70, 36–50 β-amyloid precursor protein. Proc. Natl Acad. Sci. USA
159. Murakami, K. et al. SOD1 (copper/zinc superoxide (2011). 92, 5341–5345 (1995).
dismutase) deficiency drives amyloid β protein 181. Moore, S. et al. APP metabolism regulates Tau 205. Yamaguchi, F. et al. Transgenic mice for the amyloid
oligomerization and memory loss in mouse model of protestasis in human cerebral cortex neurons. precursor protein 695 isoform have impaired spatial
Alzheimer disease. J. Biol. Chem. 286, Cell Rep. 11, 689–696 (2015). memory. Neuroreport 2, 781–784 (1991).
44557–44568 (2011). 182. Lu, T. et al. REST and stress resistance in ageing 206. Mucke, L. et al. High-level neuronal expression
160. Zis, P., Dickinson, M., Shende, S., Walker, Z. & and Alzheimer’s disease. Nature 507, 448–454 of Aβ1–42 in wild-type human amyloid protein
Strydom, A. Oxidative stress and memory decline in (2014). precursor transgenic mice: synaptotoxicity without
adults with Down syndrome: longitudinal study. 183. Kimura, R. et al. The DYRK1A gene, encoded in plaque formation. J. Neurosci. 20, 4050–4058
J. Alzheimers Dis. 31, 277–283 (2012). chromosome 21 Down syndrome critical region, (2000).
161. deHaan, J. B. et al. Elevation in the ratio of Cu/ bridges between β-amyloid production and tau 207. Balducci, C. & Forloni, G. A.P. P. Transgenic mice:
Zn‑superoxide dismutase to glutathione peroxidase phosphorylation in Alzheimer disease. Hum. Mol. their use and limitations. Neuromolecular Med. 13,
activity induces features of cellular senescence and Genet. 16, 15–23 (2007). 117–137 (2011).
this effect is mediated by hydrogen peroxide. Hum. 184. Vazquez-Higuera, J. L. et al. DYRK1A genetic variants 208. Brault, V., Pereira, P., Duchon, A. & Herault, Y.
Mol. Genet. 5, 283–292 (1996). are not linked to Alzheimer’s disease in a Spanish Modeling chromosomes in mouse to explore the
162. Anderson, J. S. et al. Abnormal brain synchrony in case-control cohort. BMC Med. Genet. 10, 129 function of genes, genomic disorders, and
Down syndrome. Neuroimage Clin. 2, 703–715 (2009). chromosomal organization. PLoS Genet. 2, e86
(2013). 185. Cooper, S. A. et al. Toward onset prevention of (2006).
163. Belichenko, P. V. et al. Excitatory-inhibitory cognitive decline in adults with Down syndrome (the 209. Park, I. H. et al. Disease-specific induced pluripotent
relationship in the fascia dentata in the Ts65Dn mouse TOP-COG study): study protocol for a randomized stem cells. Cell 134, 877–886 (2008).
model of down syndrome. J. Comp. Neurol. 512, controlled trial. Trials 15, 202 (2014). 210. Li, L. B. et al. Trisomy correction in Down syndrome
453–466 (2008). 186. Phillips, M. C. Molecular mechanisms of cellular induced pluripotent stem cells. Cell Stem Cell 11,
164. Fernandez, F. et al. Pharmacotherapy for cognitive cholesterol efflux. J. Biol. Chem. 289, 24020–24029 615–619 (2012).
impairment in a mouse model of Down syndrome. (2014). 211. Jiang, J. et al. Translating dosage compensation to
Nat. Neurosci. 10, 411–413 (2007). 187. Kim, W. S. et al. Role of ABCG1 and ABCA1 in regulation trisomy 21. Nature 500, 296–300 (2013).
165. Schmidt-Sidor, B., Wisniewski, K. E., Shepard, T. H. & of neuronal cholesterol efflux to apolipoprotein E discs 212. Chang, C. Y. et al. N‑butylidenephthalide attenuates
Sersen, E. A. Brain growth in Down syndrome and suppression of amyloid-β peptide generation. Alzheimer’s disease-like cytopathy in Down syndrome
subjects 15 to 22 weeks of gestational age and birth J. Biol. Chem. 282, 2851–2861 (2007). induced pluripotent stem cell-derived neurons.
to 60 months. Clin. Neuropathol. 9, 181–190 188. Tansley, G. H. et al. The cholesterol transporter Sci. Rep. 5, 8744 (2015).
(1990). ABCG1 modulates the subcellular distribution and
166. Takashima, S., Becker, L. E., Armstrong, D. L. & proteolytic processing of β-amyloid precursor protein.
Acknowledgements
Chan, F. Abnormal neuronal development in the visual J. Lipid Res. 48, 1022–1034 (2007).
The authors are funded by a Wellcome Trust Strategic Award
cortex of the human fetus and infant with down’s 189. Burgess, B. L. et al. ABCG1 influences the brain
(grant number: 098330/Z/12/Z) awarded to The London Down
syndrome. A quantitative and qualitative Golgi study. cholesterol biosynthetic pathway but does not affect
Syndrome (LonDownS) Consortium (J.H., A.K-S., D.N., V.L.J.T.
Brain Res. 225, 1–21 (1981). amyloid precursor protein or apolipoprotein E
E.M.C.F. and A.S.) and the Medical Research Council (pro-
167. Adorno, M. et al. Usp16 contributes to somatic stem- metabolism in vivo. J. Lipid Res. 49, 1254–1267
gramme number U117527252; awarded to V.L.J.T.), as well
cell defects in Down’s syndrome. Nature 501, (2008).
as by awards from Alzheimer Research UK (awarded to F.K.W
380–384 (2013). 190. Perry, V. H. & Holmes, C. Microglial priming in
and E.M.C.F), Alzheimer Society (awarded to E.M.C.F and
168. Canzonetta, C. et al. DYRK1A‑dosage imbalance neurodegenerative disease. Nat. Rev. Neurol. 10,
F.K.W.), Bailey Thomas Trust (awarded to A.S.), Epilepsy
perturbs NRSF/REST levels, deregulating 217–224 (2014).
Research UK (awarded to F.K.W.), Lee Kong Chian School of
pluripotency and embryonic stem cell fate in Down 191. Wilcock, D. M. & Griffin, W. S. T. Down’s syndrome,
Medicine, Nanyang Technological University Start‑up Grant,
syndrome. Am. J. Hum. Genet. 83, 388–400 neuroinflammation, and Alzheimer neuropathogenesis.
and Singapore Ministry of Education Academic Research Fund
(2008). J. Neuroinflammation 10, 84 (2013).
Tier 1(2014‑T1‑001‑173; awarded to D.N).
169. Hibaoui, Y. et al. Modelling and rescuing 192. Griffin, W. S. T. et al. Brain interleukin 1 and S-100
neurodevelopmental defect of Down syndrome using immunoreactivity are elevated in Down syndrome and
induced pluripotent stem cells from monozygotic twins Alzheimer disease. Proc. Natl Acad. Sci. USA 86,
Competing interests statement
The authors declare competing interests: see Web version
discordant for trisomy 21. EMBO Mol. Med. 6, 7611–7615 (1989).
for details.
259–277 (2014). 193. Naude, P. J. W. et al. Serum NGAL is associated with
170. Chang, K. T. & Min, K. T. Upregulation of three distinct plasma amyloid-β peptides according to the
Drosophila homologs of human chromosome 21 genes clinical diagnosis of dementia in Down syndrome. FURTHER INFORMATION
alters synaptic function: implications for Down J. Alzheimers Dis. 45, 733–743 (2015). London Down Syndrome (LonDownS) Consortium:
syndrome. Proc. Natl Acad. Sci. USA 106, 194. Mann, D. M. A. et al. Microglial cells and amyloid-β http://www.ucl.ac.uk/london-down-syndrome-consortium
17117–17122 (2009). protein (A) deposition — association with
171. Cvetkovska, V., Hibbert, A. D., Emran, F. & Chen, B. E. Aβ40‑containing plaques. Acta Neuropathol. SUPPLEMENTARY INFORMATION
Overexpression of Down syndrome cell adhesion 90, 472–477 (1995). See online article: S1 (table) | S2 (table) | S3 (table)
molecule impairs precise synaptic targeting. Nat. 195. Xue, Q. S. & Streit, W. J. Microglial pathology in Down ALL LINKS ARE ACTIVE IN THE ONLINE PDF
Neurosci. 16, 677–682 (2013). syndrome. Acta Neuropathol. 122, 455–466 (2011).

574 | SEPTEMBER 2015 | VOLUME 16 www.nature.com/reviews/neuro

© 2015 Macmillan Publishers Limited. All rights reserved

You might also like