You are on page 1of 6

MENTAL RETARDATION AND DEVELOPMENTAL DISABILITIES

RESEARCH REVIEWS 13: 215 – 220 (2007)

A YEAR OF UNPRECEDENTED PROGRESS IN


DOWN SYNDROME BASIC RESEARCH
Roger H. Reeves1,2* and Craig C. Garner3
1
Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
2
Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
3
Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University, Palo Alto, California

The years 2006 and 2007 saw the publication of three new and conserved linkage with Hsa21 on mouse chromosomes 16
different approaches to prevention or amelioration of Down syndrome (Mmu16), 17, and 10. Ts65Dn mice are trisomic for nearly half
effects on the brain and cognition. We describe the animal model sys-
tems that were critical to this progress, review these independent break-
of the mouse equivalents of Hsa21 genes, whereas other models
through studies, and discuss the implications for therapeutic approaches contain subsets of these (Fig. 1). The elevated gene expression
suggested by each. ' 2007 Wiley-Liss, Inc. due to trisomy is very comparable between these mice and
MRDD Research Reviews 2007;13:215–220. human beings. But will this comparable genetic effect have the
same outcome on the features of the mouse, i.e., will analogous
Key Words: mouse model; cognitive testing; GABA inhibitor; APP; sonic phenotypes be produced in the same way?
hedgehog; synapse function; brain development For many features of DS, there is strong evidence to vali-
date the idea that developmental processes affected by trisomy
21 in humans can be studied in mice. Quantitative phenotype
assessments in Ts65Dn mice demonstrate directly comparable
dysmorphologies in craniofacial structure, in which the same
ANIMAL MODELS OF DOWN SYNDROME
bones affected in DS are affected in the same way in mice

F
eatures of Down syndrome (DS) arise due to overex-
[Richtsmeier et al., 2000, 2002]. Histopathological analysis of
pression of genes on human chromosome 21 (Hsa21)
the cerebellum in Ts65Dn mice predicted a pattern of cell loss
usually through the triplication of the entire chromo-
that was subsequently shown to occur in humans with DS
some. Divergence from euploid of the structures and functions
[Baxter et al., 2000]. Age-related loss of forebrain cholinergic
that are affected in DS may result from upregulation of Hsa21
neurons occurs in Ts65Dn mice and in DS as well as in Alzhei-
genes in adult cells and tissues and/or because of inappropriate
mer disease. Deficits in retrograde transport of NGF are respon-
cues during development that are perpetuated in descendents
sible for this loss in mice [Cooper et al., 2001] and suggest a
of the cells that are affected initially [Potier et al., 2006; Roper
therapeutic approach to this problem in humans. Detailed stud-
and Reeves, 2006]. The central nervous system begins to de-
ies of synaptic dysfunction in mouse models point towards spe-
velop very early in embryogenesis and substantial structural
cific neuronal pathologies in DS [Belichenko et al., 2004].
and functional changes are still evident for several years fol-
Trisomic mouse models also provide a good representa-
lowing birth, and to some degree, throughout life. Thus, the
tion of important aspects of cognitive dysfunction in DS.
brain is especially susceptible to perturbations caused by subtle
Work by Nadel and coworkers has demonstrated that func-
changes in gene expression. Small structural differences can
tions of the hippocampus are among those most severely
result in a substantial degree of dysfunction at the level of
affected in DS [Nadel, 2003; Pennington et al., 2003]. A ro-
individual neurons, which will affect the complex circuits
bust learning and memory deficit in tests requiring hippocam-
involved in cognitive function.
pal function has been demonstrated in Ts65Dn mice [Escori-
Development cannot be studied in human beings at the
huela et al., 1995; Reeves et al., 1995; Holtzman et al., 1996].
level necessary to understand the complex processes that are
Hippocampal dysfunction extends to studies of the electro-
perturbed by trisomy. In vitro molecular biology and cell cul-
ture systems are important tools but do not replicate the de-
velopmental processes that are the targets for intervention to
ameliorate DS features. Animal models are essential to study Grant sponsors: Anna and John J. Sie Foundation; The National Heart, Lung and
the disruption of the developmental process caused by trisomy. Blood Institute; National Institute of Child Health and Human Development;
The laboratory mouse provides the best-characterized ex- National Cancer Institute.
*Correspondence to: Roger H. Reeves, Ph.D., Department of Physiology, Johns
perimental system for studies of mammalian aneuploidy. The Hopkins University School of Medicine, 725 N. Wolfe St., Baltimore, MD
first line of evidence supporting the use of mouse models to 21205. E-mail: rreeves@jhmi.edu
study DS is genetic. Detailed comparative and physical mapping Received 10 August 2007; Accepted 13 August 2007
Published online in Wiley InterScience (www.interscience.wiley.com).
and now comparative sequence analysis [Hattori et al., 2000; DOI: 10.1002/mrdd.20165
Mural et al., 2002; Toyoda et al., 2002] demonstrate highly
' 2007 Wiley -Liss, Inc.
features have parallels that have been
documented in trisomic Ts65Dn mice
including short stature; anomalies of the
craniofacial skeleton and skull that result
in flat facies, brachycephaly and pro-
truding tongue (small mandible); and
(with caveats) ‘‘mental retardation of the
DS type,’’ specifically impaired hippo-
campal function. Ts1Rhr mice, which
are trisomic only for the DSCR,
showed none of the features of the cra-
niofacial skeleton attributed to the
DSCR. Ms1Rhr/Ts65Dn mice are tri-
somic for all the genes triplicated in
Ts65Dn except those in the DSCR (Fig.
1). These mice had craniofacial features
similar to Ts65Dn. Thus, trisomy for
the DSCR is not sufficient and for the
most part is not necessary to produce
these features of trisomy.
The picture is more complex
when the question of mental retardation
of the DS type is addressed [Olson et al.,
2007]. Disruption of hippocampal func-
tions in DS can be modeled in the per-
Fig. 1. Aneuploid segments in mouse models used to study Down syndrome. Gene numbers
for each segment are based on conserved genes identified by Gardiner et al. [2003]. formance of Ts65Dn mice in the Morris
water maze (MWM) paradigm. In
MWM, a mouse placed in a swimming
physiological properties of neuronal cir- quantitative analysis of phenotypic out- tank is trained to navigate to a platform
cuits in the hippocampus of trisomic comes in these models. based on visual cues in the room (Fig.
mice [Siarey et al., 1997; Kleschevnikov 2a), a visiospatial integration task that is
et al., 2004]. THE CONTRIBUTION OF dependent on hippocampal function.
Trisomic mouse models have ‘‘CRITICAL REGIONS’’ ON Ts65Dn mice are significantly impaired
become an essential tool in research into Hsa21 TO PHENOTYPES OF DS in the MWM test, whereas Ts1Rhr
the basic mechanisms of gene dosage Efforts to identify regions of mice that are trisomic for only the
effects in DS. The highly parallel out- Hsa21 containing genes that contribute DSCR genes performed the same as
comes that result when the same evolu- to specific aspects of the DS phenotype euploid in both phases of the test [Olson
tionarily conserved genetic programs have focused on individuals with trans- et al., 2007]. Thus, trisomy for the
are perturbed in mice and man validate locations resulting in trisomy for a sub- DSCR is not sufficient to produce the
studies that focus on phenotypes. The set of Hsa21 genes (translocation DS). kind of hippocampal disruption seen in
three studies discussed here rely on Phenotype maps correlating dosage Ts65Dn mice and in DS, contrary to the
mouse genetic models to explore when, imbalance of specific regions with spe- prediction of the DSCR hypothesis.
where, and how trisomic development cific characteristics provide useful infor- Next, Ts65Dn mice were crossed to
and function deviate from euploid, and mation about segments in which to Ms1Rhr mice, returning the DSCR
each suggests possible therapeutic app- search for genes that perturb specific genes to the normal two copies in a
roaches to overcome these differences. developmental pathways [Delabar et al., mouse that was trisomic for all of the
To illustrate the application of 1993; Korenberg et al., 1994]. other genes triplicated in Ts65Dn.
genetic models and introduce tests of The resolution and interpretation These Ms1Rhr/Ts65Dn mice per-
hippocampal function in trisomic mice, of the ‘‘critical regions’’ identified by formed like euploid (Fig. 2b), showing
we review a recent study analyzing the phenotype maps is severely limited by that a gene (or genes) from this segment
genetic contribution to hippocampal- several factors. Notably, there is no col- is necessary to produce this specific hip-
based cognitive dysfunction measured in lection of human beings who are triso- pocampal deficit, and acts in combina-
the Morris maze deficit [Olson et al., mic for only the critical region, whereas tion with other genes from Mmu16 (and
2007]. We then review the three assessment of variable human pheno- Hsa21) to do so.
‘‘breakthrough’’ studies. The study of types requires assessment of many indi-
Salehi et al. (2006) uses a genetics-based viduals to draw meaningful conclusions. GABAA ANTAGONISTS AND
approach to identify a candidate target To address this issue directly, Olson LONG-TERM CORRECTION OF
gene, while the other two studies reach et al. [2004] used chromosome engi- LEARNING AND MEMORY
their conclusions by focusing on neering of Mmu16 to create mice with [FERNANDEZ ET AL., 2007]
description of pathogenesis and modera- a duplication or a deletion of the so- The previous experiments de-
tion of phenotypes without looking called DS critical region (DSCR) (Fig. scribe functional tests at the organismal
into the genetic basis for the origins of 1) and asked whether the 33 DSCR level that demonstrate the effects of tris-
the phenotype under consideration. All genes were necessary and/or sufficient omy. The deficit in MWM performance
three reach their important conclusions to produce DS features attributed to in Ts65Dn mice is paralleled by a
by careful attention to detailed and genes in that region. Several DSCR change in the way that neurons com-
216 MRDD Research Reviews DOI 10.1002/mrdd  TOWARDS THERAPY FOR DOWN SYNDROME  REEVES
Fig. 3. Ts65Dn mice are impaired relative
to euploid in the hippocampal-based object
recognition task (‘‘Milk’’), but treatment with
PTZ allows them to perform normally, even
several months after the end of treatment
[adapted from Fernandez et al., 2007].
Fig. 2. Testing hippocampal function in mice. (a) The Morris water maze tests the ability of a
mouse to navigate to a platform (black rectangle) based on visual cues placed around the room. back in the cage with one of the same
(b) Ts65Dn mice perform poorly in the MWM (final trials 7, 8, and 9), but when the so-called
DSCR is genetically ‘‘subtracted’’ in Ms1Rhr/Ts65Dn mice, they perform like euploid [adapted objects and a second, new object.
from Olson et al., 2007]. Euploid (normal) mice will remember
the old object and spend most of their
time examining the new one, but triso-
municate. These changes in synaptic are very well studied and thus the results mic mice show no evidence that they
plasticity are measured by sensitive elec- here can be related to a wealth of addi- recognize the first object. The second
trophysiological tests as changes in long- tional findings. The second compound is test is an ‘‘Alternating T maze,’’ a long
term potentiation (LTP) and long-term bilobalide (BB), one of several com- platform with two arms. In multiple tri-
depression (LTD), changes that are ro- pounds extracted from the leaves of the als, a mouse with normal hippocampal
bust in Ts65Dn mice [Sago et al., 1998; ginkgo biloba tree. A variety of effects, function comes to the ‘‘T,’’ remembers
Siarey et al., 1999, 2006; Kleschevnikov many conflicting, have been reported for the arm that it investigated in the pre-
et al., 2004]. In fact, Ts65Dn mice this compound, but the most remarkable ceding trial, and goes down the other
can show LTP in the dentate gyrus but is as a noncompetitive antagonist of arm. Mice in which the hippocampus is
are normally inhibited from inducing it GABAA receptors. The third com- impaired, including Ts65Dn mice, choose
due to excessive inhibitory input [Kle- pound, pentylenetetrazole (PTZ), is also the arms randomly, suggesting that they
schevnikov et al., 2004]. Studies that a noncompetitive GABAA receptor an- do not remember the preceding trial.
examined excitatory and inhibitory tagonist. It was administered to patients In the first series of experiments,
inputs to various parts of the hippocam- for many years as a circulatory and respi- Ts65Dn mice given BB or PTX at
pus found that inhibitory inputs were ratory stimulant [Levy, 1953]. It has also doses far below those used to induce
somewhat more prevalent and more been used ineffectively, at higher doses seizures showed marked improvement
active in trisomic than in euploid mice that cause convulsion, for the treatment compared to saline-treated trisomic mice,
[Hanson et al., 2007]. These inhibitory of psychiatric disorders such as schizo- performing the same as their euploid lit-
neurons are characterized by their use phrenia and at low nonepilepitic doses as termates in both the object recognition
of a neurotransmitter called GABA. a procognitive drug for the treatment of and T-maze tests. The authors observed
Use of GABAA receptor antagonist in senility in geriatric patients. Given that that if animals were injected with PTX
in vitro studies results in enhancement of it was ineffective for the treatment of ei- for 2 weeks followed by saline for
the development of normal LTP in the ther of these disorders and is potentially 2 weeks, they performed just as well as
dentate gyrus of Ts65Dn brain slices. harmful at high doses, approval for PTZ animals that had just finished the drug
These observations led Dr. Craig was revoked by the FDA in 1982. The treatment.
Garner and coworkers to examine the fact remains that large numbers of peo- Based on these results, a series of
possibility that administration of non- ple were treated with this compound for exposures to PTZ was designed. This
competitive GABAA receptor antagonists years, and although the drug showed lit- drug can be given orally, so euploid and
could have a positive effect in mice [Fer- tle efficacy, it also showed few side trisomic mice were trained to drink
nandez et al., 2007]. We considered effects in most people at low doses. chocolate milk with or without the
three antagonists of GABA, compounds Given this history, of the three com- drug daily for 3 weeks. Two to three
that counter the effects of this neuro- pounds tested in this study, PTZ is the months after the animals stopped get-
transmitter by blocking its receptor. most intriguing to consider as a drug. ting the drug, they were tested in the
The first compound is called picrotoxin To test these compounds, the object recognition test. Trisomic mice
(PTX) and is widely used in experi- investigators turned to the Ts65Dn that got PTZ performed as well as
mental paradigms to induce seizures. mouse model since hippocampal impair- euploid (Fig. 3). This long-term im-
(Indeed, all three of these molecules can ments in these mice parallel those in DS. provement was also evident in a sensi-
cause seizures, discussed further below.) They used two nonstressful tests of hip- tive electrophysiological test of LTP in
PTX has a very small window between pocampal function. The first is called the dentate gyrus, a measure of synaptic
the amount required to show a positive ‘‘Object recognition.’’ The mouse is plasticity that is normally lacking in
effect in LTP experiments and the placed in a cage with two objects for Ts65Dn mice. Trisomic mice that had
amount required to induce seizures, and 15 min, during which time it will spend received PTZ had significantly im-
thus is an unlikely candidate as a drug. about half the time examining each proved LTP relative to saline-treated
However, its effects and mode of action object. The next day, the mouse is placed Ts65Dn mice [Fernandez et al., 2007].
MRDD Research Reviews DOI 10.1002/mrdd  TOWARDS THERAPY FOR DOWN SYNDROME  REEVES 217
Serious questions remain to be is described in depth elsewhere in this
answered before clinical trials can be issue and is reviewed briefly here to put
considered. The first is the phenom- it in context with the other two studies.
enon known as kindling, in which Mobley and coworkers have used mouse
repeated medium doses of chemicals— models extensively to understand how
including all three of those used in trisomy causes changes in structure and
these experiments (20–30 mg/kg)— function of neuronal synapses, including
make animals more susceptible to seiz- some key observations about inhibitory
ures. It remains to be seen whether the input to the hippocampus as discussed
far lower doses used here (1–3 mg/kg), earlier [Kleschevnikov et al., 2004]. They
which fail to elicit seizures in euploid have also studied the loss of a specific
rats and mice, will increase seizure risk population of neurons that communicates
in trisomic mice. A related question is between the basal forebrain and the hip-
whether there is a genetic predisposition pocampus. These basal forebrain cholin-
to seizures that would make the use of ergic neurons (BFCN) utilize acetylcho-
this drug more dangerous in some peo- line as a neurotransmitter and are seen to
ple than in others and/or whether degenerate in Alzheimer disease (AD), in
young children are more susceptible DS, and in Ts65Dn mice. (In fact, the
than young adults. A second important study by Salehi et al. presents evidence
goal is the elucidation of the mecha- suggesting that rather than degenerating,
nisms by which short-term treatment of these neurons actually become quiescent,
noncompetitive GABAA receptor antag- turning off expression of the marker
onists leads to a long-term improvement commonly used to identify them.) This
Fig. 4. BFCN must transport NGF pro-
in function. This is not only important BFCN quiescence is believed to contrib- duced in the hippocampus back to the cell
for the direct consideration of safety of ute to the age-related decline in cogni- body in the basal forebrain to remain
these drugs, but is important to choos- tive function that is seen in many indi- active. (a) Retrograde transport of NGF is
ing (or designing) safer and more effec- viduals with DS. greatly impaired in Ts65Dn mice, but
affected to a lesser degree in Ts1Cje mice.
tive treatment strategies in the future. Like many neurons, the BFCN (b) Genetic ‘‘subtraction’’ of App from the
Even given these cautions, this study require specific signals to remain active trisomic gene set in Ts65Dn substantially
represents an extremely exciting break- and alive. Several years ago, Mobley and improves retrograde NGF transport
through on which to focus research coworkers [Cooper et al., 2001] used (Ts65Dn:Appþ/þ/2 vs. Ts65Dn:Appþ/þ/þ);
efforts in the near term. Ts65Dn trisomic mice to demonstrate *indicates a significant difference [adapted
from Salehi et al., 2006].
that neurons do not properly transport
Conclusion Study 1 an important signal, nerve growth factor
Decreased hippocampal perform- (NGF), from the nerve terminals in the with inherited forms of AD with very
ance in mouse models of DS is linked hippocampus back to the neuronal cell early age of onset.
to an imbalance in the weighting of body. It was known that this retrograde To determine whether dosage (3
excitatory and inhibitory circuits. At transport of NGF was a signal that copies vs. 2 copies) of App contributed
present, the underlying etiology of this maintains BFCN, and so the absence of to the NGF retrograde transport deficit
imbalance is unknown. Administration this transport in the trisomic neurons in Ts65Dn mice, a genetic cross was
of low, nonepileptic doses of noncom- was considered likely to be the cause of made between Ts65Dn, which have
petitive GABAA antagonists can restore their demise. three copies of App (Ts65Dn:Appþ/þ/
normal cognitive function in Ts65Dn At this point, the incomplete but þ), and mice with only one copy of
mice. This information suggests a new very powerful set of mouse models for App (Appþ/2) and the offspring were
and exciting avenue for the possible DS and also for AD played a prominent analyzed. The severe reduction in trans-
treatment of cognitive dysfunction in role. Ts1Cje mice [Sago et al., 1998] port of Ts65Dn mice was substantially
Down syndrome. While serious ques- are trisomic for about 80% of the improved in the trisomic mice that had
tions remain, the questions are well genes triplicated in Ts65Dn (Fig. 1). two instead of three copies of App
defined and there is every reason to When Mobley’s group compared BFCN- (Ts65Dn:Appþ/þ/2, Fig. 4b). Fur-
think they can be answered. Whether related neuropathology in euploid, thermore, mice that were euploid
PTZ ends up as a drug of choice, these Ts65Dn, and Ts1Cje mice, they found except for extra copies of the APP gene
experiments indicate that drugs directed that retrograde transport of NGF was were affected. Careful experiments es-
toward GABA pathways that presum- much closer to that of euploid mice in tablished that an APP cleavage product
ably lower inhibitory inputs in the tri- the Ts1Cje mice (Fig. 4a). Accordingly, from the COOH-terminal end of the
somic hippocampus hold great promise attention was focused on the genes that protein was responsible for the retro-
for improvement of cognition in DS. are present in three copies in Ts65Dn grade transport deficit, possibly through
but not Ts1Cje. Several of these have interaction at the level of the early
known functions in the CNS, including endosomes that carry NGF.
DEGENERATION OF BASAL Gabpa, Grik1, App, and Sod1. Of these,
FOREBRAIN CHOLINERGIC APP plays a major role in AD, as shown
NEURONS [SALEHI ET AL., 2006] by the fact that a cleavage product of Conclusion, Study 2
The second major breakthrough of the APP protein is a major constituent Restoration of NGF transport and
the past year was established by Dr. Wil- of the neuritic plaques that characterize survival of BFCN might counteract, to
liam Mobley’s group at Stanford [Salehi the histopathology of this disease. Fur- some degree, the age-related cognitive
et al., 2006]. The basis for this discovery ther, mutations in APP are associated decline seen frequently in DS. The
218 MRDD Research Reviews DOI 10.1002/mrdd  TOWARDS THERAPY FOR DOWN SYNDROME  REEVES
pharmaceutical industry is not likely to
make a large investment in DS because
the number of affected individuals is
too small to return a profit in their eco-
nomic model. In that light, the associa-
tion of APP C-terminal fragments with
the pathology that is shared in AD and
DS is fortuitous, because extensive
efforts are already focused on APP
processing as a possible intervention for
AD. The indications in this study of
APP interference with retrograde trans-
port via early endosomes suggest that
reducing levels of APP could have a
positive effect in reducing BFCN Fig. 5. Growth of the Ts65Dn brain is reduced because trisomic mice do not respond properly
degeneration, with beneficial conse- to the SHH growth factor. (a) Purified GCP from both trisomic and euploid mice respond to
quences in DS and AD. increasing concentrations of SHH, but trisomic GCP respond less. A representative experiment is
shown. (b) A single treatment at P0 with SAG is sufficient to restore the number of GCP in the
cerebellum 1 week later; *indicates a significant difference [adapted from Roper et al., 2006].

REVERSAL OF A GROWTH
FACTOR RESPONSE DEFICIT
AFFECTING BRAIN Ts65Dn mice, the cerebellum was dis- mates, the number of GCP undergoing
DEVELOPMENT [ROPER proportionately small. (The fact that the cell division was significantly reduced in
ET AL., 2006] brain was not reduced overall in Ts65Dn trisomic mice.
The third breakthrough study was not terribly surprising as much of the Collectively, these findings identi-
comes from Dr. Reeve’s laboratory reduction seen in DS occurs in the cere- fied a specific effect of trisomy on brain
[Roper et al., 2006]. It appears likely to bral cortex, which represents a much growth and defined the developmental
be farther removed from potential clini- larger proportion of the brain in human timing (P0), the affected cell type
cal application, but may have more beings than in other mammals.) Given (GCP), and the disrupted process (cell
wide-reaching effects regarding the the reduced Ts65Dn cerebellum, histo- division). Studies from a number of lab-
broad range of features that characterize logical approaches were used to deter- oratories had established that the
DS. This study also demonstrates the mine where the reduction occurred. growth factor, sonic hedgehog (SHH),
ability to reverse one very specific aspect Both the granule layer and molecular is the major signal to induce cell divi-
of the phenotype using a potential drug. layer were affected. Unexpectedly, Baxter sion of GCP. Crosses between Ts65Dn
In the course of fundamental research et al. demonstrated that not only was the and mice with a reporter of SHH path-
on the development of the trisomic volume occupied by granule cells and way activity suggested that this path-
brain, these studies identified a deficit in Purkinje cells reduced, but in addition way was somewhat downregulated in
response to a growth factor that affects there was a reduction in the cell density Ts65Dn mice. Accordingly, we isolated
many different tissues at different stages of both types of neurons, further reduc- GCP and cultured them in the presence
of development. We went on to demon- ing the total number of cerebellar neu- of increasing amounts of duly-lipidated
strate that administration of an experi- rons. This phenotype had never been SHH (Fig. 5a). This analysis showed
mental compound could return a spe- reported in DS. Using age-matched brain two important things. First, trisomic
cific aspect of the developmental process specimens from individuals with two or GCP responded less at every concentra-
to normal in the cerebellum. As in the three copies of Hsa21, the same reduc- tion of SHH, demonstrating a cell au-
preceding studies, these findings relied tion of granule cell density was seen in tonomous deficit in response to this im-
on genetic mouse models of DS. DS. portant growth factor. Second, trisomic
The experiments started several The ability of a mouse model to GCP did respond, suggesting that a
years ago with efforts to compare brain correctly predict a new phenotype of a small increase in SHH might elevate
development in trisomic Ts65Dn mice human condition is a strong indication proliferation rates of trisomic GCP to
to that in human beings with DS. The that the developmental processes are the the same levels as euploid.
cerebellum is significantly reduced in same in the two species. Accordingly, To test this, we injected a small
DS, even relative to the overall smaller we next asked at what point the pattern molecule that activates the SHH path-
size of the brain [Crome et al., 1966; of development diverged between triso- way (SHH agonist, SAG) into trisomic
Aylward et al., 1997]. Unlike many fea- mic and euploid mice [Roper et al., pups the day they were born and
tures of DS which may or may not be 2006]. Again using histological app- assessed the number of cells in the
present in an individual with trisomy roaches, we found that every parameter cerebellum 6 days later. The single
21, a small cerebellum appears to occur that could be measured 1 week after administration restored the number of
in everyone with this condition. Thus, birth (postnatal day 6, P6) was reduced GCP (Fig. 5b) and the volume of the
it was expected that a mouse with a in Ts65Dn mice, but these differences cerebellum to euploid levels. Further,
similar genetic constitution should dis- were not evident at P0. Very precise the number of dividing GCP was also
play corresponding effects on develop- statistically validated sampling of cere- restored to euploid level, suggesting
ment of the cerebellum. bellum revealed that while the number that the ameliorative effect of this sin-
Baxter et al. [2000] used high reso- of granule cell precursors (GCP) that gle treatment would continue through
lution 3d MRI to show that while the give rise to granule cell neurons was the cerebellar development [Roper et al.,
brain was the same size as euploid in same in Ts65Dn and euploid litter- 2006].
MRDD Research Reviews DOI 10.1002/mrdd  TOWARDS THERAPY FOR DOWN SYNDROME  REEVES 219
Conclusion, Study 3 antioxidants and cholinesterase inhibitors, Levy S. 1953. Pharmacological treatment of aged
SHH is important in brain devel- the prospects for improving cognitive patients in a state mental hospital. J Am
Med Assoc 153:1260–1265.
opment from very early stages. In addi- function in DS in the foreseeable future Mural RJ, Adams MD, Myers EW. 2002. A com-
tion, this potent morphogen and growth appear to be strongly positive. n parison of whole-genome shotgun-derived
factor affects the proliferation, migration, mouse chromosome 16 and the human ge-
differentiation, and survival of many nome. Science 296:1661–1671.
populations of cells beginning early in ACKNOWLEDGMENT Nadel L. 2003. Down’s syndrome: a genetic disor-
der in biobehavioral perspective. Genes
development and continuing throughout The authors thank Dr. William Brain Behav 2:156–166.
life. Thus, any stimulation of this path- Mobley for insightful comments. Olson LE, Richtsmeier JT, Leszl J, et al. 2004. A
way must be precisely targeted, both in chromosome 21 critical region does not
which cells are exposed and when during cause specific down syndrome phenotypes.
development this stimulation takes place. REFERENCES Science 306:687–690.
Aylward EH, Habbak R, Warren AC, et al. 1997. Olson LE, Roper RJ, Sengstaken CL, et al. 2007.
On the other hand, only those Trisomy for the Down syndrome ‘‘critical
Cerebellar volume in adults with Down syn-
cells with the appropriate receptors and drome. Arch Neurol 54:209–212. region’’ is necessary but not sufficient for
transducers of the SHH signal can Baxter LL, Moran TH, Richtsmeier JT, et al. brain phenotypes of trisomic mice. Hum
respond; those without the receptor are 2000. Discovery and genetic localization of Mol Genet 16:774–782.
impervious to its affects. If all trisomic Down syndrome cerebellar phenotypes using Pennington BF, Moon J, Edgin J, et al. 2003. The
the Ts65Dn mouse. Hum Mol Genet 9: neuropsychology of Down syndrome: evi-
cells show the same attenuated response dence for hippocampal dysfunction. Child
195–202.
to SHH, a more general application of a Belichenko PV, Masliah E, Kleschevnikov AM, Dev 74:75–93.
drug targeted to this specific response et al. 2004. Synaptic structural abnormalities Potier MC, Rivals I, Mercier G, et al. 2006. Tran-
deficit could have wide-reaching amelio- in the Ts65Dn mouse model of Down syn- scriptional disruptions in Down syndrome: a
rative effects. SHH is important in the drome. J Comp Neurol 480:281–298. case study in the Ts1Cje mouse cerebellum
Cooper JD, Salehi A, Delcroix JD, et al. 2001. during post-natal development. J Neuro-
formation of a substantial part of the chem 97 (Suppl 1):104–109.
Failed retrograde transport of NGF in a
brain. In addition, it affects all structures mouse model of Down’s syndrome: reversal Reeves R, Irving N, Moran T, et al. 1995. A
derived from neural crest, including the of cholinergic neurodegenerative phenotypes mouse model for Down syndrome exhibits
craniofacial skeleton, enteric ganglia, following NGF infusion. Proc Natl Acad Sci learning and behaviour deficits. Nat Genet
and components of the outflow tract of USA 98:10439–10444. 11:177–183.
Crome R, Cowie V, Slater E. 1966. A statistical Richtsmeier J, Baxter L, Reeves R. 2000. Parallels
the heart. This morphogen is primarily of craniofacial maldevelopment in Down
note on cerebellar and brain-stem weight in
responsible for establishment of pattern- Down syndrome. J Mental Defect Res syndrome and Ts65Dn mice. Dev Dyn 217:
ing anterior–posterior polarity in the 10:69–72. 137–145.
hand. Thus, an attenuated SHH response Delabar JM, Theophile D, Rahmani Z, et al. Richtsmeier JT, Zumwalt A, Carlson EJ, et al.
could contribute to multiple aspects of 1993. Molecular mapping of twenty-four 2002. Craniofacial phenotypes in segmen-
features of Down syndrome on chromosome tally trisomic mouse models for Down syn-
the DS phenotype. However, substantial drome. Am J Med Genet 107:317–324.
21. Eur J Hum Genet 1:114–124.
additional fundamental discovery will be Escorihuela RM, Fernandez-Teruel A, Vallina IF, Roper R, Reeves R. 2006. Understanding the
required to determine whether SHH is et al. 1995. A behavioral assessment of basis for Down syndrome phenotypes. PLoS
beneficial in the longer term and what Ts65Dn mice: a putative Down syndrome Genet 2:231–236.
aspects of development may specifically model. Neurosci Lett 199:143–146. Roper RJ, Baxter LL, Saran NG, et al. 2006. De-
Fernandez F, Morishita W, Zuniga E, et al. 2007. fective cerebellar response to mitogenic
benefit from this type of approach. Pharmacotherapy for cognitive impairment Hedgehog signaling in Down syndrome
in a mouse model of Down syndrome. Nat mice. Proc Natl Acad Sci USA 103:1452–
OVERALL CONCLUSIONS Neurosci 10:411–413. 1456.
The occurrence of three break- Gardiner K, Fortha A, Bechtel L, et al. 2003. Sago H, Carlson EJ, Smith DJ, et al. 1998.
Mouse models of Down syndrome: how Ts1Cje, a partial trisomy 16 mouse model
through studies in 1 year is a harbinger of useful can they be? Comparison of the gene for Down syndrome, exhibits learning and
great progress to come in DS and a call to content of human chromosome 21 with behavioral abnormalities. Proc Natl Acad Sci
the wider scientific community to con- orthologous mouse genomic regions. Gene USA 95:6256–6261.
sider what other areas of research might 318:137–147. Salehi A, Delcroix JD, Belichenko PV, et al. 2006.
benefit from the perspectives provided by Hanson JE, Blank M, Valenzuela RA, et al. 2007. Increased App expression in a mouse model
The functional nature of synaptic circuitry is of Down’s syndrome disrupts NGF transport
defined genetic effects in trisomy. Collec- altered in area CA3 of the hippocampus in a and causes cholinergic neuron degeneration.
tively, these studies represent a synthesis mouse model of Down’s syndrome. J Physiol Neuron 51:29–42.
of approaches based in development, 579 (Part 1):53–67. Siarey RJ, Carlson EJ, Epstein CJ, et al. 1999.
structure, function, and age-related Hattori M, Fujiyama A, Taylor TD, et al. 2000. Increased synaptic depression in the Ts65Dn
changes that must be the basis for any The DNA sequence of human chromosome mouse, a model for mental retardation in
21. Nature 405:311–319. Down syndrome. Neuropharmacology 38:
consideration in this complex disorder. Holtzman DM, Santucci D, Kilbridge J, et al. 1917–1920.
These multi-faceted considerations can 1996. Developmental abnormalities and age- Siarey RJ, Kline-Burgess A, Cho M, et al. 2006.
only be addressed using mouse models related neurodegeneration in a mouse model Altered signaling pathways underlying
that reflect the developmental and physi- of Down syndrome. Proc Natl Acad Sci abnormal hippocampal synaptic plasticity in
ological processes affected in DS, and that USA 93:13333–13338. the Ts65Dn mouse model of Down syn-
Kleschevnikov AM, Belichenko PV, Villar AJ, drome. J Neurochem 98:1266–1277.
provide tools essential for understanding et al. 2004. Hippocampal long-term Siarey RJ, Stoll J, Rapoport SI, et al. 1997.
the genetic basis of the divergence potentiation suppressed by increased inhi- Altered long-term potentiation in the young
between trisomic and euploid indi- bition in the Ts65Dn mouse, a genetic and old Ts65Dn mouse, a model for Down
viduals. A common element of these model of Down syndrome. J Neurosci syndrome. Neuropharmacology 36:1549–
approaches is that all were designed with 24:8153–8160. 1554.
Korenberg JR, Chen XN, Schipper R, et al. Toyoda A, Noguchi H, Taylor TD, et al. 2002.
consideration of the possibilities for phar- 1994. Down syndrome phenotypes: the con- Comparative genomic sequence analysis of
maceutical (or other) amelioration. Com- sequences of chromosomal imbalance. Proc the human chromosome 21 Down syndrome
bined with ongoing clinical trials, e.g., of Natl Acad Sci USA 91:4997–5001. critical region. Genome Res 12:1323–1332.

220 MRDD Research Reviews DOI 10.1002/mrdd  TOWARDS THERAPY FOR DOWN SYNDROME  REEVES

You might also like