Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 1.
Published in final edited form as: Curr Opin Neurobiol. 2012 May 30;22(5):880–886. doi: 10.1016/j.conb.2012.05.002

Cognitive and Pharmacological Insights from the Ts65Dn Mouse Model of Down Syndrome

Aarti Ruparelia a, Matthew L Pearn b,c, William C Mobley d,*
PMCID: PMC3434300  NIHMSID: NIHMS376458  PMID: 22658745

Abstract

Down syndrome (DS) is a multi-faceted condition resulting in the most common genetic form of intellectual disability. Mouse models of DS, especially the Ts65Dn model, have been pivotal in furthering our understanding of the genetic, molecular and neurobiological mechanisms that underlie learning and memory impairments in DS. Cognitive and pharmacological insights from the Ts65Dn mouse model have led to remarkable translational progress in the development of therapeutic targets and in the emergence of DS clinical trials. Unravelling the pathogenic role of trisomic genes on human chromosome 21 and the genotype-phenotype relationship still remains a pertinent goal for tackling cognitive deficits in DS.

Introduction

Trisomy of human chromosome 21 (Hsa21) causes overexpression of more than 500 genes, resulting in the multi-faceted genetic condition characterised as Down syndrome (DS) [1,2]. With an incidence of approximately one in 650-1000 live births worldwide, DS is the most common genetic form of intellectual disability [3]. Accelerated and precocious aging occurs in DS, as does early-onset Alzheimer's disease (AD), which is manifested in over 75% of people with DS by the age of 65 [3-5]. Learning and memory impairments in DS are marked by perturbed neurodevelopment, altered neuronal structure, and synaptic plasticity deficits. The cognitive profiles in DS vary in both expressivity and severity; conceivably from allelic differences in Hsa21 genes and the complex interplay with other non-Hsa21 genes, epigenetic influences and environmental factors. Understanding these genotype-phenotype correlations may help develop pharmacological interventions. Mouse models of DS, including the Ts65Dn mouse, recapitulate many cognitive phenotypes of DS and have been instrumental in elucidating the molecular pathogenesis underlying DS, mapping Hsa21 genes to various phenotypes, and assessing the effect of potential therapeutic targets [6-8]. Herein, we highlight recent insights obtained from the Ts65Dn mouse model to unravel mechanisms of learning and memory impairments in DS; and how these findings have led to recent breakthroughs in pharmacological interventions.

Cognitive insights from the Ts65Dn mouse

Neurodevelopment

Neurodevelopment is perturbed in DS as demonstrated by a reduced brain volume, reduced number of neurons, and abnormal neuronal morphology in several brain regions; particularly the granule cells in the cerebellar cortex [9]. Compared to healthy infants, brains of DS infants show an increase in total dendritic branching and higher total dendritic length, which then steadily decreases to lower than normal levels during adolescence and into adulthood. These structural and dendritic differences may contribute to perturbed cortical information processing and decreased synaptic plasticity [10]. It is proposed that elongation of the cell cycle length, from decreased Sonic hedgehog growth factor response, results in reduced proliferation rates, leading to impaired neurogenesis [9]. A deficient mitotic response to the Sonic hedgehog growth factor in the Ts65Dn mice is proposed to cause the decreased proliferation of the cerebellar granule cells and an alteration in neural crest progenitor cells, which could contribute to the DS-associated craniofacial dysmorphology [11,12]. Cerebellar granule cell deficits in neural progenitor cells as well as an elevated rate of cell death have been documented in other mouse models of DS [13,14]. Oxidative stress levels indicative of elevated rates of neuronal apoptosis are also increased in DS fibroblasts [15,16].

GABAergic system and synaptic plasticity

The majority of the forebrain is comprised of excitatory glutamatergic projection neurons and approximately 10% inhibitory γ-aminobutyric acid (GABA) interneurons. Neuronal development and cognitive functioning is dependent on a balanced ratio of excitatory and inhibitory neurons. A developed and functioning cortex evolves from the neurogenesis of the proper neurotransmission of excitatory and inhibitory neurons, in distinct sites of origin, followed by the migration and differentiation of these neurons within the neocortex [17-19]. Alterations in neuronal morphology, function, and neurotransmission have been proposed to cause synaptic plasticity deficits and impairments in long-term potentiation (LTP), a neural correlate for learning and memory.

Neurophysiological studies in the Ts65Dn mouse have revealed enlarged boutons and dendritic spine heads in cortical and hippocampal neurons and excessive inhibition leading to failed LTP induction in the hippocampus and fascia dentate [20-22]. This increased inhibitory input has been attributed to an altered efficiency of the GABAergic system in the DG of Ts65Dn mice, rather than a decrease in inhibitory synapse density, and is a proposed mechanism for synaptic plasticity defects in DS [21-26]. Electrophysiological data revealed enhanced GABAA and GABAB receptor-mediated neurotransmission with an accompanied reduction of paired-pulse ratios of evoked inhibitory postsynaptic currents (IPSCs); suggesting increased presynaptic release of GABA. These data correlate with larger, but not increased, number of inhibitory synapses found in the DG of Ts65Dn mice.

Contribution of Hsa21-encoded genes

The perturbed neurodevelopment and the over-inhibition in DS and Ts65Dn mice is likely caused by triplicated genes on Hsa21 (Table 1). Oligodendrocyte transcription factor 1 (Olig1) and lineage transcription factor 2 (Olig2) genes are implicated in neurogenesis and oligodendrogenesis [27,28]. Normalising these two genes to disomic levels in Ts65Dn mice corrected the enhanced inhibitory interneuron phenotype, providing a causal explanation of the gene-dosage imbalance of Olig1 and Olig2 genes in producing the excitatory-inhibitory (E-I) imbalance [29].

Table 1.

Physiological and pathogenic role of affected Hsa21 genes

Hsa21 genes Physiological role Pathogenic role Reference
OLIG1/OLIG2 Implicated in neurogenesis and oligodendrogenesis Causes GABAergic excitatory-inhibitory imbalance Lu et al., 2002; Zhou & Anderson, 2002; Chakrabarti et al., 2010
KCNJ6 Effector protein for GABAB receptors; modulates potassium channel current and density Contributes to enhanced GABAB signalling Best et al., 2007; Best et al., 2011; Kleschevnikov et al., 2012
SYNJ1 Nerve terminal protein implicated in membrane trafficking and synaptic transmission Inability to maintain stable GABAergic neurotransmission Luthi et al., 2001
DYRK1A Regulates signalling and cell proliferation; involved in neurogenesis and neurodevelopment Deregulates genes implicated in dendritic growth, cell pluripotency and embryonic stem cell fate; deregulates NFAT circuits and may cause enhanced Tau phosphorylation Altafaj et al., 2001; Lepagnol-Bestel et al., 2009; Canzonetta et al., 2008; Arron et al., 2006; Jung et al., 2011
RCAN1 Inhibits calcineurin-dependent signalling pathways affecting development; modulates NMDAR activation Decreases opening probability of NMDAR channel; deregulates NFAT circuits and may cause enhanced Tau phosphorylation Arron et al., 2006; Jung et al., 2011
DSCAM Cell adhesion molecule with a critical role in dendrite morphology and neuronal wiring Inhibits dendritic branching and causes aberrant synaptic plasticity; aberrant NMDA-mediated regulation of DSCAM local translation Alves-Sampaio et al., 2010
SIM2 Transcriptional repressor implicated in synaptic plasticity and morphology Reduces DBN1 levels causing morphological cytoskeletal changes at postsynaptic terminals in dendritic spines Ooe et al., 2004; Hayashi et al., 1996
SOD1 Cytoplasmic protein implicated in oxidative stress Reduces hippocampal neuronal progenitors and LTP; enhances sensitivity to degeneration and apoptosis; up-regulates GABAergic neurotransmission Gahtan et al., 1998; Levkovitz et al., 1999
APP Cell surface receptor and transmembrane precursor protein that promotes transcriptional activation Forms the protein basis of amyloid plaques prevalent in AD and DS; mechanism of action unknown Rovelet-Lecrux et al., 2006; Netzer et al., 2010

Enhanced postsynaptic GABAB signalling could be explained by the triplication of the KCNJ6 (potassium inwardly-rectifying channel, subfamily J, member 6) gene and increased expression of the protein it encodes, Kir3.2, a channel that modulates postsynaptic GABAB receptors. Overexpression of Kcnj6 in Ts65Dn mice leads to increased Kir3.2 channel density, increased current, and increased inhibitory GABAB signalling [30]. A recent study also documented enhanced GABAB/Kir3.2 signalling in DG granule cells of Ts65Dn mice [25]. Kcnj6 overexpression has also been suggested to lead to an imbalance between GABAB and GABAA inhibition of CA1 pyramidal neurons through a pathway specific mechanism to perturb hippocampal circuitry functioning [31]. Synaptojanin 1 (SYNJ1) encodes a nerve terminal protein that is implicated in membrane trafficking and is another Hsa21 gene that is essential for maintaining GABAergic neurotransmission stability [32].

DYRK1A (dual specificity tyrosine-(Y)-phosphorylation-regulated kinase 1A) is heavily implicated in neurodevelopment and is strongly expressed in neural precursor populations during embryonic neurogenesis. It is conceivable that altered DYRK1A expression levels perturb developmental pathways, leading to postnatal neurodevelopmental difficulties [33]. Overexpression of DYRK1A has been found to decrease neuron-restrictive silencer factor (REST/NRSF) chromatin remodelling complex levels and to deregulate genes that contribute to DS-associated neuronal phenotypes, including dendritic growth impairments, pluripotency, and embryonic stem cell fate [34,35]. RCAN1 (regulator of calcineurin 1) is a negative regulator of calcineurin that subsequently modulates NMethyl-D-aspartate receptor (NMDAR) activation kinetics by decreasing the probability of opening time of the NMDAR channel. DYRK1A directly phosphorylates RCAN1, leading to reduced nuclear factor of activated T-cells (NFATc) translocation to the nucleus. NFATc transcription factors are regulators of vertebrate development and destabilisation of this regulatory circuit through triplication of DYRK1A and RCAN1 may contribute to the enhanced Tau phosphorylation seen in DS [36,37].

DSCAM (Down syndrome cell adhesion molecule) has a critical role in dendrite morphology and neuronal wiring. Overexpression of DSCAM in hippocampal neurons inhibits dendritic branching; impairments in NMDA-mediated regulation of DSCAM local mRNA translation may be one mechanism through which aberrant dendritic morphology and synaptic plasticity deficits occurs during development [38]. Overexpression of SIM2 (single-minded homolog 2), a transcriptional repressor, dramatically reduces levels of DBN1 (Drebrin 1), a neuronal gene that modulates dendritic spine cytoskeletal dynamics at postsynaptic terminals. The reduction of DBN1 levels could explain the morphological neuronal changes and the resulting learning and memory deficits prevalent in DS [39,40].

Sod1 (superoxide dismutase 1) overexpression also reduces hippocampal neuronal progenitors and LTP, enhances sensitivity to degeneration and apoptosis, and up-regulates GABAergic neurotransmission [41,42]. Amyloid protein (APP) is strongly implicated in neurodegeneration, and triplication of this gene has been associated with early onset AD [43]. However, recent evidence for a role in neurodevelopment for APP stems from studies in which lowering of beta-amyloid levels, an APP metabolite that is the main constitute of amyloid plaques in AD, improved cognitive deficits in Ts65Dn mice [44].

Pharmacological insights from the Ts65Dn mouse

The identification of behavioural, morphological, and neurobiological alterations in the Ts65Dn mouse model have led to invaluable insights into the pathogenesis of DS that allow for potential therapeutic targets to be explored (Table 2).

Table 2.

Pharmacological interventions to tackle DS-associated cognitive deficits

Neurobiological Pathway Pharmacological Compound Cognitive Effect References
SSRIs and mood stabilisers Fluoxetine Increases neurogenesis by enhancing proliferation and survival of neurons; restores 5-HT receptor and BDNF levels Clark et al., 2006; Bianchi et al., 2010
Lithium Increases neurogenesis by restoring cell proliferation in subventricular zone Bianchi et al., 2010
Neuroprotective peptides NAPVSIPQ+SALLRSIPA Protection from oxidative damage and apoptosis; prenatal treatment prevents developmental delays, glial deficits and aberrant ADNF expression Brenneman et al., 2004; Busciglio et al., 2007; Toso et al., 2008
GABAA antagonists Pentylenetetrazol Improves hippocampal-based learning in behavioural tasks and enhances LTP Fernandez et al., 2007; Rueda et al., 2008
Picrotoxin Improves hippocampal-based learning in behavioural tasks, enhances LTP and normalises NMDAR-mediated currents Kleschevnikov et al., 2004; Fernandez et al., 2007
α5IA Improves hippocampal-based learning in behavioural tasks, normalises Sod1 overexpression and enhances learning-evoked immediate gene expression levels Braudeau et al., 2001; Roche, 2011
NMDAR antagonists Memantine Improves hippocampal-based learning and fear conditioning in behavioural tasks, reduces excessive glutamate neurotransmission and increases BDNF levels Costa et al., 206; Rueda et al., 2010; Lockrow et al., 2010; Mohan et al., 2009

SSRIs and mood stabilisers

Chronic treatment in Ts65Dn mice with fluoxetine, a serotonin selective reuptake inhibitor (SSRI), increased neurogenesis by enhancing the proliferation and survival of neurons in the DG [45]. Recently, studies examined whether early pharmacotherapy with fluoxetine could improve neurogenesis. Untreated Ts65Dn neonatal mice exhibited impaired cellular proliferation and demonstrated normal levels of serotonin (5-HT), but a lower expression of 5-HT1A receptors and brain-derived neurotrophic factor (BDNF) levels [46]. Treating Ts65Dn neonatal mice with fluoxetine not only rescued impaired proliferation and increased the number of surviving cells, but also restored the expression of 5-HT receptors and BDNF levels to that of control mice [46]. Lithium has also been examined as a potential treatment to improve neurogenesis. Treating Ts65Dn mice with lithium restored cellular proliferation in the subventricular zone [47]. These studies demonstrate the potential of early pharmacotherapy to correct for neurogenesis impairments by using readily available and approved drugs.

Neuroprotective peptides

Pharmacological intervention with neuroprotective peptides has also been demonstrated to promote neurodevelopment. Vasoactive intestinal peptide (VIP) levels are altered in DS; and cortical astrocytes in Ts65Dn neonatal mice demonstrate reduced responsiveness to VIP stimulation [48,49]. Activity-dependent neuroprotective protein (ADNP) and activity-dependent neurotrophic factor (ADNF) are neuroprotective neurotrophic factors released by VIP stimulation of astrocytes [50]. Combined treatment of DS cortical neurons with active fragments of ADNP and ADNF, NAPVSIPQ (NAP) and SALLRSIPA (SAL) respectively, increased neuronal survival, restored morphological changes and protected from oxidative damage and apoptosis [51]. The efficacy of these neuroprotective peptides in preventing developmental delay and glial deficits through prenatal treatment was examined in Ts65Dn mice. Untreated Ts65Dn mice displayed developmental delays in achieving motor and sensory milestones, downregulated ADNF expression, and glial deficits [52]. Prenatal treatment with NAP+SAL reversed all these deficits [52]. This study identifies a potential intervention during pregnancy that could improve developmental delays and glial deficits in DS.

GABAA antagonists

To restore the E-I imbalance, several pharmacological interventions have aimed to decrease the excessive inhibition of GABAergic neurotransmission prevalent in Ts65Dn mice [25,26]. Ts65Dn mice have been treated with non-competitive GABAA antagonists, pentylenetetrazol (PTZ) and picrotoxin (PTX), which inhibit GABAA receptors. Chronic treatment with PTZ reversed the deficits seen in the novel object recognition task (NORT) and spontaneous alternation tasks in Ts65Dn mice [53]. Surprisingly, the improvement in cognition and LTP was sustained for up to 2 months after initial treatment, suggesting a lasting effect of treatment on neuronal circuit modification. Chronic treatment with PTZ for 8 weeks in Ts65Dn mice did not modify sensorimotor abilities and locomotor activity in home cages; however it did rescue learning and memory performance in the Morris water maze (MWM) task [54]. Treating Ts65Dn mice with PTX also reversed deficits in NORT that were exhibited in untreated mice; these improvements were retained for up to 2 weeks [53]. In untreated Ts65Dn mice, impaired LTP was coupled with reduced synaptic activation of NMDAR due to excessive inhibition of DG cells [21]. Administering PTX to suppress inhibition resulted in improved induction of LTP and normalised NMDAR-mediated currents [21]. Recently, chronic treatment in Ts65Dn mice with an inverse agonist selective for the α5 subunit of the GABAA benzodiazepine receptor (α5IA) improved cognitive deficits in the MWM and normalised Sod1 overexpression with an enhancement in learning-evoked immediate early genes expression levels [55]. Encouraged by this body of evidence, Roche, a healthcare company, recently announced the commencement of a trial to examine the cognitive impact of reducing GABAergic neurotransmission in the hippocampus using a drug selective for the α5 subunit of GABAA receptors [56].

NMDAR antagonists

Learning is also improved by the non-competitive NMDAR antagonist, memantine, which reduces abnormal activation of glutamate neurotransmission. Administration of memantine, an open-channel antagonist, rescued Ts65Dn performance deficits in a fear conditioning test [57] and improved spatial learning in MWM task [58]. Long-term memantine treatment improved spatial reference memory in a MWM task and recovered object discrimination ability in a NORT, but spontaneous activity remained unaltered [59]. Upon histopathological analysis, no morphological modifications indicative of neuroprotection were observed in the neurons of the basal forebrain or locus coeruleus (LC), however, an increase in BDNF expression was documented in the hippocampus and frontal cortex [59]. Interestingly, acute treatment of memantine 30 mins prior to testing was sufficient to enhance performance on the NORT [59]. Despite mouse studies demonstrating promising benefits of memantine, a recently published clinical trial reported that memantine is not an effective pharmacological treatment for cognitive decline or dementia in people who are above 40 years old and have DS [60]. This suggests that therapies that are effective in people with AD may not necessarily confer benefits in DS.

Conclusion

Triplication of Hsa21 genes leads to a plethora of multi-system pathologies that characterise DS, rendering it complex to understand. Despite this, since the discovery of DS in the 19th century, the life expectancy of people with DS has increased from an average age of 12 years old in the 1940s to 60 years of age at present due to dramatic advances in medical treatment and social intervention [3]. Mouse models of DS, especially the Ts65Dn mouse, have provided an unequivocal contribution to dissecting the genetic, molecular and neurobiological processes that underlie the syndrome, and in deciphering the genotype-phenotype relationship of overexpressed Hsa21 genes in causing the clinical manifestation of DS. This approach has successfully led to the development of pharmacological targets and the emergence of DS clinical trials. However, to fully understand the genetic basis of DS and its consequent perturbations still remains a challenge and further investigations are necessary to tackle various aspects of the syndrome. The development and study of DS mouse models that more closely resemble the gene-dosage imbalance in humans with DS, and genome-wide association studies of individuals with DS, will be instrumental in identifying dosage-sensitive genes and the pathogenic mechanisms underlying DS-associated phenotypes.

Highlights.

DS causes perturbed synaptic plasticity and excessive inhibitory neurotransmission

Ts65Dn mouse model recapitulates behavioural and cognitive phenotypes of DS

Several triplicated Hsa21-associated genes in Ts65Dn mice are implicated

Insights from Ts65Dn have led to pharmacological interventions and clinical trials

Acknowledgements

We thank the Brain Research Trust for funding (A.R.). We also thank NIH (PN2 EY016525; R01 NS066072-01A1; R01 NS055371; R01 NS24054), Down Syndrome Research and Treatment Foundation, Alzhiemer's Association, Thrasher Research Fund and the Larry L. Hillblom Foundation for funding (W.C.M.).

Grant Sponsors: NIH (NS06672, NS24054, PN2EY016525), Down Syndrome Research and Treatment Foundation, Alzhiemer's Association, Thrasher Research Fund, and the Larry L. Hillblom Foundation.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of Interest

The authors declare no conflicts of interest.

References

  • 1.Ahmed MM, Sturgeon X, Ellison M, Davisson MT, Gardiner KJ. Loss of correlations among proteins in brains of the Ts65Dn mouse model of down syndrome. J Proteome Res. 2012;11:1251–1263. doi: 10.1021/pr2011582. [DOI] [PubMed] [Google Scholar]
  • 2.Hattori M, Fujiyama A, Taylor TD, Watanabe H, Yada T, Park HS, Toyoda A, Ishii K, Totoki Y, Choi DK, et al. The DNA sequence of human chromosome 21. Nature. 2000;405:311–319. doi: 10.1038/35012518. [DOI] [PubMed] [Google Scholar]
  • 3.Bittles AH, Bower C, Hussain R, Glasson EJ. The four ages of Down syndrome. The European Journal of Public Health. 2007;17:221–225. doi: 10.1093/eurpub/ckl103. [DOI] [PubMed] [Google Scholar]
  • 4.Coppus A, Evenhuis H, Verberne GJ, Visser F, van Gool P, Eikelenboom P, van Duijin C. Dementia and mortality in persons with Down's syndrome. J Intellect Disabil Res. 2006;50:768–777. doi: 10.1111/j.1365-2788.2006.00842.x. [DOI] [PubMed] [Google Scholar]
  • 5*.Beacher F, Daly E, Simmons A, Prasher V, Morris R, Robinson C, Lovestone S, Murphy K, Murphy DG. Brain anatomy and ageing in non-demented adults with Down's syndrome: an in vivo MRI study. Psychol Med. 2010;40:611–619. doi: 10.1017/S0033291709990985. [Using volumetric MRI the authors discovered neuronal correlates for ‘accelerated’ ageing in DS. Individuals with DS displayed reduced frontal, temporal and parietal lobes, and increased peripheral CSF. This study demonstrates a biological dissociation of ageing from DS pathology and the compounded risk for age-related cognitive decline and AD in DS.] [DOI] [PubMed] [Google Scholar]
  • 6.Reeves RH, Irving NG, Moran TH, Wohn A, Kitt C, Sisodia SS, Schmidt C, Bronson RT, Davisson MT. A mouse model for Down syndrome exhibits learning and behaviour deficits. Nat Genet. 1995;11:177–184. doi: 10.1038/ng1095-177. [DOI] [PubMed] [Google Scholar]
  • 7.Davisson MT, Schmidt C, Reeves RH, Irving NG, Akeson EC, Harris BS, Bronson RT. Segmental trisomy as a mouse model for Down syndrome. Prog Clin Biol Res. 1993;384:117–133. [PubMed] [Google Scholar]
  • 8.Ruparelia A, Wiseman F, Sheppard O, Tybulewicz VLJ, Fisher EMC. Down syndrome and the molecular pathogenesis resulting from trisomy of human chromosome 21. Journal of Biomedical Research. 2010;24:87–99. doi: 10.1016/S1674-8301(10)60016-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Contestabile A, Fila T, Bartesaghi R, Ciani E. Cell cycle elongation impairs proliferation of cerebellar granule cell precursors in the Ts65Dn mouse, an animal model for Down syndrome. Brain Pathol. 2009;19:224–237. doi: 10.1111/j.1750-3639.2008.00168.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Lott IT, Dierssen M. Cognitive deficits and associated neurological complications in individuals with Down's syndrome. Lancet Neurol. 2010;9:623–633. doi: 10.1016/S1474-4422(10)70112-5. [DOI] [PubMed] [Google Scholar]
  • 11.Roper RJ, Baxter LL, Saran NG, Klinedinst DK, Beachy PA, Reeves RH. Defective cerebellar response to mitogenic Hedgehog signaling in Down [corrected] syndrome mice. Proc Natl Acad Sci U S A. 2006;103:1452–1456. doi: 10.1073/pnas.0510750103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Roper RJ, VanHorn JF, Cain CC, Reeves RH. A neural crest deficit in Down syndrome mice is associated with deficient mitotic response to Sonic hedgehog. Mech Dev. 2009;126:212–219. doi: 10.1016/j.mod.2008.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Moldrich RX, Dauphinot L, Laffaire J, Vitalis T, Herault Y, Beart PM, Rossier J, Vivien D, Gehrig C, Antonarakis SE, et al. Proliferation deficits and gene expression dysregulation in Down's syndrome (Ts1Cje) neural progenitor cells cultured from neurospheres. J Neurosci Res. 2009;87:3143–3152. doi: 10.1002/jnr.22131. [DOI] [PubMed] [Google Scholar]
  • 14.O'Doherty A, Ruf S, Mulligan C, Hildreth V, Errington ML, Cooke S, Sesay A, Modino S, Vanes L, Hernandez D, et al. An aneuploid mouse strain carrying human chromosome 21 with Down syndrome phenotypes. Science. 2005;309:2033–2037. doi: 10.1126/science.1114535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Micali N, Longobardi E, Iotti G, Ferrai C, Castagnaro L, Ricciardi M, Blasi F, Crippa MP. Down syndrome fibroblasts and mouse Prep1-overexpressing cells display increased sensitivity to genotoxic stress. Nucleic Acids Res. 2010;38:3595–3604. doi: 10.1093/nar/gkq019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Busciglio J, Yankner BA. Apoptosis and increased generation of reactive oxygen species in Down's syndrome neurons in vitro. Nature. 1995;378:776–779. doi: 10.1038/378776a0. [DOI] [PubMed] [Google Scholar]
  • 17.Ang ES, Jr., Haydar TF, Gluncic V, Rakic P. Four-dimensional migratory coordinates of GABAergic interneurons in the developing mouse cortex. J Neurosci. 2003;23:5805–5815. doi: 10.1523/JNEUROSCI.23-13-05805.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Corbin JG, Nery S, Fishell G. Telencephalic cells take a tangent: non-radial migration in the mammalian forebrain. Nat Neurosci. 2001;4(Suppl):1177–1182. doi: 10.1038/nn749. [DOI] [PubMed] [Google Scholar]
  • 19.Haydar TF, Reeves RH. Trisomy 21 and early brain development. Trends Neurosci. 2011 doi: 10.1016/j.tins.2011.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Belichenko PV, Masliah E, Kleschevnikov AM, Villar AJ, Epstein CJ, Salehi A, Mobley WC. Synaptic structural abnormalities in the Ts65Dn mouse model of Down Syndrome. J Comp Neurol. 2004;480:281–298. doi: 10.1002/cne.20337. [DOI] [PubMed] [Google Scholar]
  • 21.Kleschevnikov AM, Belichenko PV, Villar AJ, Epstein CJ, Malenka RC, Mobley WC. Hippocampal long-term potentiation suppressed by increased inhibition in the Ts65Dn mouse, a genetic model of Down syndrome. J Neurosci. 2004;24:8153–8160. doi: 10.1523/JNEUROSCI.1766-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Belichenko PV, Kleschevnikov AM, Masliah E, Wu C, Takimoto-Kimura R, Salehi A, Mobley WC. Excitatory-inhibitory relationship in the fascia dentata in the Ts65Dn mouse model of Down syndrome. J Comp Neurol. 2009;512:453–466. doi: 10.1002/cne.21895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Belichenko PV, Kleschevnikov AM, Salehi A, Epstein CJ, Mobley WC. Synaptic and cognitive abnormalities in mouse models of Down syndrome: exploring genotype-phenotype relationships. J Comp Neurol. 2007;504:329–345. doi: 10.1002/cne.21433. [DOI] [PubMed] [Google Scholar]
  • 24.Belichenko NP, Belichenko PV, Kleschevnikov AM, Salehi A, Reeves RH, Mobley WC. The “Down syndrome critical region” is sufficient in the mouse model to confer behavioral, neurophysiological, and synaptic phenotypes characteristic of Down syndrome. J Neurosci. 2009;29:5938–5948. doi: 10.1523/JNEUROSCI.1547-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25*.Kleschevnikov AM, Belichenko PV, Gall J, George L, Nosheny R, Maloney MT, Salehi A, Mobley WC. Increased efficiency of the GABAA and GABAB receptor-mediated neurotransmission in the Ts65Dn mouse model of Down syndrome. Neurobiol Dis. 2012;45:683–691. doi: 10.1016/j.nbd.2011.10.009. [Enhanced inhibition was characterised by examining synaptic and molecular properties of the GABAergic system in Ts65Dn mice. GABAA receptor signaling was increased, suggesting increased presynaptic release of GABA and increased Kir3.2 levels explained the enhanced GABAB receptor signaling. This study identified the mechanistic basis of inhibitory efficiency, which was unknown.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Rissman RA, Mobley WC. Implications for treatment: GABAA receptors in aging, Down syndrome and Alzheimer's disease. J Neurochem. 2011;117:613–622. doi: 10.1111/j.1471-4159.2011.07237.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Lu QR, Sun T, Zhu Z, Ma N, Garcia M, Stiles CD, Rowitch DH. Common developmental requirement for Olig function indicates a motor neuron/oligodendrocyte connection. Cell. 2002;109:75–86. doi: 10.1016/s0092-8674(02)00678-5. [DOI] [PubMed] [Google Scholar]
  • 28.Zhou Q, Anderson DJ. The bHLH transcription factors OLIG2 and OLIG1 couple neuronal and glial subtype specification. Cell. 2002;109:61–73. doi: 10.1016/s0092-8674(02)00677-3. [DOI] [PubMed] [Google Scholar]
  • 29**.Chakrabarti L, Best TK, Cramer NP, Carney RS, Isaac JT, Galdzicki Z, Haydar TF. Olig1 and Olig2 triplication causes developmental brain defects in Down syndrome. Nat Neurosci. 2010;13:927–934. doi: 10.1038/nn.2600. [Direct gene-dosage relationship found between Olig1 and Olig2 triplication and defects in neurogenesis and neuronal development, leading to an excessive inhibition phenotype in DS. Genetic rescue through normalisation of these genes to two copies corrected the interneuron phenotype. This genetic approach of deleting just one allele of the genes is a gold standard for future work on DS to assess genotype-phenotype relationships.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Best TK, Siarey RJ, Galdzicki Z. Ts65Dn, a mouse model of Down syndrome, exhibits increased GABAB-induced potassium current. J Neurophysiol. 2007;97:892–900. doi: 10.1152/jn.00626.2006. [DOI] [PubMed] [Google Scholar]
  • 31.Best TK, Cramer NP, Chakrabarti L, Haydar TF, Galdzicki Z. Dysfunctional hippocampal inhibition in the Ts65Dn mouse model of Down syndrome. Exp Neurol. 2011 doi: 10.1016/j.expneurol.2011.11.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Luthi A, Di Paolo G, Cremona O, Daniell L, De Camilli P, McCormick DA. Synaptojanin 1 contributes to maintaining the stability of GABAergic transmission in primary cultures of cortical neurons. J Neurosci. 2001;21:9101–9111. doi: 10.1523/JNEUROSCI.21-23-09101.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Altafaj X, Dierssen M, Baamonde C, Marti E, Visa J, Guimera J, Oset M, Gonzalez JR, Florez J, Fillat C, et al. Neurodevelopmental delay, motor abnormalities and cognitive deficits in transgenic mice overexpressing Dyrk1A (minibrain), a murine model of Down's syndrome. Hum Mol Genet. 2001;10:1915–1923. doi: 10.1093/hmg/10.18.1915. [DOI] [PubMed] [Google Scholar]
  • 34.Lepagnol-Bestel AM, Zvara A, Maussion G, Quignon F, Ngimbous B, Ramoz N, Imbeaud S, Loe-Mie Y, Benihoud K, Agier N, et al. DYRK1A interacts with the REST/NRSF-SWI/SNF chromatin remodelling complex to deregulate gene clusters involved in the neuronal phenotypic traits of Down syndrome. Hum Mol Genet. 2009;18:1405–1414. doi: 10.1093/hmg/ddp047. [DOI] [PubMed] [Google Scholar]
  • 35.Canzonetta C, Mulligan C, Deutsch S, Ruf S, O'Doherty A, Lyle R, Borel C, Lin-Marq N, Delom F, Groet J, et al. DYRK1A-dosage imbalance perturbs NRSF/REST levels, deregulating pluripotency and embryonic stem cell fate in Down syndrome. Am J Hum Genet. 2008;83:388–400. doi: 10.1016/j.ajhg.2008.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Arron JR, Winslow MM, Polleri A, Chang CP, Wu H, Gao X, Neilson JR, Chen L, Heit JJ, Kim SK, et al. NFAT dysregulation by increased dosage of DSCR1 and DYRK1A on chromosome 21. Nature. 2006;441:595–600. doi: 10.1038/nature04678. [DOI] [PubMed] [Google Scholar]
  • 37**.Jung MS, Park JH, Ryu YS, Choi SH, Yoon SH, Kwen MY, Oh JY, Song WJ, Chung SH. Regulation of RCAN1 protein activity by Dyrk1A protein-mediated phosphorylation. J Biol Chem. 2011;286:40401–40412. doi: 10.1074/jbc.M111.253971. [Direct synergistic link between Dyrk1A and RCAN1 was found such that Dyrk1A-mediated phosphorylation of RCAN1 enhanced the ability of RCAN1 to inhibit the phosphotase activity of calcineurin and extended its half-life, resulting in reduced NFAT transcriptional activity and enhanced Tau phosphorylation. This study furthers our understanding of the interaction between Hsa21 genes and their contribution to DS phenotypes.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38*.Alves-Sampaio A, Troca-Marin JA, Montesinos ML. NMDA-mediated regulation of DSCAM dendritic local translation is lost in a mouse model of Down's syndrome. J Neurosci. 2010;30:13537–13548. doi: 10.1523/JNEUROSCI.3457-10.2010. [In the Ts1Cje mouse model, overexpression of DSCAM resulted in increased levels of DSCAM mRNA and protein and was found to inhibit dendritic branching. The authors propose that a loss of NMDA-mediated regulation of DSCAM dendritic local translation contributes to dendritic morphology and neurogenesis defects in development and synaptic plasticity deficits in adulthood.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ooe N, Saito K, Mikami N, Nakatuka I, Kaneko H. Identification of a novel basic helix-loop-helix-PAS factor, NXF, reveals a Sim2 competitive, positive regulatory role in dendritic-cytoskeleton modulator drebrin gene expression. Mol Cell Biol. 2004;24:608–616. doi: 10.1128/MCB.24.2.608-616.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Hayashi K, Ishikawa R, Ye LH, He XL, Takata K, Kohama K, Shirao T. Modulatory role of drebrin on the cytoskeleton within dendritic spines in the rat cerebral cortex. J Neurosci. 1996;16:7161–7170. doi: 10.1523/JNEUROSCI.16-22-07161.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Gahtan E, Auerbach JM, Groner Y, Segal M. Reversible impairment of long-term potentiation in transgenic Cu/Zn-SOD mice. Eur J Neurosci. 1998;10:538–544. doi: 10.1046/j.1460-9568.1998.00058.x. [DOI] [PubMed] [Google Scholar]
  • 42.Levkovitz Y, Avignone E, Groner Y, Segal M. Upregulation of GABA neurotransmission suppresses hippocampal excitability and prevents long-term potentiation in transgenic superoxide dismutase-overexpressing mice. J Neurosci. 1999;19:10977–10984. doi: 10.1523/JNEUROSCI.19-24-10977.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Rovelet-Lecrux A, Hannequin D, Raux G, Le Meur N, Laquerriere A, Vital A, Dumanchin C, Feuillette S, Brice A, Vercelletto M, et al. APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy. Nat Genet. 2006;38:24–26. doi: 10.1038/ng1718. [DOI] [PubMed] [Google Scholar]
  • 44.Netzer WJ, Powell C, Nong Y, Blundell J, Wong L, Duff K, Flajolet M, Greengard P. Lowering beta-amyloid levels rescues learning and memory in a Down syndrome mouse model. PLoS One. 2010;5:e10943. doi: 10.1371/journal.pone.0010943. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Clark S, Schwalbe J, Stasko MR, Yarowsky PJ, Costa AC. Fluoxetine rescues deficient neurogenesis in hippocampus of the Ts65Dn mouse model for Down syndrome. Exp Neurol. 2006;200:256–261. doi: 10.1016/j.expneurol.2006.02.005. [DOI] [PubMed] [Google Scholar]
  • 46*.Bianchi P, Ciani E, Guidi S, Trazzi S, Felice D, Grossi G, Fernandez M, Giuliani A, Calza L, Bartesaghi R. Early pharmacotherapy restores neurogenesis and cognitive performance in the Ts65Dn mouse model for Down syndrome. J Neurosci. 2010;30:8769–8779. doi: 10.1523/JNEUROSCI.0534-10.2010. [This study demonstrates that early pharmacotherapy in neonatal Ts65Dn mice with a readily available antidepressant, fluoxetine, reverses defective proliferation and promotes neurogenesis as well as improving cognitive deficits in a hippocampal-based contextual fear conditioning task. This has crucial implications for targeting treatments during early development and the use of SSRIs in improving neurogenesis deficits.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Bianchi P, Ciani E, Contestabile A, Guidi S, Bartesaghi R. Lithium restores neurogenesis in the subventricular zone of the Ts65Dn mouse, a model for Down syndrome. Brain Pathol. 2010;20:106–118. doi: 10.1111/j.1750-3639.2008.00246.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Nelson PG, Kuddo T, Song EY, Dambrosia JM, Kohler S, Satyanarayana G, Vandunk C, Grether JK, Nelson KB. Selected neurotrophins, neuropeptides, and cytokines: developmental trajectory and concentrations in neonatal blood of children with autism or Down syndrome. Int J Dev Neurosci. 2006;24:73–80. doi: 10.1016/j.ijdevneu.2005.10.003. [DOI] [PubMed] [Google Scholar]
  • 49.Sahir N, Brenneman DE, Hill JM. Neonatal mice of the Down syndrome model, Ts65Dn, exhibit upregulated VIP measures and reduced responsiveness of cortical astrocytes to VIP stimulation. J Mol Neurosci. 2006;30:329–340. doi: 10.1385/JMN:30:3:329. [DOI] [PubMed] [Google Scholar]
  • 50.Brenneman DE, Spong CY, Hauser JM, Abebe D, Pinhasov A, Golian T, Gozes I. Protective peptides that are orally active and mechanistically nonchiral. J Pharmacol Exp Ther. 2004;309:1190–1197. doi: 10.1124/jpet.103.063891. [DOI] [PubMed] [Google Scholar]
  • 51.Busciglio J, Pelsman A, Helguera P, Ashur-Fabian O, Pinhasov A, Brenneman DE, Gozes I. NAP and ADNF-9 protect normal and Down's syndrome cortical neurons from oxidative damage and apoptosis. Curr Pharm Des. 2007;13:1091–1098. doi: 10.2174/138161207780618957. [DOI] [PubMed] [Google Scholar]
  • 52.Toso L, Cameroni I, Roberson R, Abebe D, Bissell S, Spong CY. Prevention of developmental delays in a Down syndrome mouse model. Obstet Gynecol. 2008;112:1242–1251. doi: 10.1097/AOG.0b013e31818c91dc. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Fernandez F, Morishita W, Zuniga E, Nguyen J, Blank M, Malenka RC, Garner CC. Pharmacotherapy for cognitive impairment in a mouse model of Down syndrome. Nat Neurosci. 2007;10:411–413. doi: 10.1038/nn1860. [DOI] [PubMed] [Google Scholar]
  • 54.Rueda N, Florez J, Martinez-Cue C. Chronic pentylenetetrazole but not donepezil treatment rescues spatial cognition in Ts65Dn mice, a model for Down syndrome. Neurosci Lett. 2008;433:22–27. doi: 10.1016/j.neulet.2007.12.039. [DOI] [PubMed] [Google Scholar]
  • 55**.Braudeau J, Dauphinot L, Duchon A, Loistron A, Dodd RH, Herault Y, Delatour B, Potier MC. Chronic Treatment with a Promnesiant GABA-A alpha5-Selective Inverse Agonist Increases Immediate Early Genes Expression during Memory Processing in Mice and Rectifies Their Expression Levels in a Down Syndrome Mouse Model. Adv Pharmacol Sci. 2011;2011:153218. doi: 10.1155/2011/153218. [Chronic treatment in Ts65Dn mice with a GABAA α5 subunit inverse agonist restores cognitive deficits, normalises the expression level of Sod1 and rescues deficits in immediate early gene activation, particularly of c-Fos and Arc. This study has been pivotal in the development of clinical trials using this compound to reverse DS-associated cognitive deficits.] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Roche: Roche starts early stage clinical trial in Down syndrome. 2011 http://www.rocheusa.com/portal/usa/press_releases_nutley?siteUuid=re7180004&paf_gear_id=38400020&pageId=re7425113&synergyaction=show&paf_dm=full&nodeId=1415-fbfa4d37db2611e0953b3d6bec9c2782&currentPage=0.
  • 57.Costa AC, Scott-McKean JJ, Stasko MR. Acute injections of the NMDA receptor antagonist memantine rescue performance deficits of the Ts65Dn mouse model of Down syndrome on a fear conditioning test. Neuropsychopharmacology. 2008;33:1624–1632. doi: 10.1038/sj.npp.1301535. [DOI] [PubMed] [Google Scholar]
  • 58.Rueda N, Llorens-Martin M, Florez J, Valdizan E, Banerjee P, Trejo JL, Martinez-Cue C. Memantine normalizes several phenotypic features in the Ts65Dn mouse model of Down syndrome. J Alzheimers Dis. 2010;21:277–290. doi: 10.3233/JAD-2010-100240. [DOI] [PubMed] [Google Scholar]
  • 59.Lockrow J, Boger H, Bimonte-Nelson H, Granholm AC. Effects of long-term memantine on memory and neuropathology in Ts65Dn mice, a model for Down syndrome. Behav Brain Res. 2011;221:610–622. doi: 10.1016/j.bbr.2010.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hanney M, Prasher V, Williams N, Jones EL, Aarsland D, Corbett A, Lawrence D, Yu LM, Tyrer S, Francis PT, et al. Memantine for dementia in adults older than 40 years with Down's syndrome (MEADOWS): a randomised, double-blind, placebo-controlled trial. Lancet. 2012;379:528–536. doi: 10.1016/S0140-6736(11)61676-0. [DOI] [PubMed] [Google Scholar]

RESOURCES