Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Sep 23.
Published in final edited form as: Curr Alzheimer Res. 2016;13(1):35–52. doi: 10.2174/1567205012666150921095505

Cognitive Impairment, Neuroimaging, and Alzheimer Neuropathology in Mouse Models of Down Syndrome

Eric D Hamlett 1, Heather A Boger 1, Aurélie Ledreux 1, Christy M Kelley 2, Elliott J Mufson 3, Maria F Falangola 4, David N Guilfoyle 5, Ralph A Nixon 6, David Patterson 7, Nathan Duval 7, Ann-Charlotte E Granholm 1,*
PMCID: PMC5034871  NIHMSID: NIHMS816589  PMID: 26391050

Abstract

Down syndrome (DS) is the most common non-lethal genetic condition that affects approximately 1 in 700 births in the United States of America. DS is characterized by complete or segmental chromosome 21 trisomy, which leads to variable intellectual disabilities, progressive memory loss, and accelerated neurodegeneration with age. During the last three decades, people with DS have experienced a doubling of life expectancy due to progress in treatment of medical comorbidities, which has allowed this population to reach the age when they develop early onset Alzheimer’s disease (AD). Individuals with DS develop cognitive and pathological hallmarks of AD in their fourth or fifth decade, and are currently lacking successful prevention or treatment options for dementia. The profound memory deficits associated with DS-related AD (DS-AD) have been associated with degeneration of several neuronal populations, but mechanisms of neurodegeneration are largely unexplored. The most successful animal model for DS is the Ts65Dn mouse, but several new models have also been developed. In the current review, we discuss recent findings and potential treatment options for the management of memory loss and AD neuropathology in DS mouse models. We also review age-related neuropathology, and recent findings from neuroimaging studies. The validation of appropriate DS mouse models that mimic neurodegeneration and memory loss in humans with DS can be valuable in the study of novel preventative and treatment interventions, and may be helpful in pinpointing gene-gene interactions as well as specific gene segments involved in neurodegeneration.

Keywords: Cholinergic neurons, Diffusional Kurtosis Imaging, Down syndrome, Intellectual disability, memory loss, neuro inflammation

1. INTRODUCTION

Down syndrome (DS) is a developmental genetic condition caused by trisomy of human chromosome 21 (Hsa21). Ninety-nine percent of individuals with DS have an extra copy of Hsa21, and for 90–95% the extra copy is a maternal Hsa21 [1]. DS occurs in 1 in 700 to 1200 live births worldwide, and is estimated to affect approximately 250,000 people in the USA [2]. DS is caused by an extra copy of the long arm of human chromosome 21, which leads to perturbations in expression of genes included in the trisomy segment. Neuropathological and clinical features of Alzheimer’s disease (AD) present in the fourth through sixth decade in individuals with DS [3, 4], and include neurofibrillary tangles (NFTs), amyloid plaques, basal forebrain cholinergic neuron (BFCN) loss, inflammation and dementia resembling AD [5]. People with DS exhibit a higher penetrance of AD pathology in those that survive long enough compared to the disomic population [6, 7]. The co-occurrence of dementia resembling AD in DS (DS-AD) rises steeply with age [8, 9], making DS an excellent natural genetic model for the study of AD-like biological mechanisms and potential biomarkers of both disorders. Moreover, unlike presenilin-linked familial AD, the age of onset of dementia is regulated by the apolipoprotein E (APOE) allele among individuals with DS and can therefore be stratified by the presence of the ε4 allele like typical late onset AD [10]. Despite the molecular and genetic similarities between AD and DS-AD, there is a lack of information on the biological mechanisms underlying the onset of AD pathobiology in people with DS. The development of appropriate mouse models for DS during the last several decades has enabled in depth studies of biological mechanisms, many of which have led to clinical trials of several classes of drugs for DS-AD.

Biological mechanisms involved in DS-AD include extracellular amyloid β protein (Aβ) accumulation, intraneuronal neurofibrillary tangles (NFTs) deposition, BFCN cell loss [11], neuron loss in locus coeruleus (LC) [12], hippocampal abnormalities [13], imbalance of neurotrophic factors [14], alterations in long-term potentiation (LTP) [15], abnormal endosomal signaling [16], presence of neuroinflammation [17], and oxidative stress [18]. Due to its localization on Hsa21, the amyloid precursor protein (APP) gene is triplicated in DS and Aβ deposition is frequently profound in persons with DS. Aβ accumulation in amyloid plaques typically does not begin in the DS brain until after the age of 30 years [19], although diffuse plaques have been observed in individuals with DS in their teens and 20’s [20, 21]. Work from multiple groups including our own has demonstrated increased levels of APP and Aβ, but not in classic amyloid plaque formation, in the Ts65Dn mouse model of DS [2226]. In addition to the pronounced Aβ deposition in subjects with DS, they also have the presence of intracellular tau-containing NFTs [18, 27]. Previous studies have shown that neuropathology accumulates progressively when individuals with DS are in their 30s and 40s [28]. While a proposed time course for the progression of NFT pathology in the entorhinal cortex is in the mid-30s, hippocampus in the mid-40s, and neocortex in the mid-50s, no consistent appearance of tangle pathology between DS brains has been demonstrated [19, 2830]. However, there is consensus that NFTs do not appear before the age of 20 in DS [7, 31]. The progressive accumulation of AD pathology suggests that there is a preclinical phase in DS-AD with a delay of at least 10 years between the onset of AD pathology and dementia diagnosis, similar to what is described for AD [32, 33], providing a window for intervention. However, due to the difficulty in obtaining clinically and pathologically well-characterized human DS brain tissue, the need to develop and investigate animal models of this disorder is critical.

The use of mouse models to study DS allows approaches that are not possible in humans. Mouse models are beneficial for: 1) Disease progression studies in a controlled environment, 2) Trials of prevention or intervention strategies, 3) Validation of imaging and surgical procedures, and 4) Studies of gene-gene interactions in the Hsa21 region involved in DS. There are a minimum of 166 protein coding genes in Hsa21, and the regions on Hsa21 are syntenically conserved with three regions located on murine chromosome (Mmu) 10, 16 and 17 [34, 35]. The Ts65Dn mouse, contains a majority of the Hsa21 orthologous protein-coding genes that map to Mmu16, but lacks trisomy for about 45% of Hsa21 ortholog genes [36]. Thus, this model has genetic limitations as a model of age-related AD pathology in DS. Nonetheless, Ts65Dn mice exhibit several deficits seen in individuals with DS, including progressive memory decline [22, 37], adult-onset degeneration of BFCNs and LC-NE neurons [3840], hippocampal abnormalities [41, 42], and increased APP production [43, 44]. Recent efforts have been made to make a DS mouse model with overexpression of the Hsa21 gene segment in the attempt to more closely mimic human AD pathology such as classical diffuse or neuritic plaques. In the current review we will discuss findings related to aging and AD neuropathology in Ts65Dn mice, as well as comparative studies in other DS mouse models.

2. SUMMARY OF MOUSE MODELS OF DOWN SYNDROME

One of the first models of DS to be developed was the complete trisomy of the murine chromosome 16 (Ts16), which proved to be lethal [45]. Bambrick et al. [46] demonstrated that hippocampal neurons from the Ts16 mouse die at an accelerated rate via caspase-mediated apoptosis and that caspase inhibitors prevent abnormal neuronal loss in Ts16 mice. Because complete Mmu16 trisomy is lethal, this model can only be used for in vitro studies. Other DS mouse models have also been developed to investigate the role of different gene segments of Hsa21 on AD pathology and memory impairment associated with DS-AD. A summary of these different mouse models is provided in Fig. 1. The Ts65Dn mouse model for DS, developed in the early 90’s by Muriel Davisson [47], is the most widely used mouse model for DS, and represents about 120 orthologs of Hsa21 protein encoding genes through a segmental trisomy for Mmu16 (Fig. 1). Mrpl39 is the proximal endpoint for the segmental trisomy in Ts65Dn [48]. This mouse model is also trisomic from a segment of about 10 Mb of the Mmu17, which contains 60 protein encoding genes, none of which are homologous to Hsa21 genes. Most of these have orthologues on Hsa6 [35, 48]. Although the aneuploidy in Ts65Dn is not lethal, there is some evidence that these mice have a shorter lifespan than diploid mice [49]. Another mouse model for DS, Ts1Cje, has shorter Mmu16 trisomy than the Ts65Dn mouse, contains 62 orthologs of Hsa21 genes, and excludes the gene segment containing APP and superoxide dismutase 1 (SOD1, Fig. 1). These mice display certain neurodegenerative features of the processes identified in Ts65Dn mice, including oxidative stress [50], tau hyper-phosphorylation, mitochondrial dysfunction [51], and show some learning and memory deficits [52]. The Ms1Ts65 mice are trisomic for the region of difference between Ts1Cje and Ts65Dn, contains 56 orthologs of Hsa21 genes within genetic segment from Mrp139 to Sod1.

Fig. 1.

Fig. 1

Summary of available DS mouse models to date. Hsa = human chromosome, Mmu = murine chromosome, gray lines delineates included gene segments per mouse, and light gray dotted box represents the so called Down syndrome Critical Region (DSCR).

The closely related Ts65Dn, Ts1Cje, and Ms1Ts65 models have been examined by Sago and collaborators in order to assess differences in behavioral phenotypes from their aneuploid segments. Spontaneous locomotor testing showed that Ts65Dn mice were hyperactive, Ts1Cje mice hypoactive, and Ms1Ts65 mice exhibited normal spontaneous activity [53]. The performance in Morris water maze of Ts1Cje mice was similar to that of Ts65Dn mice, but reverse probe tests demonstrated a more severe deficit in Ts65Dn compared to Ts1Cje mice, suggesting consolidation problems unique to the Ts65Dn mice. This is consistent with observations by Lockrow et al., showing that Ts65Dn mice exhibit significant overnight consolidation problems in a water radial arm maze [54]. The memory deficits in Ms1Ts65 mice were much less severe than in Ts65Dn mice. Sago and collaborators [53] concluded that triplication of SOD1 to Mx1 plays a significant role in memory loss observed in Ts65Dn, and that an imbalance of the APP-SOD1 gene segment also contributes to the poor performance. These findings further indicate that genes in the Ms1Ts65 trisomic region interact with others in the Ts1Cje region to produce hyperactivity in Ts65Dn mice. Histomorphological studies of these related DS models have revealed key hallmark changes that may underlie the behavioral phenotypes observed. APP overexpression, seen in Ts65Dn but not in Ts1Cje mice, is necessary to produce cholinergic cell loss [55] and DS-AD neuronal endosomal abnormalities seen at the earliest stage of disease [25, 56, 57] and during infancy long before disease hallmarks are present [57]. Moreover, APP overexpression is similarly linked to cholinergic degeneration associated with NGF transport deficits observed with aging in Ts65Dn mice. These findings suggest that over-expression of APP plays a significant role in AD neurodegeneration and dementia in DS.

Another relatively novel model for DS is the Ts2Cje mouse model Ts(Rb(12.1716))2Cje (Ts2) [58]. Ts2 mice have a similar aneuploid segment to Ts65Dn mice except that a chromosomal rearrangement of the Ts65Dn genome caused a translocation to Mmu12 forming a Robertsonian chromosome [59]. Studies performed in the Nixon and Falangola laboratories indicate that the pattern of brain network abnormalities is similar between the Ts2 and the Ts65Dn mice (see Neuroimaging section below) and further analyses by Jiang et al., (submitted report) show that the sequence of characteristic age-related endosome abnormalities and neurodegenerative changes of basal forebrain cholinergic neurons are similar, confirming the validity of these models for neurobiological studies of DS abnormalities. Finally, Yu and collaborators have developed another novel mouse model for DS that carries complete aneuploidy (spanning the entire Hsa21 syntenic region) [60]. After generating separate aneuploid mouse models: Mmu10− Dp(10)1Yey/+ (Ts1Yey), Mmu17− Dp(16)1Yey/+ (Ts2Yey), and Mmu16− Dp(17)1Yey/+ (Ts3Yey), the triple aneuploid mice (Ts1Yey; Ts2Yey;Ts3Yey) were established by standard breeding [60]. The investigators found that the genotype of Ts3Yey resulted in abnormal hippocampal LTP, but the genotype of Ts2Yey resulted in impaired performance in learning and memory tasks, as well as abnormal hippocampal LTP, suggesting that genes syntenic to Hsa21 on Mmu16 may represent a crucial element for memory loss in DS. Trisomy-based differences in brain morphology were similar in Ts3Yey compared to Ts65Dn mice [60]. These results validate the genetic basis for behavioral and morphological DS mouse model phenotypes, and carry promise for developing more appropriate and complete mouse models for DS in the near future. Moreover, this collection of mouse models allows for assessment of the importance of the Mmu17 and Mmu10 homologous regions in generation of the phenotypes of DS. For example, Zhang et al. [61] have recently shown that the Mmu17 region is a significant determinant of DS related cognitive deficits.

While many mouse models of DS involve aneuploidy of Mmu segments, a notable DS model was created from the trans-species insertion of Hsa21. The Tc1 mouse model for DS carries most of human chromosome 21 in addition to the normal complement of mouse chromosomes, and is trisomic for approximately 212 Hsa21 protein-encoding genes ([36]; Fig. 1). Gardiner and collaborators have shown that Tc1 mice show many of the phenotypes characteristic of DS, including abnormalities in learning and memory and synaptic plasticity [36]. In comparison to Ts65Dn mice, the Tc1 exhibit elevated S100B calcium-binding protein, AMPK, and the mTORC1 proteins RAPTOR and downstream kinase P70S6 [62], which are important regulators of cellular metabolism and aging. Therefore, these mice may represent a novel and yet fairly unexplored model for DS, aging and AD. Due to a genetic rearrangement involving the APP gene, expression of the full length APP transcript cannot be detected in the Tc1 brain, and no human APP protein is found in the Tc1 brain [63]. Therefore, even in the absence of a functional third copy of the APP gene, Tc1 mice still demonstrate a DS phenotype and associated molecular signaling changes that may underlie neuropathology. These data establish the importance of aneuploidy of non-APP genes in DS neuropathology. Although the Tc1 DS mouse model may be an excellent model to examine aging, to our knowledge no aging or intervention studies have been carried out to date. Tc1 mice are mosaic for and have some Hsa21 rearrangements attributed to gamma irradiation of the Hsa21 during production of the Tc1 mice, which complicates their use for some studies but they can, nonetheless, be useful for comparison studies of Hsa/Mmu traits [63]. Interestingly, the production of the Ts65Dn mouse model also involved irradiation of the testes of the male mice used to make these mice [47]. The extent of damage due to Cesium137 irradiation of the extra chromosome in Ts65Dn mice has not been tested.

To our knowledge, age-related memory deficits and AD pathology that occur in Ts65Dn mice have not been examined in other DS mouse models. The newly developed DS mouse models may more closely mimic neuropathology occurring in humans with DS-AD, and most are made available as a shared resource to researchers. Attrition studies have not been conducted officially on any of the DS mouse models to date, although we have found that Ts65Dn mice can age quite successfully, until at least 20 months of age, with similar age-related attrition as seen in normosomic littermates (Granholm, personal communication). A summary of studies focused on cognitive impairment in DS mouse models, along with the age of the cohorts studied, is shown in Section 3 (Table 1). In addition to the mouse models listed, there are numerous mouse models transgenic for one or more Hsa21 genes and in which one or more Hsa21 genes have been inactivated by targeted mutagenesis (knockout mice). These mice can be used either separately or in conjunction with the models described above. For example, APP knockout mice have been used to reduce the APP gene in Ts65Dn mice to two copies, thus allowing for the dissection of the effects of trisomy of a single gene. Until recently, this technology was somewhat limited by the availability of appropriate knockout mice. However, with the advent of CRISPR/Cas9 technology, the generation of appropriate mice for these applications is likely to improve markedly [64].

Table 1.

Behavioral alterations in DS mouse models.

Mouse model: Behavior: Direction of change Age References
Tc1 -Sleep disturbance Fragmented sleep pattern adult [191]
Ts65Dn -Nest building task Severely impaired young [192]
Ms2Yah -social novelty interaction
-contextual fear-conditioning
Decreased
increased associative memory
young [193]
Ts[Rb(12.1716)]2Cje; Ts2Cje (Ts2) -nest building behavior
-spontaneous alternation
Impaired
Impaired
Young adult [88]
Ts65Dn/DnJ -Object recognition and consolidation Impaired Adult [194]
Ts65Dn -Spatial working and reference memory Impaired (age-dependent) 6–18 month old [67, 68, 195]
Ts1Cje -Morris Water maze
-Spontaneous locomotion
Less severely affected
Hypoactive
Adult [53]
Ts65Dn Elevated plus maze Increased activity Adult [196]
Ms1Ts65 (trisomic for APP to SOD1) -Morris water maze
-Spontaneous locomotion
Less severe deficits
Normal activity
Adult [53]
Ts65Dn -Impulsivity
-motor activity
Unaltered Adult/ aged [75]
Ts65Dn -conditioned taste aversion Impaired 12 months [158]
Ts65Dn -Locomotor activity increased 2–12 months [137, 197]
Ts65Dn -WRAM Severely impaired 13–17 months [174, 198]
Ts65Dn Sustained attention task Impaired (error-induced stereotypy) 15–17 month [65]
Dp(10,16,17)1Yey Morris water maze Impaired Young [61]

3. COGNITIVE IMPAIRMENT

Several research groups have demonstrated that Ts65Dn mice exhibit a progressive loss of learning and memory function with age, which starts when they are approximately 6 months old and worsens with age [37, 65, 66]. The Ts65Dn mouse has undergone extensive behavioral testing, and has been shown to exhibit severe spatial and recognition memory deficits [6769]. Impaired performance has been observed in several different memory tasks including the 8-arm water radial arm maze [70], novel object recognition tasks [52, 71], and the Morris water maze [7274], see also Table 1. In a meta-analysis of Ts65Dn performance in the WRAM maze at 4, 8, and 10 months of age (Fig. 2A), we found significant genotype effects in the last four trials (F=4.708; p=0.0026), strongly suggesting deficiencies in the learning process in Ts65Dn mice compared to Normosomic (NS) littermates. In post hoc analyses using Bonferroni correction, we also found that 10 month old Ts65Dn mice exhibited the worst performance of both genotypes at any age tested (p=0.0030), suggesting a progressive reduction in performance in this task with age. Stasko and collaborators [73] performed memory and learning studies of Ts65Dn mice, and noted that the behavioral phenotype of Ts65Dn mice was milder than previously described in the literature. These studies were performed on young adult mice, and did not include aged cohorts, demonstrating the importance of aging in DS mouse investigations. Ts65Dn performance on several different behavioral tasks gets worse from 6 to 12 months of age and 4-month old mice are not severely deficient, at least not on spatial memory tasks [23]. Ts65Dn mice exhibit hyperactivity already at a young age [75], and a meta-analysis of Ts65Dn spontaneous activity demonstrated that hyperactivity also increases as the mice age (Fig. 2B).

Fig. 2.

Fig. 2

(A) = Analysis of WRAM performance in Ts65Dn mice (gray symbols) and age-matched NS mice (black symbols) from several different experiments across age. Note that the Ts65Dn mice exhibit no statistical difference in average errors the first 4 trials, indicating that they are capable of performing the task, but have significantly more average errors during the last 4 trials, indicating significant reduction in learning. (B) Analysis of total spontaneous activity (in cm) in Ts65Dn mice (gray) and age-matched normosomic mice (NS, black) across several different experiments. Note that there is significant hyperactivity at all ages examined (6–13 months of age). (C) Analysis of locus coeruleus (LC) phenotypic loss from 4 to 13 months of age. TS = Ts65Dn; NS = normosomic. Comparative study across several different cohorts of mice. There is a progressive loss of LC neurons with age, most notable at 13 months of age.

As can be seen in Table 1, few studies [76] have been conducted on the aging process of other DS mouse models. However, it will be exciting to evaluate the aging process and the development of AD pathology and age-related memory loss in these newly developed DS mouse models.

4. NEUROIMAGING OF MOUSE MODELS OF DOWN SYNDROME

Despite the fact that mouse models of DS have been characterized cognitively and histomorphologically, no neuroimaging biomarker evaluation of developmental or neurodegenerative abnormalities has been performed. Only a few in vivo imaging studies have been published in humans [7684] or mouse models [8589] of DS. In humans, positron emission tomography (PET) measurements of β-amyloid burden [77, 79, 81], regional PET measurements of the cerebral metabolic rate for glucose [76] and structural and functional connectivity differences by anatomical and functional magnetic resonance imaging (MRI) studies [78, 80, 82, 84] demonstrate the safety, efficacy, and acceptability of these imaging techniques to study DS-AD progression. Using diffusion tensor imaging (DTI), Powell et al. [83] recently reported that changes in white matter integrity, particularly in the frontal tracts, were associated with poorer cognition in adults with DS. This suggested that late myelinating fiber bundles in the frontal cortex are vulnerable to aging in DS. Hence, neuroimaging is an ideal method for analyzing the anatomical and functional phenotypes of DS mouse models, which may exhibit unknown cellular defects that develop in three-dimensions (3D) over time.

Thus far, only a few in vivo neuroimaging studies in mouse models of DS have been published using MRI [8589]. Chen et al. [85] reported decreased transverse relaxation time (T2) in the cortical and hippocampal cholinergic circuitry in middle-aged (> 12 months old) Ts65Dn mice, demonstrating the utility of quantitative in vivo MRI for identifying AD-relevant cholinergic changes in animal models of DS. Structural MRI revealed ventricular enlargement that may be associated with impaired neurogenesis in the brains of Ts1Cje (Ts1) and Ts2Cje (Ts2) mouse models [86]. In vivo magnetic resonance spectroscopy (MRS), demonstrated reduced glutamate in the hippocampus of Ts2 mice, which was accompanied by reduced mRNA and protein levels of N-methyl-D-aspartate receptors (NMDA-R1) [88]. Recently, in a study using high-field MRS, investigators reported lower levels of glutamine and higher levels of myo-inositol in the hippocampus of Ts65Dn mice, and elevated myo-inositol has also been found via MRS in humans with DS [87]. Lastly, our group recently published the first diffusion MRI (dMRI) results in Ts65Dn mice, using diffusional kurtosis imaging (DKI) [89]. Diffusion MRI is uniquely positioned for exploring brain tissue microstructure, function and connectivity, and to provide evidence of brain plasticity in vivo, allowing longitudinal assessment of ongoing processes. DKI is a specific dMRI technique that quantifies the non-Gaussian behavior of water diffusion [9092]. Aside from yielding all of the diffusion indices conventionally obtained with diffusion tensor imaging (DTI), DKI also gives metrics of non-Gaussian diffusional, such as mean (MK), axial (K//) and radial (K) kurtoses, which more comprehensively characterize brain cytoarchitecture. Our own animal investigations, as well as studies from other groups, have shown that DK metrics are sensitive to changes in brain microstructural complexity that may be associated with brain development [93], Aβ deposition [94], stroke [95, 96], and myelin abnormalities [97].

In our diffusion MRI study in the Ts65Dn mice, DKI metrics allowed detection of early brain tissue abnormalities, with distinct patterns in age-related microstructural trajectories in frontal cortex, striatum and hippocampus of the Ts65Dn mouse model of DS [89]. We showed that Ts65Dn mice have higher DKI metrics in the frontal cortex compared with normosomic (NS) littermates at 2 months and do not change significantly through 8 months of age. We also showed that, in the dorsal hippocampus, Ts65Dn mice have lower DK metrics compared with NS and do not change significantly with age, except for the axial kurtosis (K//). Although interpretation of changes in diffusion MRI metrics is complex, DKI metrics tend to increase with diffusional heterogeneity and are altered by water exchange or by diffusion barriers. Previous studies suggest that increased kurtosis values in the frontal cortex of 2-month old Ts65Dn mice reflect increased heterogeneity due to abnormal cortical lamination and altered structural dendrite and spine abnormalities in this region [98, 99]. Also, lower DK values in the dorsal hippocampus of Ts65Dn mice may be related to defects in axonal spread and enlargements of dendritic spines [98, 99] causing diffusion dead-space micro-domains, as well as, abnormal myelination of the hippocampal formation, as described in humans [100, 101]. Lower radial kurtosis (K) values in the striatum may also be related to abnormalities in the myelination process [101].

Since the Ts2 mouse model shows a similar degree of neurogenesis impairment, plasticity alterations and behavioral impairments as described in Ts65Dn mice [59], we investigated if DKI metrics would also detect brain tissue abnormalities. Therefore, we studied 10-month old male Ts2 mice (Ts2; n = 5) and control NS littermates of Ts2 mice (NS; n = 4). The in vivo MRI experiments were performed on a 7T Agilent MR system at Nathan Kline Institute (NKI) using a respiratory gated 6-shot SE-EPI sequence for DKI acquisition (see Table 2 for imaging details). All of the dMRI metrics were estimated from the diffusion and diffusional kurtosis tensors [90], and we studied the same brain regions of interest (ROIs) as in our previous study on the Ts65Dn mice (e.g., frontal cortex (FC), striatum (ST) and hippocampus (Dorsal - HD; Ventral - HV) [89]. Similar to the Ts65Dn results (Table 2), DT metrics showed no group differences in any of the brain ROIs assessed in the Ts2 mice. In contrast, DK metrics showed significant differences compared with NS mice, with decrease in all DK metrics in the ST, HD and HV, which supports the known morphological and plasticity alteration similarities between these two DS mouse models [59]. The changes observed in the hippocampus in both DS mouse models at later ages (8–10 months of age), when cognitive defects are present [52, 69, 74, 102], may be associated with neurodegenerative changes related to neuroinflammation, increased oxidative stress, BFCN degeneration and age-related increased APP transcription and Aβ levels, as previously described [5, 103].

Table 2.

Diffusion metrics estimates (mean ± standard error of the mean (SEM)) in each brain region of interest for the 8-month old Ts65Dn and 10-month old Ts2 mice. The sequence parameters were: TR/TE=4000/29 ms, δ/Δ=5/17 ms, slice thick-ness=1 mm, 14 slices with 0.1 mm gap, data matrix=96×96 zero filled to 128 × 128, image resolution=208×208 μm2, 1 average, 30 gradient directions and two b-values for each gradient direction (1 and 2 ms/μm2). A non-diffusion weighted image (b = 0 ms/μm2) was also acquired. DKI post-processing was performed using in-house Diffusional Kurtosis Estimator (DKE) (http://nitrc.org/projects/dke) [199]. Parametric maps of the conventional diffusion tensor (DT) metrics of mean (MD), axial (D) and radial (D) diffusivities, as well as the additional DK metrics of mean kurtosis (MK), axial kurtosis (K|), and radial kurtosis (K) were subsequently computed. One-way analysis of variance (ANOVA) was performed to compare the mean of diffusion metrics between the two groups, with significant p-values in bold; ROIs: frontal cortex (FC), striatum (ST) and hippocampus (dorsal – HD; ventral - HV).

MD
μm2/ms
D//
μm2/ms
D
μm2/ms
MK K// K

Ts65Dn FC NS 0.79 ± 0.06 0.92 ± 0.09 0.72 ± 0.05 0.75 ± 0.05 0.72 ± 0.04 0.75 ± 0.06
TS 0.78 ± 0.02 0.92 ± 0.03 0.71 ±0.02 0.68 ± 0.04 0.64 ± 0.05 0.66 ± 0.06
p-values 0.81 0.94 0.64 0.04 0.03 0.06
ST NS 0.75 ± 0.02 0.91 ± 0.04 0.67 ± 0.02 0.86 ± 0.06 0.86 ± 0.05 0.97 ± 0.07
TS 0.74 ± 0.02 0.88 ± 0.03 0.67 ± 0.01 0.80 ± 0.05 0.78 ± 0.06 0.86 ± 0.09
p-values 0.48 0.25 0.92 0.15 0.03 0.04
HD NS 0.79 ± 0.01 0.87 ± 0.01 0.74 ± 0.01 0.82 ± 0.01 0.83 ± 0.01 0.85 ± 0.02
TS 0.78 ± 0.01 0.87 ± 0.01 0.73 ± 0.00 0.69 ± 0.02 0.71 ± 0.02 0.70 ± 0.03
p-values 0.35 0.84 0.24 0.001 0.001 0.002
HV NS 0.79 ± 0.01 0.88 ± 0.02 0.75 ± 0.01 0.80 ± 0.02 0.77 ± 0.03 0.82 ± 0.03
TS 0.81 ± 0.02 0.91 ± 0.02 0.76 ± 0.02 0.71 ± 0.04 0.71 ± 0.04 0.71 ± 0.04
p-values 0.4 0.22 0.52 0.06 0.26 0.07

Ts2 FC NS 0.85 ± 0.01 1.06 ± 0.02 0.80 ± 0.05 0.54 ± 0.03 0.71 ± 0.05 0.46 ± 0.02
Ts2 0.87 ± 0.01 1.07 ± 0.02 0.76 ± 0.05 0.48 ± 0.02 0.62 ± 0.04 0.40 ± 0.02
p-values 0.31 0.57 0.07 0.11 0.22 0.08
ST NS 0.82 ± 0.01 1.08 ± 0.03 0.81 ± 0.08 0.76 ± 0.02 0.91 ± 0.02 0.72 ± 0.04
Ts2 0.84 ± 0.01 1.07 ± 0.02 0.72 ± 0.07 0.65 ± 0.02 0.84 ± 0.02 0.59 ± 0.04
p-values 0.37 0.66 0.48 0.002 0.04 0.05
HD NS 0.89 ± 0.02 1.05 ± 0.04 0.82 ± 0.01 0.65 ± 0.02 0.83 ± 0.03 0.56 ± 0.03
Ts2 0.91 ± 0.01 1.09 ± 0.04 0.83 ± 0.01 0.56 ± 0.03 0.72 ± 0.03 0.45 ± 0.03
p-values 0.25 0.55 0.67 0.01 0.02 0.02
HV NS 0.86 ± 0.02 1.05 ± 0.04 0.78 ± 0.02 0.63 ± 0.02 0.82 ± 0.02 0.50 ± 0.02
Ts2 0.90 ± 0.02 1.09 ± 0.04 0.81 ± 0.02 0.52 ± 0.02 0.72 ± 0.02 0.40 ± 0.02
p-values 0.11 0.4 0.21 0.01 0.01 0.02

Taken together, neuroimaging studies demonstrate that structural MRI, MRS and dMRI can detect changes in the brain of DS mouse models. DKI provides a sensitive indicator of complex neurite architecture abnormalities seen in both Ts65Dn and Ts2 mouse models of DS, which supports the viability of using DKI-based metrics as potential neuroimaging biomarkers of disease processes and for monitoring future neuroprotective interventions in DS. These studies may also assist in the interpretation of results from future translational studies related to human brain microstructure abnormalities in DS.

5. NEURONAL CELL LOSS

BFCNs provide the major cholinergic innervation to frontal cortical regions and the hippocampus, and thereby influence the processing of information necessary for attention and cognition in both animal models and in humans [104106]. The phenotypic loss of cholinergic neurons is a hallmark of AD, and correlates strongly with memory loss [107]. Individuals with DS exhibit progressive degeneration of BFCNs [11, 108110]. BFCN neuropathology is mirrored in the Ts65Dn mouse model of DS-AD, with an age-related degeneration, commencing around 6–8 months of age [3840, 44, 55, 111] with significant atrophy of BFCN cell bodies shown at 6 months of age, and significant phenotypic loss between 8 and 10 months of age (Table 3; Fig. 3A and B). Further, a decrease in immunohistochemistry for both choline acetyltransferase (ChAT) and the high-affinity nerve growth factor (NGF) receptor TrkA has been demonstrated in BFCN neurons at this age. These findings are consistent with the increased deficiency on hippocampal-dependent spatial memory tasks, such as the WRAM, during this age span [39].

Table 3.

Literature summary of BFCN cell loss in Ts65Dn mice compared with age-matched NS mice.

Sex Age Marker Region Results References
M 4 mos ChAT, TrkA MS, VDB – cell size [39]
M 4 mos,
8 mos
MS, VDB – region area
M 6–10 mos ChAT MS, VDB ↓ cell density
M 6 mos,
8 mos,
10 mos
TrkA MS ↓ cell size
M 8mos,
10 mos
ChAT MS ↓ cell size
M 6 mos p75NTR MS – cell count [38]
M 12 mos,
18 mos
p75NTR MS ↓ cell count
↓ cell size
ns 6 mos p75NTR MS – cell count
↓ cell size
[40]
ns 20 mos p75NTR MS ↓ cell count
↓ cell size
M 8–10 mos TrkA MS ↓ cell count
↓ cell size
[70]
M 10 mos ChAT MS ↓ cell count [174]
M 10 mos ChAT VDB – cell count
F 18 mos ChAT MS ↓ cell count
F 18 mos ChAT VDB – cell count
M,F 10 mos p75NTR MS – cell count [111]
M,F 19 mos ChAT, p75NTR MS ↓ cell count
M,F 19 mos p75NTR VDB – cell count
F 11–14 mos ChAT MS – cell count
– cell size
[114]
ns 12 mos ChAT MS/VDB/
HDB*
↓ cell count
– cell size
[44]
M 19–22 mos p75NTR, TrkA MS ↓ cell count
↓ cell size
[55]

NS, normosomic mice born to Ts65Dn dams; ChAT, choline acetyltransferase; F, female; HDB, horizontal limb of diagonal band; M, male; MS, medial septum; ns, not specified; p75NTR, protein 75 kilodaltons neurotrophic receptor; sc, subcutaneous; TrkA, tyrosine kinase receptor A; Tx, treatment; VDB, vertical limb of diagonal band

ns = sex of mice studied not specified

NS = normosomic

*

Regions combined

Fig 3.

Fig 3

Cell loss in basal forebrain (TrkA staining), hippocampus (Calbindin), and locus coeruleus (tyrosine hydroxylase, TH) in middleaged Ts65Dn mice compared to age-matched normosomic mice (NS). In all these three neuronal populations, there is a progressive loss of phenotype with age, starting at 6 months of age. For BFCNs, this phenotypic loss is significant at 8 months of age, and for the LC and hippocampal neurons at 10–12 months of age. As can be seen in these representative images, phenotypic loss is accompanied by neurodegenerative shrinkage of cell bodies. Scale bar in (A) represents 150 microns, in (D) 60 microns, and in (F) 50 microns.

Evaluation of the age-dependent changes in the activity of ChAT and the acetylcholine-degrading enzyme acetylcholinesterase (AChE) revealed that in 10-month old Ts65Dn mice, ChAT activity was significantly higher compared to NS littermates in the hippocampus and prefrontal cortex, but not in other neocortical regions [111]. At 19 months of age, no differences in ChAT activity were found between the two genotypes, despite significant BFCN degeneration at this age [44]. Thus, alterations of ChAT activity in these forebrain areas seem to recapitulate changes described in patients with mild cognitive impairment (MCI) or mild AD, who exhibit an increase in ChAT activity in cortical regions, presumably as an adaptative reaction to BFCN degeneration [107]. Contestabile et al. [111] reported that neurochemical markers associated with disease progression, including inflammatory markers and APP cleavage products, were altered in some brain regions at the oldest age examined (19 months), suggesting continued progression of AD neuropathology with age in Ts65Dn mice. A summary of cholinergic deficits that occur in Ts65Dn mice and other DS models is provided in Table 3.

Further, female mice had smaller BFCNs in all cholinergic subregions examined compared with genotype-matched males. These findings demonstrate that differences between the genders in BFCNs of young-adult Ts65Dn and NS mice are region- and genotype-specific. The gender differences in BFCN degeneration also have clinical relevance. Studies have shown that women with DS are likely to have early menopause, and that the age of menopause onset correlates significantly with onset of dementia [113]. Our previous findings demonstrated that estrogen treatment prevented age-related BFCN loss and spatial memory loss in female Ts65Dn mice [114], providing evidence that adequate hormone replacement therapy may be a relevant investigative path for older adult women with DS.

Other neuronal populations also degenerate in human and mouse models of DS. Calbindin-D28K (Cal) neurons are known to degenerate in the human brain with age, with greater loss associated with AD [112]. Cal is a neuronal calcium binding protein, which may act as a buffer for neuronal calcium. Cal-positive pyramidal cells do not accumulate NFT in AD, which suggests that loss of Cal expression in specific hippocampal neuronal populations may be associated with reduced resistance to injury and loss [116]. Previous work in our laboratory demonstrated an age-related and significant loss of Cal immune-reactive neurons in the hippocampal CA1 region in Ts65Dn mice (Fig. 3C and D) [22]. These neurons are regulated by neurotrophin expression and play an important role in AD neuropathology observed in the hippocampus [116, 117]. Neurotrophin regulation provides a possible biological mechanism for hippocampal neuronal loss in aged Ts65Dn mice, since these mice exhibit age-related loss of mature NGF and BDNF levels in the hippocampus [14, 43, 44, 68] (also discussed in the Cuello et al. chapter in the current issue [118]).

Finally, a great deal of interest has been generated in the loss of LC-NE neurons and noradrenergic (NE) cortical innervation in both DS-AD and AD [119123]. NE neurotransmission exerts effects on neurons, glia, and blood vessels throughout the neuro-axis [124, 125]. LC-NE lesions using the selective NE neurotoxin, N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP-4), give rise to aggravated amyloid accumulation, oxidative stress, and memory loss in transgenic AD models [126, 127], and our previous work has shown that DSP-4 lesions in Ts65Dn mice give rise to accelerated memory loss, inflammation, and neurodegeneration [128]. LC-NE neurons degenerate with age in Ts65Dn mice, to the same extent as BFCN neurons (Fig. 2C, Fig. 3E and F) [128]. Effects of LC-NE degeneration on brain function are mediated directly, via neurotransmission, and indirectly, via aggravation of Aβ accumulation, inflammation, and oxidative stress pathways [129, 130]. NE circuitry exerts a direct influence on BFCNs via α1-adrenergic receptor activity [131], and pharmacological stimulation of NE receptors leads to improved cognitive performance in rodent models and in humans [132135]. Salehi et al. [136] demonstrated successful recovery from memory loss in Ts65Dn mice using the NE precursor Droxidopa (L-threo-dihydroxyphenylserine). In support of this study, a meta-analysis of several different groups of Ts65Dn mice confirmed the reported LC degeneration with age in Ts65Dn mice (Fig. 2C, 3E and F), see also [128]. In further studies aimed at rescuing LC function, Fortress and Hamlett, et al. used a Gq-coupled designer receptor exclusively activated by a designer drug (DREADD-hM3Dq) to selectively stimulate aged LC-NE neurons in Ts65Dn mice [137]. The DREADD receptor was administered via AAV into the LC under a synthetic promoter to dopamine-beta-hydroxylase (DBH), PRSx8, to selectively stimulate LC neurons by exogenous administration of the inert DREADD ligand, clozapine-N-oxide (CNO). DREADD stimulation of LC-NE neurons enhanced performance in a novel object recognition task and reduced hyperactivity in Ts65Dn mice, without significant behavioral effects in normosomic age-matched controls. We further confirmed that the NE transmitter system was responsible for the enhanced memory function, by administering the NE pro-drug L-threo-dihydroxyphenylserine (L-DOPS) in separate groups of Ts65Dn and NS mice, which produced similar behavioral results [137].

A recent in vivo study demonstrated that administration of physiological levels of NE prevented Aβ (Aβ1–42 and Aβ25–35) toxicity in human NT2N (hNT) neurons and rat primary hippocampal neurons as well as inhibiting Aβ25–35-mediated increases in intracellular reactive oxygen species, mitochondrial membrane depolarization, and caspase activation in hNT neurons [138]. Taken together, these data suggest that the neuroprotective effects of LC-NE afferent projections result from stimulating neurotrophic NGF and BDNF autocrine or paracrine loops via β-adrenoceptor activation of the cAMP response element binding protein pathway. Thus, NE stimulation may prevent memory loss in Ts65Dn mice and may hold promise for treatment in individuals with DS and dementia as the LC-NE transmitter system is intricately involved in attention, executive function, and several other domains of learning and memory [139, 140]. LC-enhancing medications, such as Atomoxetine or L-DOPS [141] have been utilized for individuals with Attention Deficit Disorder (ADHD), and could be considered in future clinical treatment paradigms for DS-AD patients, since attention deficits and executive dysfunction are common also in DS [142].

6. AMYLOID AND TAU PATHOLOGY

Individuals with DS develop Aβ depositions which are characteristic of early-onset AD in mid-life, presumably due to an extra copy of the chromosome 21- located APP gene [7, 16, 31, 143, 144]. The APP gene is triplicated in the Ts65Dn mouse, and this gene triplication is required for early endosomal abnormalities [25], diminished NGF transport, BFCN loss and cerebellar neuron loss in the Ts65Dn mouse [55]. These pathologies occur despite the absence of amyloid plaques in the Ts65Dn mouse [22, 44], though elevations in both APP, ßCTF and Aβ occur with aging in the hippocampus of this model. APP accumulates beyond the gene dosage in the striatum at 6–8 months of age and in the hippocampus at 13–16 months of age in Ts65Dn mice [43]. Similar elevation of soluble peptide levels (sAPPa and sAPPβ) were also seen at 12 months, but not at 4 months of age [24]. Recent evidence implicates ßCTF, which is elevated in AD and DS brain as well as DS models [145, 146] as the APP metabolite primarily responsible for the development of the endosome defects leading to cholinergic neuronal deficits [55]. In support of the ßCTF pathological role, endosomal transport and cholinergic deficits in fibroblasts from individuals with DS and neuron models require elevated levels of the ßCTF, and not Aß [145, 146]. Most recently, the adaptor protein APPL1 (adaptor protein containing pleckstrin homology domain, phosphotyrosine binding (PTB) domain, and leucine zipper motif) was identified as the critical molecule that directly binds to endosomal ßCTF and activates rab5, the effector of endosome dysfunction leading to defective endosome transport and hallmark cholinergic degenerative changes [146, 147]. Although most investigators have not found extracellular Aβ aggregates in mouse models of DS, Lomoio et al. described small deposits of Aβ in the cerebellum of middle-aged T65Dn mice [26]. These investigators also found evidence for tau protein hyper-phosphorylation in cerebellar neurons, suggesting that Ts65Dn mice are affected by a cerebellar tauopathy. Degeneration of cholinergic and cerebellar neurons in the Ts65Dn mouse has been shown to result in part from the additional gene load of APP [55], clearly linking the APP hypothesis with neurodegeneration that occurs in DS-AD (see Head et al. in the current issue [21]).

There is substantial evidence that peptide monomers and oligomers of Aβ, derived from β-secretase activity, are toxic in the brain, disrupting cell signaling, causing synapse degeneration, and in some cases leading to neuronal death, especially in vulnerable neuronal populations such as the BFCNs [148] although evidence implicating Aß toxicity as the primary factor leading to cholinergic dysfunction in DS are less clear. APP-FL levels are elevated according to gene dose in Ts65Dn mice, while sAPP accumulates with age [24, 43]. The homeostasis between the a - and β-secretase cleavage pathways may be modified by oxidative stress, since anti-oxidant treatments can reduce Aβ formation [149]. We have shown that vitamin E treatment in Ts65Dn mice prevents AD neuropathology (including neuroinflammation, BFCN cell loss, and gliosis) and memory loss, and also augments frontal cortex carboxyl terminal fragment (CTF) peptide cleavage [70]. Specifically, our findings suggested that vitamin E gave rise to increased CTFa peptide relative to APP-FL and CTFβ in Ts65Dn mice with aging [70], suggesting that the anti-oxidant treatment could, at least partially, rectify aberrant APP processing in DS.

The over-expression of APP may have consequences not only for age-related AD neuropathology, but also for brain development in DS. APP affects neural precursor cell proliferation, cell fate, and maturation of neurons, as well as gliogenesis [98, 150]. There are both intracellular and secreted domains of APP that appear to play a role for brain development [151]. This study showed that normalization of APP expression was able to restore neurogenesis and reduce astrocytosis, as well as increase neurite length of trisomic neuronal precursor cells, strongly suggesting that normal expression levels of APP are needed for development of both neuronal and glial elements of the brain. In a recent study by this group [151], the investigators found that deregulation of GSK3β activity was due to higher levels of the intracellular fragment of the trisomic gene APP that directly bound to GSK3β, and by restoring GSK3β phosphorylation either with lithium or the 5-HT receptor agonist fluoxetine, they could rescue neurogenesis and behavior in Ts65Dn mice, and restore GSK3β phosphorylation [151], therefore providing potential novel treatment paradigms for DS-AD. Fluoxetine is also shown to normalize GABA neurotransmission as well as reverse memory loss in adult Ts65Dn mice [152], providing further incentive towards exploring this serotonin agonist clinically in DS-AD, even though some studies indicate adverse effects of fluoxetine in this model [153].

Hyperphosphorylation of tau (p-Tau) is another prominent neuropathological hallmark of AD and DS-AD [5, 27, 154156]. Tau abnormalities have been observed in the Ts1Cje [51] and the Ts65Dn mouse models of DS [26, 58, 157, 158]. Studies indicate that dual-specificity tyrosine-phosphorylation regulated kinase 1A (Dyrk1A), which is overexpressed in DS and in Ts65Dn mice, may play a role for AD pathology development related to p-Tau. Qian et al. [157] have shown that Dyrk1A regulates tau expression in a dose-dependent manner, and increased tau levels were found in the brains of Ts65Dn mice that overexpress Dyrk1A by stabilizing its mRNA. Further, tau aggregations, which are elevated in Ts65Dn mice, were reduced following hippocampal implantation of neural precursor cells [158]. It is possible that the neural precursor cells increase endogenous growth factors or anti-inflammatory factors to exert these neuroprotective effects.

7. COMPARISONS BETWEEN DS AND AD MOUSE MODELS

The major mouse models employed for the study of AD are humanized, transgenic mice with human gene insertions, which increase the level of human Aβ production, most commonly through transgenes that include mutations in APP or presenilin, or combinations of the two [159]. Many of these transgenic models develop similar pathology: a progressive accumulation of Aβ deposits that begin in the hippocampus and entorhinal cortex. These deposits are surrounded by astrocytic gliosis and dystrophic neurites, and are highly similar to those seen in early AD in humans [160]. Along with plaque deposition, AD mouse models also exhibit elevated oxidative stress and neuroinflammation, but most AD models fail to show significant neuronal loss in any brain region, contrary to DS mouse models [161]. Transgenic AD mouse models expressing high levels of Aβ have memory loss and neuroinflammation/oxidative stress processes that are typical for the early phase of AD (for review, see Bilkei-Gorzo [162]). Neuropharmacological agents that have some clinical beneficial effects in humans with AD are often neuroprotective in AD mouse models, including the Tg2576, APP23, APP/PS1 and 3x-Tg AD models [162]. Of the most used AD models, the triple transgenic La Ferla model appears to have the highest validity in terms of development of AD pathology, neuronal degeneration, and memory loss [162]. The exogenous promoter for APP in these mice may not respond appropriately to inflammation since it is not murine APP, and therefore may not accurately model the “inflammatory hypothesis” in AD, which posits that inflammatory cells drive the production of Aβ through cytokine production. On the other hand, inflammation in the AD mouse models does appear to be driven by Aβ accumulation [163, 164], and anti-inflammatory treatment can reduce amyloid accumulation in AD mouse models [165]. The relationship between inflammation and amyloid accumulation appears to be different in DS compared to AD mouse models. As discussed above, inflammation, as well as memory deficits and oxidative stress are present in the hippocampus of Ts65Dn mice at 4 months of age, a period prior to significant increases in APP or Aβ levels [22, 43], and in the notable absence of Aβ deposits, which rarely develop in trisomy models for DS.

8. NEUROINFLAMMATORY AND OXIDATIVE STRESS PATHWAYS

As in AD, individuals with DS-AD consistently exhibit chronic neuroinflammation, with increases in microglial and astrocytic activation coupled with IL-1β and TNF-α cytokine release [17, 166, 167]. Microglial activation has been demonstrated in several brain regions in AD, including entorhinal cortex, hippocampus and surrounding cortex, and basal forebrain. BFCNs are highly sensitive to inflammation and oxidative stress [131, 168, 169], but specific biological mechanisms for their selective loss in AD and in DS-AD have not been revealed, although TNF-α induced cortical inflammation affecting cholinergic terminals can give rise to retrograde degeneration of BFCNs [170]. Since Ts65Dn mice exhibit significant degeneration of both BFCNs and LC-NE neurons, which both exert a massive innervation into the limbic system, it is not surprising that age-related astrocytosis and microgliosis are accelerated in the hippocampus of this mouse model of DS. Depletion of NE terminals in murine models of AD using the selective NE toxin DSP-4 results in increased inflammatory cytokine production, activated microglial morphology, and accelerated Aβ deposition [125, 126, 169, 171], and an NE lesion has similar results in Ts65Dn mice [128]. The tetracycline derivative minocycline has shown promise as a neuroprotective treatment for neurodegenerative disorders, based on its anti-inflammatory entities [172, 173]. Lockrow and collaborators demonstrated that minocycline treatment inhibited microglial activation, prevented progressive BFCN decline, and markedly improved performance of middle-aged Ts65Dn mice on a working and reference memory task [174], strongly suggesting that anti-inflammatory treatment strategies may succeed in DS-AD to at least partially prevent AD neuropathology and memory loss. Based on these studies, it is difficult to determine whether BFCN and LC-NE degeneration activates inflammatory pathways, or if the cytokine production by astrocytes and microglia, in turn, cause the neuronal degeneration in DS-AD and AD. Most likely, both processes have interactive and escalating effects on each other, leading, in the end, to memory loss and AD pathology, providing hope that anti-inflammatory therapies may be useful for prevention in DS-AD.

Another hallmark now recognized in the progression of AD is oxidative stress. Brain regions affected in AD show elevated oxidative stress markers [175, 176], and neurofibrillary tangles and amyloid plaques, both hallmarks of AD, colocalize with markers of oxidative stress in AD mouse models [161]. As in AD, individuals with DS-AD have elevated levels of DNA damage and lipid peroxidation [18, 177], and a pro-oxidant state is present early in the life, possibly due to the fact that superoxide dismutase (SOD) is located on the trisomic segment of Hsa21, leading to a dysregulation of the reactive oxygen species (ROS) scavenger pathway [178, 179]. Enhanced oxidative stress has also been observed in DS mouse models [50, 180]. DS neurons show increased sensitivity to oxidative stress in vitro [177, 181], and previous work suggests that vitamin E may directly protect neuronal function and improve survival. Studies in trisomy of Mmu16 show that defects in mitochondrial complex I contributed to elevations in reactive oxygen species (ROS) [180]. The potent antioxidant vitamin E was sufficient to protect BFCNs and LC neurons via reduced levels of ROS and neuroinflammation in aged Ts65Dn mice [70]. Similarly, previous studies assessing various antioxidants, including vitamin E, melatonin, and resveratrol have also shown similar benefit to mice transgenic for APP [161, 182]. Vitamin E delivery reduces isoprostane levels in both young and aged Tg2576 mice, yet only successfully reduces amyloid pathology in young mice [182]. These findings indicate that vitamin E appears capable of prevention, but perhaps not rescue from AD-like pathology. In a recent 2-year randomized, double-blind, placebo-controlled trial, investigators explored whether Vitamin E supplementation was safe and tolerable for individuals with DS over the age of 40 [183]. Vitamin E was well tolerated but ineffective as a treatment for dementia in individuals with DS-AD, although Vitamin E supplementation has also been tested in children and teenagers with DS, showing that antioxidant intervention with vitamins E and C attenuated the systemic oxidative damage present in DS children and teenagers [178], therefore holding promise as a neuroprotective therapy if initiated at an earlier age.

9. NEUROPROTECTION TRIALS IN MICE

Prior to the recent approval of memantine, a glutamate NMDA receptor antagonist, the primary treatment option for AD and DS-AD was acetylcholinesterase inhibitors (AChEIs), such as the second generation drugs donepizil, galantamine, and rivastigmine [184]. Each of these drugs has shown minor yet significant improvements to cognition and global status in clinical studies of mild to moderate AD patients. In particular, four small clinical trials using Donepezil, which included individuals with DS-AD suggest beneficial effects, including decreased confusion and cognitive improvement, though these trials were short (often under 6 months) and all involved relatively few patients [185, 186]. Despite studies in animal models that indicate that AChE inhibitors may act to modulate APP processing away from the amyloidogenic pathway [187, 188], more prolonged treatment paradigms using AChE inhibitors in AD patients have not lead to sustainable improvement or marked reductions in progression of the AD disease process. The consensus from meta-analyses of these trials is that AChEI are not capable of either neuroprotective or disease modifying effects [189]. Despite the short list of drugs available for DS-AD and AD patients in the clinic, a multitude of drug therapies have been tested and found relatively successful in DS and AD mouse models. A recent review by Costa et al. [190] summarizes both preclinical and clinical drug trials for the adult DS population. Table 4 shows a brief summary of drug trials in DS mouse models. The anti-oxidant and anti-inflammatory treatments that have been initiated in DS mouse models show promise, along with transmitter-modulating drugs such as the GABA receptor inhibitors and the NE-enhancing drugs (see Table 4), suggesting that the DS mouse models can be used to validate novel pathways towards successful drug therapy in DS-AD.

Table 4.

Summary of pharmaceutical trials in DS mouse models.

Drug Mechanism Mouse
model
Results References Clinical trials
Yes/No
Estrogen Anti-oxidant/protective Ts65Dn Protection/rescue [22, 114] No
Donepezil AChE inhibitor Ts65Dn No beneficial effects [200] FDA approved
Memantine NMDA reverse agonist Ts65Dn Symptomatic [54, 201, 202] Ongoing
Vitamin E Anti-oxidant Ts65Dn Preventive [70] Yes [178, 183, 203]
Fluoxetine Serotonin reuptake inhibitor Ts65Dn Increased neurogenesis
Normalize GABA release
[152, 153, 204] No formal trial
GABA α5-selective inverse agonists GABA inhibition Ts65Dn Rescue learning and memory 115 Ongoing
GABA(B) receptor antagonist CGP55845 GABA inhibition T65Dn Restored spatial memory and novel object recognition [197]
pentylenetetrazole (PTZ) GABA(A) antagonist Ts65Dn rescued TS performance in the Morris water maze [200]
L-DOPS NE precursor Ts65Dn Rescue AD pathology and memory [136, 137] No
epigallocatechin-gallate (EGCG) DYRK1A inhibitor Ts65Dn Rescues cognitive deficits [205] Pilot studies

10. CONCLUSION

This review demonstrates the need for comparative aging studies in several newly developed mouse models of DS, to determine which gene segments on Mmu10, 17 and 16 are involved in the neuropathology and memory loss observed in DS with aging. Development of more appropriate models will allow better pre-clinical studies of drug interventions as well as validation of studies performed in existing models in the past. This review also demonstrates the need for identifying in vivo neuroimaging biomarkers for the different neuropathological processes in DS especially as a function of aging, and mouse studies are crucial for validating neuroprotective interventions. A wealth of previous findings suggest that inflammation, oxidative stress, amyloid and tau toxicity, as well as an imbalance in neurotrophic factor maturation, transport, and cleavage are involved in the pathogenesis and dementia observed in DS-AD.

Supplementary Material

Ann Charlotte Granholm Bentley Profile Picture

Acknowledgments

Thanks are due to the many contributors to our previous work, including Mona Buhusi, Ashley Fortress, Chris Hunter, Jason Lockrow, Alfred Moore, Claudia Umphlet, and Linda Crnic. ACG would like to acknowledge funding from Sie Foundation, National Institutes on Aging (AG012122), and the Alzheimer’s Association. DP would like to acknowledge the support of the Lowe Fund of the Denver Foundation and the Alvin Itkin Foundation. RAN acknowledges the support of the National Institutes on Aging (P01 AG017617).

LIST OF ABBREVIATIONS

AD

Alzheimer’s disease

APP

Amyloid precursor protein

ChAT

Choline acetyltransferase

D//

Axial diffusivity

D

Radial diffusivity

DKI

Diffusional kurtosis imaging

dMRI

Diffusion MRI

DS

Down syndrome

DTI

Diffusion tensor imaging

F

Female

HDB

Horizontal limb of diagonal band

Hsa

Human chromosome

K//

Axial kurtosis

K

Radial kurtosis

L-DOPS

L-threo-dihydroxyphenylserine

M

Male

MD

Mean diffusivity

MK

Mean kurtosis

Mmu

Murine chromosome

MRI

Magnetic resonance imaging

MRS

Magnetic resonance spectroscopy

MS

Medial septum

Mx1

Myxovirus (Influenza) resistance 1

NS

Normosomic mice born to Ts65Dn dams

p75NTR

Protein 75 kilodaltons neurotrophic receptor

sc

Subcutaneous

SOD1

Superoxide dismutase 1

TrkA

Tyrosine kinase receptor A

TS

Trisomic

Tx

Treatment

VDB

Vertical limb of diagonal band

WRAM

Water radial arm maze

Biography

graphic file with name nihms816589b1.gif

Ann-C.E. Granholm

Footnotes

CONFLICT OF INTEREST

The author(s) confirm that this article content has no conflicts of interest.

References

  • 1.Hassold T, Sherman S. Down syndrome: genetic recombination and the origin of the extra chromosome 21. Clin Genet. 2000;57(2):95–100. doi: 10.1034/j.1399-0004.2000.570201.x. [DOI] [PubMed] [Google Scholar]
  • 2.Presson AP, Partyka G, Jensen KM, Devine OJ, Rasmussen SA, McCabe LL, et al. Current estimate of Down Syndrome population prevalence in the United States. J Pediatr. 2013;163(4):1163–8. doi: 10.1016/j.jpeds.2013.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lott IT, Dierssen M. Cognitive deficits and associated neurological complications in individuals with Down’s syndrome. Lancet Neurol. 2010;9(6):623–33. doi: 10.1016/S1474-4422(10)70112-5. [DOI] [PubMed] [Google Scholar]
  • 4.Head E, Silverman W, Patterson D, Lott IT. Aging and down syndrome. Curr Gerontol Geriatr Res. 2012;2012:412536. doi: 10.1155/2012/412536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hartley D, Blumenthal T, Carrillo M, DiPaolo G, Esralew L, Gardiner K, et al. Down syndrome and Alzheimer’s disease: Common pathways, common goals. Alzheimers Dement. 2015;11(6):700–9. doi: 10.1016/j.jalz.2014.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Lai F, Williams RS. A prospective study of Alzheimer disease in Down syndrome. Arch Neurol. 1989;46(8):849–53. doi: 10.1001/archneur.1989.00520440031017. [DOI] [PubMed] [Google Scholar]
  • 7.Wisniewski KE, Dalton AJ, McLachlan C, Wen GY, Wisniewski HM. Alzheimer’s disease in Down’s syndrome: clinicopathologic studies. Neurology. 1985;35(7):957–61. doi: 10.1212/wnl.35.7.957. [DOI] [PubMed] [Google Scholar]
  • 8.Lai F. Clinicopathologic features of Alzheimer disease in Down syndrome. Prog Clin Biol Res. 1992;379:15–34. [PubMed] [Google Scholar]
  • 9.Menendez M. Down syndrome, Alzheimer’s disease and seizures. Brain Dev. 2005;27(4):246–52. doi: 10.1016/j.braindev.2004.07.008. [DOI] [PubMed] [Google Scholar]
  • 10.Rubinsztein DC, Hon J, Stevens F, Pyrah I, Tysoe C, Huppert FA, et al. Apo E genotypes and risk of dementia in Down syndrome. Am J Med Genet. 1999;88(4):344–347. doi: 10.1002/(sici)1096-8628(19990820)88:4<344::aid-ajmg10>3.0.co;2-1. [DOI] [PubMed] [Google Scholar]
  • 11.Schliebs R, Arendt T. The cholinergic system in aging and neuronal degeneration. Behav Brain Res. 2010;221(2):555–63. doi: 10.1016/j.bbr.2010.11.058. [DOI] [PubMed] [Google Scholar]
  • 12.Sanchez MM, Moghadam S, Naik P, Martin KJ, Salehi A. Hippocampal network alterations in Alzheimer’s disease and Down syndrome: from structure to therapy. J Alzheimers Dis. 2011;26(3):29–47. doi: 10.3233/JAD-2011-0050. [DOI] [PubMed] [Google Scholar]
  • 13.Ferrer I, Gullotta F. Down’s syndrome and Alzheimer’s disease: dendritic spine counts in the hippocampus. Acta Neuropathol. 1990;79(6):680–5. doi: 10.1007/BF00294247. [DOI] [PubMed] [Google Scholar]
  • 14.Iulita MF, Cuello AC. Nerve growth factor metabolic dysfunction in Down’s syndrome brains. Brain. 2014;137(3):860–72. doi: 10.1093/brain/awt372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.García-Cerro S, Martínez P, Vidal V, Corrales A, Flórez J, Vidal R, et al. Overexpression of Dyrk1A is implicated in several cognitive, electrophysiological and neuromorphological alterations found in a mouse model of Down syndrome. PLoS One. 2014;9(9):e106572. doi: 10.1371/journal.pone.0106572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Cataldo AM, Petanceska S, Terio NB, Peterhoff CM, Durham R, Mercken M, et al. Abeta localization in abnormal endosomes: association with earliest Abeta elevations in AD and Down syndrome. Neurobiol Aging. 2004;25(10):1263–72. doi: 10.1016/j.neurobiolaging.2004.02.027. [DOI] [PubMed] [Google Scholar]
  • 17.Wilcock DM. Neuroinflammation in the aging down syndrome brain: lessons from Alzheimer’s disease. Curr Gerontol Geriatr Res. 2012;2012:170276. doi: 10.1155/2012/170276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lott IT, Head E, Doran E, Busciglio J. Beta-amyloid, oxidative stress and down syndrome. Curr Alzheimer Res. 2006;3(5):521–8. doi: 10.2174/156720506779025305. [DOI] [PubMed] [Google Scholar]
  • 19.Mann DM, Esiri MM. The pattern of acquisition of plaques and tangles in the brains of patients under 50 years of age with Down’s syndrome. J Neurol Sci. 1989;89(2–3):169–79. doi: 10.1016/0022-510x(89)90019-1. [DOI] [PubMed] [Google Scholar]
  • 20.Lemere CA, Blusztajn JK, Yamaguchi H, Wisniewski T, Saido TC, Selkoe DJ. Sequence of deposition of heterogeneous amyloid beta-peptides and APO E in Down syndrome: implications for initial events in amyloid plaque formation. Neurobiol Dis. 1996;3(1):16–32. doi: 10.1006/nbdi.1996.0003. [DOI] [PubMed] [Google Scholar]
  • 21.Head E, Powell D, Gold BT, Schmitt FA. Alzheimer’s Disease in Down Syndrome. Eur J Neurodegener Dis. 2012;1(3):353–364. [PMC free article] [PubMed] [Google Scholar]
  • 22.Hunter CL, Bimonte-Nelson HA, Nelson M, Eckman CB, Granholm AC. Behavioral and neurobiological markers of Alzheimer’s disease in Ts65Dn mice: effects of estrogen. Neurobiol Aging. 2004;25(7):873–84. doi: 10.1016/j.neurobiolaging.2003.10.010. [DOI] [PubMed] [Google Scholar]
  • 23.Hunter CL, Bimonte-Nelson HA, Granholm AC. Behavioral comparison of 4 and 6 month-old Ts65Dn mice: age-related impairments in working and reference memory. Behav Brain Res. 2003;138(2):121–31. doi: 10.1016/s0166-4328(02)00275-9. [DOI] [PubMed] [Google Scholar]
  • 24.Choi JH, Berger JD, Mazzella MJ, Morales-Corraliza J, Cataldo AM, Nixon RA, et al. Age-dependent dysregulation of brain amyloid precursor protein in the Ts65Dn Down syndrome mouse model. J Neurochem. 2009;110(6):1818–27. doi: 10.1111/j.1471-4159.2009.06277.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Cataldo AM, Petarceska S, Peterhoff CM, Terio NB, Epsteir CJ, Villar A, et al. App gene dosage modulates endosomal abnormalities of Alzheimer’s disease in a segmental trisomy 16 mouse model of down syndrome. J Neurosci. 2003;23(17):6788–92. doi: 10.1523/JNEUROSCI.23-17-06788.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lomoio S, Scherini E, Necchi D. Beta-amyloid overload does not directly correlate with SAPK/JNK activation and tau protein phosphorylation in the cerebellar cortex of Ts65Dn mice. Brain Res. 2009;1297:198–206. doi: 10.1016/j.brainres.2009.08.052. [DOI] [PubMed] [Google Scholar]
  • 27.Cárdenas AM, Ardiles AO, Barraza N, Baéz-Matus X, Caviedes P. Role of tau protein in neuronal damage in Alzheimer’s disease and Down syndrome. Arch Med Res. 2012;43(8):645–54. doi: 10.1016/j.arcmed.2012.10.012. [DOI] [PubMed] [Google Scholar]
  • 28.Hof PR, Bouras C, Perl DP, Sparks DL, Mehta N, Morrison JH. Age-related distribution of neuropathologic changes in the cerebral cortex of patients with Down’s syndrome. Quantitative regional analysis and comparison with Alzheimer’s disease. Arch Neurol. 1995;52(4):379–91. doi: 10.1001/archneur.1995.00540280065020. [DOI] [PubMed] [Google Scholar]
  • 29.Mann DM, Yates PO, Marcyniuk B. Alzheimer’s presenile dementia, senile dementia of Alzheimer type and Down’s syndrome in middle age form an age related continuum of pathological changes. Neuropathol Appl Neurobiol. 1984;10(3):185–207. doi: 10.1111/j.1365-2990.1984.tb00351.x. [DOI] [PubMed] [Google Scholar]
  • 30.Mann DM, Yates PO, Marcyniuk B, Ravindra CR. The topography of plaques and tangles in Down’s syndrome patients of different ages. Neuropathol Appl Neurobiol. 1986;12(5):447–57. doi: 10.1111/j.1365-2990.1986.tb00053.x. [DOI] [PubMed] [Google Scholar]
  • 31.Wisniewski KE, Wisniewski HM, Wen GY. Occurrence of neuropathological changes and dementia of Alzheimer’s disease in Down’s syndrome. Ann Neurol. 1985;17(3):278–82. doi: 10.1002/ana.410170310. [DOI] [PubMed] [Google Scholar]
  • 32.Wisniewski HM, Rabe A. Discrepancy between Alzheimer-type neuropathology and dementia in persons with Down’s syndrome. Ann N Y Acad Sci. 1986;477:247–60. doi: 10.1111/j.1749-6632.1986.tb40344.x. [DOI] [PubMed] [Google Scholar]
  • 33.Schupf N, Sergievsky GH. Genetic and host factors for dementia in Down’s syndrome. Br J Psychiatry. 2002;180:405–10. doi: 10.1192/bjp.180.5.405. [DOI] [PubMed] [Google Scholar]
  • 34.Sturgeon X, Gardiner KJ. Transcript catalogs of human chromosome 21 and orthologous chimpanzee and mouse regions. Mamm Genom. 2011;22(5–6):261–71. doi: 10.1007/s00335-011-9321-y. [DOI] [PubMed] [Google Scholar]
  • 35.Busciglio J, Capone G, O’Bryan J, Gardiner KJ. Down syndrome: genes, model systems, and progress towards pharmacotherapies and clinical trials for cognitive deficits. Cytogenet Genom Res. 2013;141(4):260–71. doi: 10.1159/000354306. [DOI] [PubMed] [Google Scholar]
  • 36.Gardiner K, Fortna A, Bechtel L, Davisson MT. Mouse models of Down syndrome: how useful can they be? Comparison of the gene content of human chromosome 21 with orthologous mouse genomic regions. Gene. 2003;318:137–47. doi: 10.1016/s0378-1119(03)00769-8. [DOI] [PubMed] [Google Scholar]
  • 37.Reeves RH, Irving NG, Moran TH, Wohn A, Kitt C, Sisodia SS, et al. A mouse model for Down syndrome exhibits learning and behaviour deficits. Nat Genet. 1995;11(2):177–84. doi: 10.1038/ng1095-177. [DOI] [PubMed] [Google Scholar]
  • 38.Cooper JD, Salehi A, Delcroix JD, Howe CL, Belichenko PV, Chua-Couzens J, et al. Failed retrograde transport of NGF in a mouse model of Down’s syndrome: reversal of cholinergic neurodegenerative phenotypes following NGF infusion. Proc Natl Acad Sci U S A. 2001;98(18):10439–44. doi: 10.1073/pnas.181219298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Granholm AC, Sanders LA, Crnic LS. Loss of cholinergic phenotype in basal forebrain coincides with cognitive decline in a mouse model of Down’s syndrome. Exp Neurol. 2000;161(2):647–63. doi: 10.1006/exnr.1999.7289. [DOI] [PubMed] [Google Scholar]
  • 40.Holtzman DM, Santucci D, Kilbridge J, Chua-Couzens J, Fontana DJ, Daniels SE, et al. Developmental abnormalities and age-related neurodegeneration in a mouse model of Down syndrome. Proc Natl Acad Sci U S A. 1996;93(23):13333–8. doi: 10.1073/pnas.93.23.13333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Insausti AM, Megías M, Crespo D, Cruz-Orive LM, Dierssen M, Vallina IF, et al. Hippocampal volume and neuronal number in Ts65Dn mice: a murine model of Down syndrome. Neurosci Lett. 1998;253(3):175–8. doi: 10.1016/s0304-3940(98)00641-7. [DOI] [PubMed] [Google Scholar]
  • 42.Siarey RJ, Stoll J, Rapoport SI, Galdzicki Z. Altered long-term potentiation in the young and old Ts65Dn mouse, a model for Down Syndrome. Neuropharmacology. 1997;36(11–12):1549–54. doi: 10.1016/s0028-3908(97)00157-3. [DOI] [PubMed] [Google Scholar]
  • 43.Hunter CL, Isacson O, Nelson M, Bimonte-Nelson H, Seo H, Lin L, et al. Regional alterations in amyloid precursor protein and nerve growth factor across age in a mouse model of Down’s syndrome. Neurosci Res. 2003;45(4):437–45. doi: 10.1016/s0168-0102(03)00005-1. [DOI] [PubMed] [Google Scholar]
  • 44.Seo H, Isacson O. Abnormal APP, cholinergic and cognitive function in Ts65Dn Down’s model mice. Exp Neurol. 2005;193(2):469–80. doi: 10.1016/j.expneurol.2004.11.017. [DOI] [PubMed] [Google Scholar]
  • 45.Haydar TF, Blue ME, Molliver ME, Krueger BK, Yarowsky PJ. Consequences of trisomy 16 for mouse brain development: corticogenesis in a model of Down syndrome. J Neurosci. 1996;16(19):6175–82. doi: 10.1523/JNEUROSCI.16-19-06175.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Bambrick LL, Krueger BK. Neuronal apoptosis in mouse trisomy 16: mediation by caspases. J Neurochem. 1999;72(4):1769–72. doi: 10.1046/j.1471-4159.1999.721769.x. [DOI] [PubMed] [Google Scholar]
  • 47.Davisson MT, Schmidt C, Akeson EC. Segmental trisomy of murine chromosome 16: a new model system for studying Down syndrome. Prog Clin Biol Res. 1990;360:263–80. [PubMed] [Google Scholar]
  • 48.Duchon A, Raveau M, Chevalier C, Nalesso V, Sharp AJ, Herault Y. Identification of the translocation breakpoints in the Ts65Dn and Ts1Cje mouse lines: relevance for modeling Down syndrome. Mamm Genome. 2011;22(11–12):674–84. doi: 10.1007/s00335-011-9356-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sanders NC, Williams DK, Wenger GR. Does the learning deficit observed under an incremental repeated acquisition schedule of reinforcement in Ts65Dn mice, a model for Down syndrome, change as they age? Behav Brain Res. 2009;203(1):137–42. doi: 10.1016/j.bbr.2009.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ishihara K, Amano K, Takaki E, Ebrahim AS, Shimohata A, Shibazaki N, et al. Increased lipid peroxidation in Down’s syndrome mouse models. J Neurochem. 2009;110(6):1965–76. doi: 10.1111/j.1471-4159.2009.06294.x. [DOI] [PubMed] [Google Scholar]
  • 51.Shukkur EA, Shimohata A, Akagi T, Yu W, Yamaguchi M, Murayama M, et al. Mitochondrial dysfunction and tau hyperphosphorylation in Ts1Cje, a mouse model for Down syndrome. Hum Mol Genet. 2006;15(18):2752–62. doi: 10.1093/hmg/ddl211. [DOI] [PubMed] [Google Scholar]
  • 52.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(18):5938–48. doi: 10.1523/JNEUROSCI.1547-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sago H, Carlson EJ, Smith DJ, Rubin EM, Crnic LS, Huang TT, et al. Genetic dissection of region associated with behavioral abnormalities in mouse models for Down syndrome. Pediatr Res. 2000;48(5):606–13. doi: 10.1203/00006450-200011000-00009. [DOI] [PubMed] [Google Scholar]
  • 54.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. 2010;221(2):610–22. doi: 10.1016/j.bbr.2010.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Salehi A, Delcroix JD, Belichenko PV, Zhan K, Wu C, Valletta JS, et al. Increased App expression in a mouse model of Down’s syndrome disrupts NGF transport and causes cholinergic neuron degeneration. Neuron. 2006;51(1):29–42. doi: 10.1016/j.neuron.2006.05.022. [DOI] [PubMed] [Google Scholar]
  • 56.Cataldo AM, Mathews PM, Boiteau AB, Hassinger LC, Peterhoff CM, Jiang Y, et al. Down syndrome fibroblast model of Alzheimer-related endosome pathology: accelerated endocytosis promotes late endocytic defects. Am J Pathol. 2008;173(2):370–84. doi: 10.2353/ajpath.2008.071053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Cataldo AM, Peterhoff CM, Troncoso JC, Gomez-Isla T, Hyman BT, Nixon RA. Endocytic pathway abnormalities precede amyloid beta deposition in sporadic Alzheimer’s disease and Down syndrome: differential effects of APOE genotype and presenilin mutations. Am J Pathol. 2000;157(1):277–86. doi: 10.1016/s0002-9440(10)64538-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Levine S, Saltzman A, Levy E, Ginsberg SD. Systemic pathology in aged mouse models of Down’s syndrome and Alzheimer’s disease. Exp Mol Pathol. 2009;86(1):18–22. doi: 10.1016/j.yexmp.2008.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Villar AJ, Belichenko PV, Gillespie AM, Kozy HM, Mobley WC, Epstein CJ. Identification and characterization of a new Down syndrome model, Ts[Rb(12.1716)]2Cje, resulting from a spontaneous Robertsonian fusion between T(171)65Dn and mouse chromosome 12. Mamm Genom. 2005;16(2):79–90. doi: 10.1007/s00335-004-2428-7. [DOI] [PubMed] [Google Scholar]
  • 60.Yu T, Li Z, Jia Z, Clapcote SJ, Liu C, Li S, et al. A mouse model of Down syndrome trisomic for all human chromosome 21 syntenic regions. Hum Mol Genet. 2010;19(14):2780–91. doi: 10.1093/hmg/ddq179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Zhang L, Meng K, Jiang X, Liu C, Pao A, Belichenko PV, et al. Human chromosome 21 orthologous region on mouse chromosome 17 is a major determinant of Down syndrome-related developmental cognitive deficits. Hum Mol Genet. 2014;23(3):578–89. doi: 10.1093/hmg/ddt446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ahmed MM, Dhanasekaran AR, Tong S, Wiseman FK, Fisher EM, Tybulewicz VL, et al. Protein profiles in Tc1 mice implicate novel pathway perturbations in the Down syndrome brain. Hum Mol Genet. 2013;22(9):1709–24. doi: 10.1093/hmg/ddt017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Gribble SM, Wiseman FK, Clayton S, Prigmore E, Langley E, Yang F, et al. Massively parallel sequencing reveals the complex structure of an irradiated human chromosome on a mouse background in the Tc1 model of Down syndrome. PLoS One. 2013;8(4):e60482. doi: 10.1371/journal.pone.0060482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Singh P, Schimenti JC, Bolcun-Filas E. A mouse geneticist’s practical guide to CRISPR applications. Genetics. 2015;199(1):1–15. doi: 10.1534/genetics.114.169771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Driscoll LL, Carroll JC, Moon J, Crnic LS, Levitsky DA, Strupp BJ. Impaired sustained attention and error-induced stereotypy in the aged Ts65Dn mouse: a mouse model of Down syndrome and Alzheimer’s disease. Behav Neurosci. 2004;118(6):1196–205. doi: 10.1037/0735-7044.118.6.1196. [DOI] [PubMed] [Google Scholar]
  • 66.Hyde LA, Crnic LS. Age-related deficits in context discrimination learning in Ts65Dn mice that model Down syndrome and Alzheimer’s disease. Behav Neurosci. 2001;115(6):1239–46. [PubMed] [Google Scholar]
  • 67.Demas GE, Nelson RJ, Krueger BK, Yarowsky PJ. Spatial memory deficits in segmental trisomic Ts65Dn mice. Behav Brain Res. 1996;82(1):85–92. doi: 10.1016/s0166-4328(97)81111-4. [DOI] [PubMed] [Google Scholar]
  • 68.Bimonte-Nelson HA, Hunter CL, Nelson ME, Granholm AC. Frontal cortex BDNF levels correlate with working memory in an animal model of Down syndrome. Behav Brain Res. 2003;139(1–2):47–57. doi: 10.1016/s0166-4328(02)00082-7. [DOI] [PubMed] [Google Scholar]
  • 69.Demas GE, Nelson RJ, Krueger BK, Yarowsky PJ. Impaired spatial working and reference memory in segmental trisomy (Ts65Dn) mice. Behav Brain Res. 1998;90(2):199–201. doi: 10.1016/s0166-4328(97)00116-2. [DOI] [PubMed] [Google Scholar]
  • 70.Lockrow J, Prakasam A, Huang P, Bimonte-Nelson H, Sambamurti K, Granholm AC. Cholinergic degeneration and memory loss delayed by vitamin E in a Down syndrome mouse model. Exp Neurol. 2009;216(2):278–89. doi: 10.1016/j.expneurol.2008.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Fernandez F, Garner CC. Episodic-like memory in Ts65Dn, a mouse model of Down syndrome. Behav Brain Res. 2008;188(1):233–7. doi: 10.1016/j.bbr.2007.09.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Costa AC, Stasko MR, Schmidt C, Davisson MT. Behavioral validation of the Ts65Dn mouse model for Down syndrome of a genetic background free of the retinal degeneration mutation Pde6b(rd1) Behav Brain Res. 2010;206(1):52–62. doi: 10.1016/j.bbr.2009.08.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Stasko MR, Costa AC. Experimental parameters affecting the Morris water maze performance of a mouse model of Down syndrome. Behav Brain Res. 2004;154(1):1–17. doi: 10.1016/j.bbr.2004.01.012. [DOI] [PubMed] [Google Scholar]
  • 74.Escorihuela RM, Vallina IF, Martínez-Cué C, Baamonde C, Dierssen M, Tobeña A, et al. Impaired short- and long-term memory in Ts65Dn mice, a model for Down syndrome. Neurosci Lett. 1998;247(2–3):171–4. doi: 10.1016/s0304-3940(98)00317-6. [DOI] [PubMed] [Google Scholar]
  • 75.Whitney KN, Wenger GR. Impulsivity and motor activity in aged, male Ts65Dn mice. Exp Clin Psychopharmacol. 2013;21(5):345–54. doi: 10.1037/a0033965. [DOI] [PubMed] [Google Scholar]
  • 76.Sabbagh MN, Chen K, Rogers J, Fleisher AS, Liebsack C, Bandy D, et al. Florbetapir PET, FDG PET, and MRI in Down syndrome individuals with and without Alzheimer’s dementia. Alzheimers Dement. 2015:84–9. doi: 10.1016/j.jalz.2015.01.006. pii: S1552-5260(15) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Handen BL, Cohen AD, Channamalappa U, Bulova P, Cannon SA, Cohen WI, et al. Imaging brain amyloid in nondemented young adults with Down syndrome using Pittsburgh compound B. Alzheimers Dement. 2012;8(6):496–501. doi: 10.1016/j.jalz.2011.09.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Carducci F, Onorati P, Condoluci C, Di Gennaro G, Quarato PP, Pierallini A, et al. Whole-brain voxel-based morphometry study of children and adolescents with Down syndrome. Funct Neurol. 2013;28(1):19–28. [PMC free article] [PubMed] [Google Scholar]
  • 79.d’Abrera JC, Holland AJ, Landt J, Stocks-Gee G, Zaman SH. A neuroimaging proof of principle study of Down’s syndrome and dementia: ethical and methodological challenges in intrusive research. J Intellect Disabil Res. 2013;57(2):105–118. doi: 10.1111/j.1365-2788.2011.01495.x. [DOI] [PubMed] [Google Scholar]
  • 80.Jacola LM, Byars AW, Hickey F, Vannest J, Holland SK, Schapiro MB. Functional magnetic resonance imaging of story listening in adolescents and young adults with Down syndrome: evidence for atypical neurodevelopment. J Intellect Disabil Res. 2014;58(10):892–902. doi: 10.1111/jir.12089. [DOI] [PubMed] [Google Scholar]
  • 81.Hartley SL, Handen BL, Devenny DA, Hardison R, Mihaila I, Price JC, et al. Cognitive functioning in relation to brain amyloid-β in healthy adults with Down syndrome. Brain. 2014;137(Pt 9):2556–63. doi: 10.1093/brain/awu173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Imai M, Watanabe H, Yasui K, Kimura Y, Shitara Y, Tsuchida S, et al. Functional connectivity of the cortex of term and preterm infants and infants with Down’s syndrome. Neuroimage. 2014;85:272–278. doi: 10.1016/j.neuroimage.2013.04.080. [DOI] [PubMed] [Google Scholar]
  • 83.Powell D, Caban-Holt A, Jicha G, Robertson W, Davis R, Gold BT, et al. Frontal white matter integrity in adults with Down syndrome with and without dementia. Neurobiol Aging. 2014;35(7):1562–9. doi: 10.1016/j.neurobiolaging.2014.01.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Vega JN, Hohman TJ, Pryweller JR, Dykens EM, Thornton-Wells TA. Resting-state functional connectivity in individuals with down syndrome and williams syndrome compared with typically developing controls. Brain Connect. 2015 doi: 10.1089/brain.2014.0266. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chen Y, Dyakin VV, Branch CA, Ardekani B, Yang D, Guilfoyle DN, et al. In vivo MRI identifies cholinergic circuitry deficits in a Down syndrome model. Neurobiol Aging. 2009;30(9):1453–65. doi: 10.1016/j.neurobiolaging.2007.11.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ishihara K, Amano K, Takaki E, Shimohata A, Sago H, Epstein CJ, et al. Enlarged brain ventricles and impaired neurogenesis in the Ts1Cje and Ts2Cje mouse models of Down syndrome. Cereb Cortex. 2010;20(5):1131–43. doi: 10.1093/cercor/bhp176. [DOI] [PubMed] [Google Scholar]
  • 87.Santin MD, Valabrègue R, Rivals I, Pénager R, Paquin R, Dauphinot L, et al. In vivo 1H MRS study in microlitre voxels in the hippocampus of a mouse model of Down syndrome at 11.7 T. NMR Biomed. 2014;27(10):1143–50. doi: 10.1002/nbm.3155. [DOI] [PubMed] [Google Scholar]
  • 88.Kaur G, Sharma A, Xu W, Gerum S, Alldred MJ, Subbanna S, et al. Glutamatergic transmission aberration: a major cause of behavioral deficits in a murine model of Down’s syndrome. J Neurosci. 2014;34(15):5099–106. doi: 10.1523/JNEUROSCI.5338-13.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Nie X, Hamlett ED, Granholm AC, Hui ES, Helpern JA, Jensen JH, et al. Evidence of altered age-related brain cytoarchitecture in mouse models of down syndrome: a diffusional kurtosis imaging study. Magn Reson Imaging. 2014;33(4):437–47. doi: 10.1016/j.mri.2014.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Jensen JH, Helpern JA, Ramani A, Lu H, Kaczynski K. Diffusional kurtosis imaging: the quantification of non-gaussian water diffusion by means of magnetic resonance imaging. Magn Reson Med. 2005;53(6):1432–40. doi: 10.1002/mrm.20508. [DOI] [PubMed] [Google Scholar]
  • 91.Lu H, Jensen JH, Ramani A, Helpern JA. Three-dimensional characterization of non-gaussian water diffusion in humans using diffusion kurtosis imaging. NMR Biomed. 2006;19(2):236–47. doi: 10.1002/nbm.1020. [DOI] [PubMed] [Google Scholar]
  • 92.Jensen JH, Helpern JA. MRI quantification of non-Gaussian water diffusion by kurtosis analysis. NMR Biomed. 2010;23(7):698–710. doi: 10.1002/nbm.1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Cheung MM, Hui ES, Chan KC, Helpern JA, Qi L, Wu EX. Does diffusion kurtosis imaging lead to better neural tissue characterization? A rodent brain maturation study. Neuroimage. 2009;45(2):386–92. doi: 10.1016/j.neuroimage.2008.12.018. [DOI] [PubMed] [Google Scholar]
  • 94.Vanhoutte G, Pereson S, Delgado Y, Palacios R, Guns PJ, Asselbergh B, et al. Diffusion kurtosis imaging to detect amyloidosis in an APP/PS1 mouse model for Alzheimer’s disease. Magn Reson Med. 2013;69(4):1115–21. doi: 10.1002/mrm.24680. [DOI] [PubMed] [Google Scholar]
  • 95.Grinberg F, Farrher E, Ciobanu L, Geffroy F, Le Bihan D, Shah NJ. Non-Gaussian diffusion imaging for enhanced contrast of brain tissue affected by ischemic stroke. PLoS One. 2014;9(2):e89225. doi: 10.1371/journal.pone.0089225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Weber RA, Hui ES, Jensen JH, Nie X, Falangola MF, Helpern JA, et al. Diffusional kurtosis and diffusion tensor imaging reveal different time-sensitive stroke-induced microstructural changes. Stroke. 2015;46(2):545–50. doi: 10.1161/STROKEAHA.114.006782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Falangola MF, Guilfoyle DN, Tabesh A, Hui ES, Nie X, Jensen JH, et al. Histological correlation of diffusional kurtosis and white matter modeling metrics in cuprizone-induced corpus callosum demyelination. NMR Biomed. 2014;27(8):948–57. doi: 10.1002/nbm.3140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Belichenko PV, Masliah E, Kleschevnikov AM, Villar AJ, Epstein CJ, Salehi A, et al. Synaptic structural abnormalities in the Ts65Dn mouse model of Down Syndrome. J Comp Neurol. 2004;480(3):281–98. doi: 10.1002/cne.20337. [DOI] [PubMed] [Google Scholar]
  • 99.Popov VI, Kleschevnikov AM, Klimenko OA, Stewart MG, Belichenko PV. Three-dimensional synaptic ultrastructure in the dentate gyrus and hippocampal area CA3 in the Ts65Dn mouse model of Down syndrome. J Comp Neurol. 2011;519(7):1338–54. doi: 10.1002/cne.22573. [DOI] [PubMed] [Google Scholar]
  • 100.Ábrahám H, Vincze A, Veszprémi B, Kravják A, Gömöri É, Kovács GG, et al. Impaired myelination of the human hippocampal formation in Down syndrome. Int J Dev Neurosci. 2012;30(2):147–58. doi: 10.1016/j.ijdevneu.2011.11.005. [DOI] [PubMed] [Google Scholar]
  • 101.Wisniewski KE, Schmidt-Sidor B. Postnatal delay of myelin formation in brains from Down syndrome infants and children. Clin Neuropathol. 1989;8:55–62. [PubMed] [Google Scholar]
  • 102.Gardiner KJ. Pharmacological approaches to improving cognitive function in Down syndrome: current status and considerations. Drug Des Develop Ther. 2015;9:103–125. doi: 10.2147/DDDT.S51476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lockrow JP, Fortress AM, Granholm AC. Age-related neurodegeneration and memory loss in down syndrome. Curr Gerontol Geriatr Res. 2012;2012:463909. doi: 10.1155/2012/463909. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.McGeer PL. The 12th J. A. F. Stevenson memorial lecture. Aging, Alzheimer’s disease, and the cholinergic system. Can J Physiol Pharmacol. 1984;62(7):741–54. doi: 10.1139/y84-123. [DOI] [PubMed] [Google Scholar]
  • 105.Perry E, Walker M, Grace J, Perry R. Acetylcholine in mind: a neurotransmitter correlate of consciousness? Trends Neurosci. 1999;22(6):273–80. doi: 10.1016/s0166-2236(98)01361-7. [DOI] [PubMed] [Google Scholar]
  • 106.Dunnett SB, Everitt BJ, Robbins TW. The basal forebrain-cortical cholinergic system: interpreting the functional consequences of excitotoxic lesions. Trends Neurosci. 1991;14(11):494–501. doi: 10.1016/0166-2236(91)90061-x. [DOI] [PubMed] [Google Scholar]
  • 107.Ikonomovic MD, Mufson EJ, Wuu J, Cochran EJ, Bennett DA, DeKosky ST. Cholinergic plasticity in hippocampus of individuals with mild cognitive impairment: correlation with Alzheimer’s neuropathology. J Alzheimers Dis. 2003;5(1):39–48. doi: 10.3233/jad-2003-5106. [DOI] [PubMed] [Google Scholar]
  • 108.Isacson O, Seo H, Lin L, Albeck D, Granholm AC. Alzheimer’s disease and Down’s syndrome: roles of APP, trophic factors and ACh. Trends Neurosci. 2002;25(2):79–84. doi: 10.1016/s0166-2236(02)02037-4. [DOI] [PubMed] [Google Scholar]
  • 109.Yates CM, Simpson J, Gordon A, Maloney AF, Allison Y, Ritchie IM, et al. Catecholamines and cholinergic enzymes in pre-senile and senile Alzheimer-type dementia and Down’s syndrome. Brain Res. 1983;280(1):119–26. doi: 10.1016/0006-8993(83)91179-4. [DOI] [PubMed] [Google Scholar]
  • 110.Mann DM, Yates PO, Marcyniuk B, Ravindra CR. Pathological evidence for neurotransmitter deficits in Down’s syndrome of middle age. J Ment Defic Res. 1985;29(Pt 2):125–35. doi: 10.1111/j.1365-2788.1985.tb00320.x. [DOI] [PubMed] [Google Scholar]
  • 111.Contestabile A, Fila T, Bartesaghi R, Contestabile A, Ciani E. Choline acetyltransferase activity at different ages in brain of Ts65Dn mice, an animal model for Down’s syndrome and related neurodegenerative diseases. J Neurochem. 2006;97(2):515–26. doi: 10.1111/j.1471-4159.2006.03769.x. [DOI] [PubMed] [Google Scholar]
  • 112.Kelley CM, Powers BE, Velazquez R, Ash JA, Ginsberg SD, Strupp BJ, et al. Sex differences in the cholinergic basal forebrain in the Ts65Dn mouse model of Down syndrome and Alzheimer’s disease. Brain Pathol. 2014;24(1):33–44. doi: 10.1111/bpa.12073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Coppus AM, Evenhuis HM, Verberne GJ, Visser FE, Eikelenboom P, van Gool WA, et al. Early age at menopause is associated with increased risk of dementia and mortality in women with Down syndrome. J Alzheimers Dis. 2010;19(2):545–50. doi: 10.3233/JAD-2010-1247. [DOI] [PubMed] [Google Scholar]
  • 114.Granholm AC, Ford KA, Hyde LA, Bimonte HA, Hunter CL, Nelson M, et al. Estrogen restores cognition and cholinergic phenotype in an animal model of Down syndrome. Physiol Behav. 2002;77(2–3):371–85. doi: 10.1016/s0031-9384(02)00884-3. [DOI] [PubMed] [Google Scholar]
  • 115.Lally G, Faull RL, Waldvogel HJ, Ferrari S, Emson PC. Calcium homeostasis in ageing: studies on the calcium binding protein calbindin D28K. J Neural Transm. 1997;104(10):1107–12. doi: 10.1007/BF01273323. [DOI] [PubMed] [Google Scholar]
  • 116.Sutherland MK, Wong L, Somerville MJ, Yoong LK, Bergeron C, Parmentier M, et al. Reduction of calbindin-28k mRNA levels in Alzheimer as compared to Huntington hippocampus. Brain Res Mol Brain Res. 1993;18(1–2):32–42. doi: 10.1016/0169-328x(93)90171-k. [DOI] [PubMed] [Google Scholar]
  • 117.Pappas IS, Parnavelas JG. Neurotrophins and basic fibroblast growth factor induce the differentiation of calbindin-containing neurons in the cerebral cortex. Exp Neurol. 1997;144(2):302–14. doi: 10.1006/exnr.1997.6411. [DOI] [PubMed] [Google Scholar]
  • 118.Iulita MF, Claudio Cuello CA. The NGF metabolic pathway in the CNS and its dysregulation in down syndrome and Alzheimer’s disease. Curr Alzheimer Res. 2015 doi: 10.2174/1567205012666150921100030. [DOI] [PubMed] [Google Scholar]
  • 119.Chan-Palay V. Alterations in the locus coeruleus in dementias of Alzheimer’s and Parkinson’s disease. Prog Brain Res. 1991;88:625–30. doi: 10.1016/s0079-6123(08)63839-x. [DOI] [PubMed] [Google Scholar]
  • 120.Whitehouse PJ, Struble RG, Hedreen JC, Clark AW, Price DL. Alzheimer’s disease and related dementias: selective involvement of specific neuronal systems. CRC Crit Rev Clin Neurobiol. 1985;1(4):319–39. [PubMed] [Google Scholar]
  • 121.Tomlinson BE, Irving D, Blessed G. Cell loss in the locus coeruleus in senile dementia of Alzheimer type. J Neurol Sci. 1981;49(3):419–28. doi: 10.1016/0022-510x(81)90031-9. [DOI] [PubMed] [Google Scholar]
  • 122.Mann DM, Yates PO, Marcyniuk B. A comparison of changes in the nucleus basalis and locus caeruleus in Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 1984;47(2):201–3. doi: 10.1136/jnnp.47.2.201. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Haglund M, Sjöbeck M, Englund E. Locus ceruleus degeneration is ubiquitous in Alzheimer’s disease: possible implications for diagnosis and treatment. Neuropathology. 2006;26(6):528–32. doi: 10.1111/j.1440-1789.2006.00725.x. [DOI] [PubMed] [Google Scholar]
  • 124.Heneka MT, Nadrigny F, Regen T, Martinez-Hernandez A, Dumitrescu-Ozimek L, Terwel D, et al. Locus ceruleus controls Alzheimer’s disease pathology by modulating microglial functions through norepinephrine. Proc Natl Acad Sci U S A. 2010;107(13):6058–63. doi: 10.1073/pnas.0909586107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Feinstein DL, Heneka MT, Gavrilyuk V, Dello Russo C, Weinberg G, Galea E. Noradrenergic regulation of inflammatory gene expression in brain. Neurochem Int. 2002;41(5):357–65. doi: 10.1016/s0197-0186(02)00049-9. [DOI] [PubMed] [Google Scholar]
  • 126.Jardanhazi-Kurutz D, Kummer MP, Terwel D, Vogel K, Dyrks T, Thiele A, et al. Induced LC degeneration in APP/PS1 transgenic mice accelerates early cerebral amyloidosis and cognitive deficits. Neurochem Int. 2010;57(4):375–82. doi: 10.1016/j.neuint.2010.02.001. [DOI] [PubMed] [Google Scholar]
  • 127.Kalinin S, Gavrilyuk V, Polak PE, Vasser R, Zhao J, Heneka MT, et al. Noradrenaline deficiency in brain increases beta-amyloid plaque burden in an animal model of Alzheimer’s disease. Neurobiol Aging. 2007;28(8):1206–14. doi: 10.1016/j.neurobiolaging.2006.06.003. [DOI] [PubMed] [Google Scholar]
  • 128.Lockrow J, Boger H, Gerhardt G, Aston-Jones G, Bachman D, Granholm AC. A noradrenergic lesion exacerbates neurodegeneration in a Down syndrome mouse model. J Alzheimers Dis. 2011;23(3):471–89. doi: 10.3233/JAD-2010-101218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Counts SE, Mufson EJ. Noradrenaline activation of neurotrophic pathways protects against neuronal amyloid toxicity. J Neurochem. 2010;113(3):649–60. doi: 10.1111/j.1471-4159.2010.06622.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Marien MR, Colpaert FC, Rosenquist AC. Noradrenergic mechanisms in neurodegenerative diseases: a theory. Brain Res Brain Res Rev. 2004;45(1):38–78. doi: 10.1016/j.brainresrev.2004.02.002. [DOI] [PubMed] [Google Scholar]
  • 131.Wenk GL, McGann K, Hauss-Wegrzyniak B, Rosi S. The toxicity of tumor necrosis factor-alpha upon cholinergic neurons within the nucleus basalis and the role of norepinephrine in the regulation of inflammation: implications for Alzheimer’s disease. Neuroscience. 2003;121(3):719–29. doi: 10.1016/s0306-4522(03)00545-1. [DOI] [PubMed] [Google Scholar]
  • 132.Rizk P, Salazar J, Raisman-Vozari R, Marien M, Ruberg M, Colpaert F, et al. The alpha2-adrenoceptor antagonist dexefaroxan enhances hippocampal neurogenesis by increasing the survival and differentiation of new granule cells. Neuropsychopharmacology. 2006;31(6):1146–57. doi: 10.1038/sj.npp.1300954. [DOI] [PubMed] [Google Scholar]
  • 133.Decker MW, McGaugh JL. Effects of concurrent manipulations of cholinergic and noradrenergic function on learning and retention in mice. Brain Res. 1989;477(1–2):29–37. doi: 10.1016/0006-8993(89)91391-7. [DOI] [PubMed] [Google Scholar]
  • 134.Harley C. Noradrenergic and locus coeruleus modulation of the perforant path-evoked potential in rat dentate gyrus supports a role for the locus coeruleus in attentional and memorial processes. Prog Brain Res. 1991;88:307–21. doi: 10.1016/s0079-6123(08)63818-2. [DOI] [PubMed] [Google Scholar]
  • 135.Gibbs ME, Summers RJ. Role of adrenoceptor subtypes in memory consolidation. Prog Neurobiol. 2002;67(5):345–91. doi: 10.1016/s0301-0082(02)00023-0. [DOI] [PubMed] [Google Scholar]
  • 136.Salehi A, Faizi M, Colas D, Valletta J, Laguna J, Takimoto-Kimura R, et al. Restoration of norepinephrine-modulated contextual memory in a mouse model of Down syndrome. Sci Transl Med. 2009;1(7):7ra17. doi: 10.1126/scitranslmed.3000258. [DOI] [PubMed] [Google Scholar]
  • 137.Fortress AM, Hamlett ED, Vazey EM, Aston-Jones G, Cass WA, Boger HA, et al. Designer receptors enhance memory in a mouse model of down syndrome. J Neurosci. 2015;35(4):1343–53. doi: 10.1523/JNEUROSCI.2658-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Kalinin S, Polak PE, Madrigal JL, Gavrilyuk V, Sharp A, Chauhan N, et al. Beta-amyloid-dependent expression of NOS2 in neurons: prevention by an alpha2-adrenergic antagonist. Antioxid Redox Signal. 2006;8(5–6):873–83. doi: 10.1089/ars.2006.8.873. [DOI] [PubMed] [Google Scholar]
  • 139.Milstein JA, Lehmann O, Theobald DE, Dalley JW, Robbins TW. Selective depletion of cortical noradrenaline by anti-dopamine beta-hydroxylase-saporin impairs attentional function and enhances the effects of guanfacine in the rat. Psychopharmacology (Berl) 2007;190(1):51–63. doi: 10.1007/s00213-006-0594-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Bosboom JL, Stoffers D, Wolters ECh. Cognitive dysfunction and dementia in Parkinson’s disease. J Neural Transm. 2004;111(10–11):1303–15. doi: 10.1007/s00702-004-0168-1. [DOI] [PubMed] [Google Scholar]
  • 141.Asherson P, Stes S, Nilsson Markhed M, Berggren L, Svanborg P, Kutzelnigg A, et al. The effects of atomoxetine on emotional control in adults with ADHD: An integrated analysis of multicenter studies. Eur Psychiatry. 2015;30(4):511–20. doi: 10.1016/j.eurpsy.2014.12.002. [DOI] [PubMed] [Google Scholar]
  • 142.Lanfranchi S, Jerman O, Dal Pont E, Alberti A, Vianello R. Executive function in adolescents with Down Syndrome. J Intellect Disabil Res. 2010;54(4):308–19. doi: 10.1111/j.1365-2788.2010.01262.x. [DOI] [PubMed] [Google Scholar]
  • 143.Beyreuther K, Dyrks T, Hilbich C, Mönning U, König G, Multhaup G, et al. Amyloid precursor protein (APP) and beta A4 amyloid in Alzheimer’s disease and Down syndrome. Prog Clin Biol Res. 1992;379:159–82. [PubMed] [Google Scholar]
  • 144.Cork LC. Neuropathology of Down syndrome and Alzheimer disease. Am J Med Genet Suppl. 1990;7:282–6. doi: 10.1002/ajmg.1320370756. [DOI] [PubMed] [Google Scholar]
  • 145.Jiang Y, Mullaney KA, Peterhoff CM, Che S, Schmidt SD, Boyer-Boiteau A, et al. Alzheimer’s-related endosome dysfunction in Down syndrome is Aβ-independent but requires APP and is reversed by BACE-1 inhibition. Proc Natl Acad Sci. 2010;107(4):1630–35. doi: 10.1073/pnas.0908953107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Kim S, Sato Y, Mohan PS, Peterhoff C, Pensalfini A, Rigoglioso A, et al. Evidence that the Rab5 effector APPL1 mediates APP-ßCTF induced dysfunction of endosomes in Down syndrome and Alzheimer’s disease. Mol Psych. 2015 doi: 10.1038/mp.2015.97. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Cossec JC, Simon A, Marquer C, Moldrich RX, Leterrier C, Rossier J, et al. Clathrin-dependent APP endocytosis and Abeta secretion are highly sensitive to the level of plasma membrane cholesterol. Biochim Biophys Acta. 2010;1801:846–852. doi: 10.1016/j.bbalip.2010.05.010. [DOI] [PubMed] [Google Scholar]
  • 148.Nyakas C, Granic I, Halmy LG, Banerjee P, Luiten PG. The basal forebrain cholinergic system in aging and dementia. Rescuing cholinergic neurons from neurotoxic amyloid-beta42 with memantine. Behav Brain Res. 2011;221(2):594–603. doi: 10.1016/j.bbr.2010.05.033. [DOI] [PubMed] [Google Scholar]
  • 149.Karuppagounder SS, Pinto JT, Xu H, Chen HL, Beal MF, Gibson GE. Dietary supplementation with resveratrol reduces plaque pathology in a transgenic model of Alzheimer’s disease. Neurochem Int. 2009;54(2):111–8. doi: 10.1016/j.neuint.2008.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Trazzi S, Fuchs C, Valli E, Perini G, Bartesaghi R, Ciani E. The amyloid precursor protein (APP) triplicated gene impairs neuronal precursor differentiation and neurite development through two different domains in the Ts65Dn mouse model for Down syndrome. J Biol Chem. 2013;288(29):20817–29. doi: 10.1074/jbc.M113.451088. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 151.Trazzi S, Fuchs C, De Franceschi M, Mitrugno VM, Bartesaghi R, Ciani E. APP-dependent alteration of GSK3beta activity impairs neurogenesis in the Ts65Dn mouse model of Down syndrome. Neurobiol Dis. 2014;67:24–36. doi: 10.1016/j.nbd.2014.03.003. [DOI] [PubMed] [Google Scholar]
  • 152.Begenisic T, Baroncelli L, Sansevero G, Milanese M, Bonifacino T, Bonanno G, et al. Fluoxetine in adulthood normalizes GABA release and rescues hippocampal synaptic plasticity and spatial memory in a mouse model of Down syndrome. Neurobiol Dis. 2014;63:12–9. doi: 10.1016/j.nbd.2013.11.010. [DOI] [PubMed] [Google Scholar]
  • 153.Heinen M, Hettich MM, Ryan DP, Schnell S, Paesler K, Ehninger D. Adult-onset fluoxetine treatment does not improve behavioral impairments and may have adverse effects on the Ts65Dn mouse model of Down syndrome. Neural Plast. 2012;2012:467251. doi: 10.1155/2012/467251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Richards SJ, Edwards P, Dunnett SB. Animal models of the neuropathology observed in Alzheimer’s disease and Down syndrome. Prog Clin Biol Res. 1992;379:245–58. [PubMed] [Google Scholar]
  • 155.Streit WJ, Braak H, Xue QS, Bechmann I. Dystrophic (senescent) rather than activated microglial cells are associated with tau pathology and likely precede neurodegeneration in Alzheimer’s disease. Acta Neuropathol. 2009;118(4):475–85. doi: 10.1007/s00401-009-0556-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.van Leeuwen FW, van Tijn P, Sonnemans MA, Hobo B, Mann DM, Van Broeckhoven C, et al. Frameshift proteins in autosomal dominant forms of Alzheimer disease and other tauopathies. Neurology. 2006;66(2 Suppl 1):S86–92. doi: 10.1212/01.wnl.0000193882.46003.6d. [DOI] [PubMed] [Google Scholar]
  • 157.Qian W, Jin N, Shi J, Yin X, Jin X, Wang S, et al. Dual-specificity tyrosine phosphorylation-regulated kinase 1A (Dyrk1A) enhances tau expression. J Alzheimers Dis. 2013;37(3):529–38. doi: 10.3233/JAD-130824. [DOI] [PubMed] [Google Scholar]
  • 158.Rachubinski AL, Maclean KN, Evans JR, Bjugstad KB. Modulating cognitive deficits and tau accumulation in a mouse model of aging Down syndrome through neonatal implantation of neural progenitor cells. Exp Gerontol. 2012;47(9):723–33. doi: 10.1016/j.exger.2012.06.013. [DOI] [PubMed] [Google Scholar]
  • 159.German DC, Eisch AJ. Mouse models of Alzheimer’s disease: insight into treatment. Rev Neurosci. 2004;15(5):353–69. doi: 10.1515/revneuro.2004.15.5.353. [DOI] [PubMed] [Google Scholar]
  • 160.King DL, Arendash GW. Behavioral characterization of the Tg2576 transgenic model of Alzheimer’s disease through 19 months. Physiol Behav. 2002;75(5):627–42. doi: 10.1016/s0031-9384(02)00639-x. [DOI] [PubMed] [Google Scholar]
  • 161.Resende R, Moreira PI, Proença T, Deshpande A, Busciglio J, Pereira C, et al. Brain oxidative stress in a triple-transgenic mouse model of Alzheimer disease. Free Radic Biol Med. 2008;44(12):2051–7. doi: 10.1016/j.freeradbiomed.2008.03.012. [DOI] [PubMed] [Google Scholar]
  • 162.Bilkei-Gorzo A. Genetic mouse models of brain ageing and Alzheimer’s disease. Pharmacol Ther. 2014;142(2):244–57. doi: 10.1016/j.pharmthera.2013.12.009. [DOI] [PubMed] [Google Scholar]
  • 163.Morgan D. Amyloid, memory and neurogenesis. Exp Neurol. 2007;205(2):330–5. doi: 10.1016/j.expneurol.2007.03.006. [DOI] [PubMed] [Google Scholar]
  • 164.Alamed J, Wilcock DM, Diamond DM, Gordon MN, Morgan D. Two-day radial-arm water maze learning and memory task; robust resolution of amyloid-related memory deficits in transgenic mice. Nat Protoc. 2006;1(4):1671–9. doi: 10.1038/nprot.2006.275. [DOI] [PubMed] [Google Scholar]
  • 165.Scholtzova H, Wadghiri YZ, Douadi M, Sigurdsson EM, Li YS, Quartermain D, et al. Memantine leads to behavioral improvement and amyloid reduction in Alzheimer’s-disease-model transgenic mice shown as by micromagnetic resonance imaging. J Neurosci Res. 2008;86(12):2784–91. doi: 10.1002/jnr.21713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Lott IT, Head E. Alzheimer disease and Down syndrome: factors in pathogenesis. Neurobiol Aging. 2005;26(3):383–9. doi: 10.1016/j.neurobiolaging.2004.08.005. [DOI] [PubMed] [Google Scholar]
  • 167.Griffin WS. Inflammation and neurodegenerative diseases. Am J Clin Nutr. 2006;83(2):470S–474S. doi: 10.1093/ajcn/83.2.470S. [DOI] [PubMed] [Google Scholar]
  • 168.Wenk GL, McGann K, Mencarelli A, Hauss-Wegrzyniak B, Del Soldato P, Fiorucci S. Mechanisms to prevent the toxicity of chronic neuroinflammation on forebrain cholinergic neurons. Eur J Pharmacol. 2000;402(1–2):77–85. doi: 10.1016/s0014-2999(00)00523-9. [DOI] [PubMed] [Google Scholar]
  • 169.Carnevale D, De Simone R, Minghetti L. Microglia-neuron interaction in inflammatory and degenerative diseases: role of cholinergic and noradrenergic systems. CNS Neurol Disord Drug Targets. 2007;6(6):388–97. doi: 10.2174/187152707783399193. [DOI] [PubMed] [Google Scholar]
  • 170.Zassler B, Weis C, Humpel C. Tumor necrosis factor-alpha triggers cell death of sensitized potassium chloride-stimulated cholinergic neurons. Brain Res Mol Brain Res. 2003;113(1–2):78–85. doi: 10.1016/s0169-328x(03)00092-5. [DOI] [PubMed] [Google Scholar]
  • 171.Heneka MT, Galea E, Gavriluyk V, Dumitrescu-Ozimek L, Daeschner J, O’Banion MK, et al. Noradrenergic depletion potentiates beta -amyloid-induced cortical inflammation: implications for Alzheimer’s disease. J Neurosci. 2002;22(7):2434–42. doi: 10.1523/JNEUROSCI.22-07-02434.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Appleby BS, Cummings JL. Discovering new treatments for Alzheimer’s disease by repurposing approved medications. Curr Top Med Chem. 2013;13(18):2306–27. doi: 10.2174/15680266113136660162. [DOI] [PubMed] [Google Scholar]
  • 173.Cuello AC, Ferretti MT, Leon WC, Iulita MF, Melis T, Ducatenzeiler A, et al. Early-stage inflammation and experimental therapy in transgenic models of the Alzheimer-like amyloid pathology. Neurodegener Dis. 2010;7(1–3):96–8. doi: 10.1159/000285514. [DOI] [PubMed] [Google Scholar]
  • 174.Hunter CL, Bachman D, Granholm AC. Minocycline prevents cholinergic loss in a mouse model of Down’s syndrome. Ann Neurol. 2004;56(5):675–88. doi: 10.1002/ana.20250. [DOI] [PubMed] [Google Scholar]
  • 175.Parihar MS, Hemnani T. Alzheimer’s disease pathogenesis and therapeutic interventions. J Clin Neurosci. 2004;11(5):456–67. doi: 10.1016/j.jocn.2003.12.007. [DOI] [PubMed] [Google Scholar]
  • 176.Smith MA, Hirai K, Hsiao K, Pappolla MA, Harris PL, Siedlak SL, et al. Amyloid-beta deposition in Alzheimer transgenic mice is associated with oxidative stress. J Neurochem. 1998;70(5):2212–5. doi: 10.1046/j.1471-4159.1998.70052212.x. [DOI] [PubMed] [Google Scholar]
  • 177.Jovanovic SV, Clements D, MacLeod K. Biomarkers of oxidative stress are significantly elevated in Down syndrome. Free Radic Biol Med. 1998;25(9):1044–8. doi: 10.1016/s0891-5849(98)00137-3. [DOI] [PubMed] [Google Scholar]
  • 178.Parisotto EB, Garlet TR, Cavalli VL, Zamoner A, da Rosa JS, Bastos J, et al. Antioxidant intervention attenuates oxidative stress in children and teenagers with Down syndrome. Res Dev Disabil. 2014;35(6):1228–36. doi: 10.1016/j.ridd.2014.03.013. [DOI] [PubMed] [Google Scholar]
  • 179.Behar TN, Colton CA. Redox regulation of neuronal migration in a Down Syndrome model. Free Radic Biol Med. 2003;35(6):566–75. doi: 10.1016/s0891-5849(03)00329-0. [DOI] [PubMed] [Google Scholar]
  • 180.Bambrick LL, Fiskum G. Mitochondrial dysfunction in mouse trisomy 16 brain. Brain Res. 2008;1188:9–16. doi: 10.1016/j.brainres.2007.10.045. [DOI] [PubMed] [Google Scholar]
  • 181.Busciglio J, Yankner BA. Apoptosis and increased generation of reactive oxygen species in Down’s syndrome neurons in vitro. Nature. 1995;378(6559):776–9. doi: 10.1038/378776a0. [DOI] [PubMed] [Google Scholar]
  • 182.Sung S, Yao Y, Uryu K, Yang H, Lee VM, Trojanowski JQ, et al. Early vitamin E supplementation in young but not aged mice reduces Abeta levels and amyloid deposition in a transgenic model of Alzheimer’s disease. FASEB J. 2004;18(2):323–5. doi: 10.1096/fj.03-0961fje. [DOI] [PubMed] [Google Scholar]
  • 183.Lott IT, Doran E, Nguyen VQ, Tournay A, Head E, Gillen DL. Down syndrome and dementia: a randomized, controlled trial of antioxidant supplementation. Am J Med Genet A. 2011;155A(8):1939–48. doi: 10.1002/ajmg.a.34114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Giacobini E. Present and future of Alzheimer therapy. J Neural Transm Suppl. 2000;59:231–42. doi: 10.1007/978-3-7091-6781-6_24. [DOI] [PubMed] [Google Scholar]
  • 185.Prasher VP. Review of donepezil, rivastigmine, galantamine and memantine for the treatment of dementia in Alzheimer’s disease in adults with Down syndrome: implications for the intellectual disability population. Int J Geriatr Psychiatry. 2004;19(6):509–15. doi: 10.1002/gps.1077. [DOI] [PubMed] [Google Scholar]
  • 186.Kondoh T, Kanno A, Itoh H, Nakashima M, Honda R, Kojima M, et al. Donepezil significantly improves abilities in daily lives of female Down syndrome patients with severe cognitive impairment: a 24-week randomized, double-blind, placebo-controlled trial. Int J Psychiatry Med. 2011;41(1):71–89. doi: 10.2190/PM.41.1.g. [DOI] [PubMed] [Google Scholar]
  • 187.Villarroya M, García AG, Marco JL. New classes of AChE inhibitors with additional pharmacological effects of interest for the treatment of Alzheimer’s disease. Curr Pharm Des. 2004;10(25):3177–84. doi: 10.2174/1381612043383368. [DOI] [PubMed] [Google Scholar]
  • 188.Scarpini E, Scheltens P, Feldman H. Treatment of Alzheimer’s disease: current status and new perspectives. Lancet Neurol. 2003;2(9):539–47. doi: 10.1016/s1474-4422(03)00502-7. [DOI] [PubMed] [Google Scholar]
  • 189.Petersen RC, Thomas RG, Grundman M, Bennett D, Doody R, Ferris S, et al. Vitamin E and donepezil for the treatment of mild cognitive impairment. N Engl J Med. 2005;352(23):2379–88. doi: 10.1056/NEJMoa050151. [DOI] [PubMed] [Google Scholar]
  • 190.Costa AC, Scott-McKean JJ. Prospects for improving brain function in individuals with Down syndrome. CNS Drugs. 2013;27(9):679–702. doi: 10.1007/s40263-013-0089-3. [DOI] [PubMed] [Google Scholar]
  • 191.Heise I, Fisher SP, Banks GT, Wells S, Peirson SN, Foster RG, et al. Sleep-like behavior and 24-h rhythm disruption in the Tc1 mouse model of Down syndrome. Genes Brain Behav. 2015;14(2):209–16. doi: 10.1111/gbb.12198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Heller HC, Salehi A, Chuluun B, Das D, Lin B, Moghadam S, et al. Nest building is impaired in the Ts65Dn mouse model of Down syndrome and rescued by blocking 5HT2a receptors. Neurobiol Learn Mem. 2014;116:162–71. doi: 10.1016/j.nlm.2014.10.002. [DOI] [PubMed] [Google Scholar]
  • 193.Sahún I, Marechal D, Pereira PL, Nalesso V, Gruart A, Garcia JM, et al. Cognition and hippocampal plasticity in the mouse is altered by monosomy of a genomic region implicated in Down syndrome. Genetics. 2014;197(3):899–912. doi: 10.1534/genetics.114.165241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Smith GK, Kesner RP, Korenberg JR. Dentate gyrus mediates cognitive function in the Ts65Dn/DnJ mouse model of Down syndrome. Hippocampus. 2014;24(3):354–62. doi: 10.1002/hipo.22229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Galdzicki Z, Siarey RJ. Understanding mental retardation in Down’s syndrome using trisomy 16 mouse models. Genes Brain Behav. 2003;2(3):167–78. doi: 10.1034/j.1601-183x.2003.00024.x. [DOI] [PubMed] [Google Scholar]
  • 196.Coussons-Read ME, Crnic LS. Behavioral assessment of the Ts65Dn mouse, a model for Down syndrome: altered behavior in the elevated plus maze and open field. Behav Genet. 1996;26(1):7–13. doi: 10.1007/BF02361154. [DOI] [PubMed] [Google Scholar]
  • 197.Kleschevnikov AM, Belichenko PV, Faizi M, Jacobs LF, Htun K, Shamloo M, et al. Deficits in cognition and synaptic plasticity in a mouse model of Down syndrome ameliorated by GABAB receptor antagonists. J Neurosci. 2012;32(27):9217–27. doi: 10.1523/JNEUROSCI.1673-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Ash JA, Velazquez R, Kelley CM, Powers BE, Ginsberg SD, Mufson EJ, et al. Maternal choline supplementation improves spatial mapping and increases basal forebrain cholinergic neuron number and size in aged Ts65Dn mice. Neurobiol Dis. 2014;70:32–42. doi: 10.1016/j.nbd.2014.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Tabesh A, Jensen JH, Ardekani BA, Helpern JA. Estimation of tensors and tensor-derived measures in diffusional kurtosis imaging. Magn Reson Med. 2011;65(3):823–36. doi: 10.1002/mrm.22655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Rueda N, Flórez J, Martínez-Cué C. Chronic pentylenetetrazole but not donepezil treatment rescues spatial cognition in Ts65Dn mice, a model for Down syndrome. Neurosci Lett. 2008;433(1):22–7. doi: 10.1016/j.neulet.2007.12.039. [DOI] [PubMed] [Google Scholar]
  • 201.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(7):1624–32. doi: 10.1038/sj.npp.1301535. [DOI] [PubMed] [Google Scholar]
  • 202.Rueda N, Llorens-Martín M, Flórez J, Valdizán E, Banerjee P, Trejo JL, et al. Memantine normalizes several phenotypic features in the Ts65Dn mouse model of Down syndrome. J Alzheimers Dis. 2010;21(1):277–90. doi: 10.3233/JAD-2010-100240. [DOI] [PubMed] [Google Scholar]
  • 203.Bennett FC, McClelland S, Kriegsmann EA, Andrus LB, Sells CJ. Vitamin and mineral supplementation in Down’s syndrome. Pediatrics. 1983;72(5):707–13. [PubMed] [Google Scholar]
  • 204.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(1):256–61. doi: 10.1016/j.expneurol.2006.02.005. [DOI] [PubMed] [Google Scholar]
  • 205.De la Torre R, De Sola S, Pons M, Duchon A, de Lagran MM, Farré M, et al. Epigallocatechin-3-gallate, a DYRK1A inhibitor, rescues cognitive deficits in Down syndrome mouse models and in humans. Mol Nutr Food Res. 2014;58(2):278–88. doi: 10.1002/mnfr.201300325. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Ann Charlotte Granholm Bentley Profile Picture

RESOURCES