Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jun 1.
Published in final edited form as: FASEB J. 2023 Jun;37(6):e22944. doi: 10.1096/fj.202202111RR

Basal forebrain cholinergic neurons are vulnerable in a mouse model of Down syndrome and their molecular fingerprint is rescued by maternal choline supplementation

Melissa J Alldred 1,2, Harshitha Pidikiti 1, Adriana Heguy 3, Panos Roussos 1,6, Stephen D Ginsberg 1,2,4,5
PMCID: PMC10292934  NIHMSID: NIHMS1897993  PMID: 37191946

Abstract

Basal forebrain cholinergic neuron (BFCN) degeneration is a hallmark of Down syndrome (DS) and Alzheimer’s disease (AD). Current therapeutics in these disorders have been unsuccessful in slowing disease progression, likely due to poorly understood complex pathological interactions and dysregulated pathways. The Ts65Dn trisomic mouse model recapitulates both cognitive and morphological deficits of DS and AD, including BFCN degeneration and has shown lifelong behavioral changes due to maternal choline supplementation (MCS). To test the impact of MCS on trisomic BFCNs, we performed laser capture microdissection to individually isolate choline acetyltransferase-immunopositive neurons in Ts65Dn and disomic littermates, in conjunction with MCS at the onset of BFCN degeneration. We utilized single population RNA sequencing (RNA-seq) to interrogate transcriptomic changes within medial septal nucleus (MSN) BFCNs. Leveraging multiple bioinformatic analysis programs on differentially expressed genes (DEGs) by genotype and diet, we identified key canonical pathways and altered physiological functions within Ts65Dn MSN BFCNs, which were attenuated by MCS in trisomic offspring, including the cholinergic, glutamatergic and GABAergic pathways. We linked differential gene expression bioinformatically to multiple neurological functions, including motor dysfunction/movement disorder, early onset neurological disease, ataxia and cognitive impairment via Ingenuity Pathway Analysis. DEGs within these identified pathways may underlie aberrant behavior in the DS mice, with MCS attenuating the underlying gene expression changes. We propose MCS ameliorates aberrant BFCN gene expression within the septohippocampal circuit of trisomic mice through normalization of principally the cholinergic, glutamatergic and GABAergic signaling pathways, resulting in attenuation of underlying neurological disease functions.

Keywords: RNA-seq, laser capture microdissection, maternal choline supplementation, medial septum, Down syndrome, Alzheimer’s disease, trisomy, selective vulnerability, bioinformatics

Graphical Abstract

graphic file with name nihms-1897993-f0001.jpg

Single population RNA sequencing was employed using basal forebrain cholinergic neurons (BFCNs) in the Ts65Dn trisomic mouse model of Down syndrome and Alzheimer’s disease to determine if maternal choline supplementation (MCS) had a neuroprotective effect. We identified key canonical pathways attenuated by MCS in 6 month old trisomic offspring compared to disomic littermates. We propose MCS ameliorates aberrant BFCN gene expression within the septohippocampal circuit of trisomic mice through normalization of principally the cholinergic, glutamatergic, and GABAergic signaling pathways.

Introduction

Down syndrome (DS) is a neurodevelopmental and neurodegenerative genetic disorder caused by triplication of human chromosome 21 (HSA21), which is observed in ~1 of 700 births, and is primary cause of intellectual disability (1, 2). DS results in systemic and neurological conditions, including deficits in learning and memory (38). Individuals with DS also display the onset of neurodegeneration associated with Alzheimer’s disease (AD) in early mid-life, including amyloid plaques, neurofibrillary tangles, and cognitive decline, with age-associated escalation (916). While persons with DS have significantly improved lifespans due to medical intervention (1720), these individuals have increased health complications, with the development of dementia accounting for >70% of deaths in individuals over the age of 35 (21). Age-related cognitive decline in DS and AD is associated with degeneration of the cholinergic basal forebrain system, including neuronal loss in subregions of the basal forebrain and specific loss of cholinergic basal forebrain neurons in the nucleus basalis and cholinergic fiber projections to the hippocampus and neocortex (4, 2226).

Many human DS neuropathological features are recapitulated in the Ts65Dn mouse model, including basal forebrain cholinergic neuron (BFCN) degeneration, AD-like hippocampal-dependent learning and memory deficits, and septohippocampal circuit degeneration, notably including cholinergic, glutamatergic, and GABAergic dysfunction (9, 2735). The medial septal nucleus/ventral diagonal band (MSN) of the BFCN system projects to the hippocampus and is critical for learning and memory (36, 37). Degeneration of the BFCN projection system is strongly associated with DS cognitive decline (3032, 3842). BFCN degeneration is a cardinal feature of the Ts65Dn mouse, beginning at approximately 6 months of age (MO) (29, 31, 32, 40, 4349). BFCN loss and alteration of hippocampal innervation are consistently reported in older (>10 MO) Ts65Dn mice (32, 40, 42, 50, 51). Ts65Dn mice have recently been shown to have significant changes in gene expression within MSN BFCNs at 6 MO by laser capture microdissection (LCM) coupled with single population RNA sequencing (RNA-seq) (52) and in older trisomic mice by LCM coupled with custom-designed microarray analysis (48). Currently, there is no therapeutic intervention that may slow or stop the BFCN degeneration seen in both mouse and human DS and AD.

Choline is an essential nutrient required for development and homeostasis, and increased consumption of choline is necessary during pregnancy in human and rodent models (5355). Choline metabolite plasma levels are depleted in pregnant dams when standard choline levels are present in rodent chow (56, 57). These reports support the hypothesis that current dietary recommendations for choline are insufficient during pregnancy and lactation (45, 54, 55, 58). Choline is necessary for proper brain development and function, as it is requisite for biosynthesis of acetylcholine, the primary dietary methyl donor, and is a key substrate in the phosphatidylethanolamine N-methyltransferase (PEMT) pathway. Acetylcholine is a critical neurotransmitter which regulates multiple neurodevelopmental niches including proliferation, migration, and synapse formation, among others (44, 45, 59, 60), while methylation affects gene expression regulation through epigenetic programming (6164). The PEMT pathway utilizes choline as the substrate for formation of several structural membrane phospholipids, including sphingomyelin and phosphatidylcholine (65, 66).

Maternal choline supplementation (MCS) involves increasing dietary choline intake during pregnancy and lactation (67). MCS is inexpensive and well-tolerated, with numerous publications showing beneficial effects in rodent and human trials (41, 44, 55, 6873). MCS positively impacts multiple aspects of learning and memory in offspring of both normal rodents and in disease models (45, 65). MCS has been utilized in the Ts65Dn mouse model with positive behavioral, cellular and gene expression effects, including benefits in spatial and attentional memory recorded up to 16 MO in Ts65Dn mice, although these effects are not as profound in aged cohorts (31, 41, 43, 70, 74). Further, these trisomic mice have shown morphological benefits in the basal forebrain and hippocampus, along with amelioration of dysfunctional gene expression within hippocampal pyramidal and BFCNs, although these inquiries were limited by the relatively small number of genes queried by microarray analyses (41, 44, 48, 75, 76). Recent human studies have shown long-term clinical benefits of MCS during the 3rd trimester, including improved processing speed in infants and sustained attention in children (55, 77). Understanding the underlying effects of MCS in the adult brain is critical, especially in those with genetic and/or neurodevelopmental abnormalities.

We employed LCM and single population RNA-seq to profile vulnerable BFCNs in the Ts65Dn mouse model of DS in the context of MCS. We postulate by isolating vulnerable MSN BFCNs from heterogenous cell populations and identifying dysregulated genes that are MCS responsive, we will identify targets that underlie the beneficial behavioral and functional changes. Characterizing these select gene and biological pathway targets at the onset of BFCN degeneration will pinpoint the underlying lifelong organizational benefits provided by MCS. These targets may then be utilized for therapeutic intervention that may help slow or stop the progression of BFCN degeneration associated with DS and AD. Further, we hypothesize differentially expressed genes (DEGs) impacted by MCS could identify previously unknown targets for DS and AD prevention.

Materials and Methods

Mice:

Animal protocols were approved by the Nathan Kline Institute/New York University Grossman School of Medicine (NYUGSOM) IACUC in accordance with NIH guidelines. Breeder pairs (female Ts65Dn and male C57Bl/6J Eicher x C3H/HeSnJ F1 mice) were purchased from Jackson Laboratories (Bar Harbor, ME) and mated at the Nathan Kline Institute. Breeder pairs were assigned to receive one of two choline-controlled experimental diets: i) control rodent diet containing 1.1 g/kg choline chloride (AIN-76A; Dyets Inc., Bethlehem, PA), or ii) choline-supplemented diet containing 5.0 g/kg choline chloride (AIN-76A; Dyets Inc.), as described previously. Pups received choline normal or choline supplemented diet as per their respective dam’s diet from E0-P21 and starting at weaning (P21) all offspring had ad libitum access to water and the control diet (31, 41, 42, 78, 79) (Fig. 1A). The choline-supplemented diet provides approximately 4.5 times the concentration of choline in the control diet and is within the normal physiological range (80). The control diet supplies an adequate level of choline, so the offspring are not choline deficient, meaning this is not a study of choline deficiency. Tail clips were taken and genotyped (81) at weaning and mice were aged to ~6 MO (Fig. 1A).

Figure 1:

Figure 1:

Overview of experimental workflow A. The MCS paradigm is shown, with dams and pups fed a normal choline (1.1 g/kg choline) or choline supplemented (+; 5.0 g/kg choline) diet during the perinatal period (E0-P21). Upon weaning (P21), pups are moved to a normal choline diet until sacrifice at 6 MO. B. Brains are immediately flash frozen, cryocut at 20 μm, adhered to PEN membrane slides and immunostained for ChAT-ir. C. LCM is performed on 500 individual MSN BFCNs under a 40X objective Scale bar 50 μm. Multiple sections are collected and combined for RNA purification. D. QC is performed on total RNA. RNA-seq library preparation is done and QC on resulting cDNA library for each sample. E. Illumina HiSeq 4000 single reads were performed by the NYUGSOM GTC. F. FastQ files were generated for the reads for each sample and bioinformatic analysis is initiated.

Tissue Preparation:

Brain tissues were accessed from unsupplemented Ts65Dn (Ts; n=6), MCS Ts65Dn (Ts+; n=8), unsupplemented disomic (2N; n=6) and MCS disomic (2N+; n =4) male mice (age range: 5.7–6.4 MO, mean age 6.0 MO). Mice were transcardially perfused with ice-cold 0.15 M phosphate buffer as previously described (75, 76, 78, 79). Brains were immediately flash frozen and 20 μm-thick tissue sections were cryostat cut (−25 °C) in the coronal plane (CM1860UV, Leica, Buffalo Grove, IL) and mounted on polyethylene naphthalate (PEN) membrane slides (Leica, Fig. 1B). Slides were kept under desiccant at −80 0C until used for immunohistochemistry. RNase-free precautions were employed, and solutions were made with 18.2 mega Ohm RNase-free water (Nanopure Diamond, Barnstead, Dubuque, IA).

Immunohistochemistry and neuron collection:

PEN membrane slides were equilibrated to room temperature (RT) under desiccant (−20 0C for 5 min, 4 0C for 10 min, RT for 5 min) followed by a rapid staining protocol utilizing antibody against choline acetyltransferase (ChAT) (AB144P, Millipore) to preserve intact RNA in the unfixed tissue as previously described (Fig. 1B) (52). ChAT-immunoreactive (ir) neurons were identified within the MSN and isolated by LCM (LMD7000; Leica; Fig 1C) as previously described (52). Approximately 500 ChAT-ir neurons were isolated per brain before proceeding to RNA isolation and RNA-seq library preparation.

RNA purification:

RNA from ~500 BFCNs was purified using miRNeasy Micro kit (Qiagen) according to manufacturers’ specifications. A DNase digestion was performed twice sequentially before the final washes and RNA purification. RNA quality control (QC) was performed (RNA 6000 pico kit, Agilent, Santa Clara, CA; Fig 1D).

Library Preparation and RNA-seq:

The SMARTer Stranded Total RNA-Seq kit-Pico input Mammalian (Takara Bio, Mountain View, CA) was employed with minor modifications to utilize full volume of RNA (52). Samples were quantified (Agilent 2100 HS DNA kit; Fig. 1D), and samples below 2 nM of library were excluded. Samples were pooled in equimolar concentrations and assayed on an HiSeq-4000 (Illumina, San Diego, CA) using a single read 50 cycle protocol at NYUGSOM Genome Technology Center (GTC) (Fig. 1E & F). Unsupplemented (e.g., normal choline diet) trisomic (Ts) and disomic (2N) RNA-seq profiles were previously generated (52), utilizing a different bioinformatics interrogation approach and were reanalyzed herein to compare expression profiles with offspring from dams exposed to MCS.

RNA-seq processing:

FastQ files were utilized for all four conditions (2N, 2N+, Ts, Ts+) to analyze data in parallel. FastQ files were generated and QC of the raw reads was performed by FastQC v0.11.9 (82). Read trimming was performed as necessary by Trimmomatic 0.39 (83). If QC passed and showed no adapter contamination, this step was skipped. Sequence reads were indexed and aligned to the reference genome (Gencode GRCm39-mm10) using STAR Aligner (2.7.10a) (84). Quantification was performed on aligned reads using Picard (2.27.1) (85) for different measures and RSEM (1.3.3) for output (86). QC was performed on aligned reads using RSeQC (v4.0.0) (88). Differential gene expression was performed using R version R-v4.2.0/RStudio v1+554 using gene results with the mouse reference genome (Gencode GRCm39-mm10) (Supplemental Fig. 1, Fig. 2A).

Figure 2:

Figure 2:

Pipeline for bioinformatic workflow using RNA-seq libraries derived from LCM-captured MSN BFCNs. A. Workflow for RNA-seq bioinformatics pipeline. B. PCA shows overall gene expression profiles for each mouse, with mean expression for each group as a larger sized dot. (black= 2N, blue= Ts, grey = 2N+, light blue= Ts+) C. Heatmaps illustrate individual gene expression differences from each individual mouse with DEGs by genotype (2N and Ts; 2,510 DEGs) and disease diet (Ts+ and Ts; 2,098 DEGs) binned by upregulation (pink) or downregulation (green).

Statistical analysis:

The Gene Count matrix obtained from RSEM was analyzed. Genes with over 0.1 counts per million (CPM) were retained followed by TMM normalization (87) implemented by edgeR (89) for downstream analysis. This step removes lowly expressed genes as they provide little evidence of differential expression and increase statistical errors and false discovery rates (90, 91). Analyses were performed using the Dream pipeline (92) which is built on top of limma-voom from the VariancePartition (93) package. In addition to Group and RNA concentration, the following variables were included as covariates: Intergenic percentage, Intronic percentage, Uniquely mapped, mRNA base percentage, Usable base percentage, and Correct strand reads percentage. With the exception of Group and RNA concentration, the other covariates are computed from RNA-seq reads by Picard (86). TopTable (edgeR; v3.38.1) extracts genes that are present for all comparisons. Gene expression differences at (p <0.05) were considered statistically significant. Protein coding genes were extracted using the R Bioconductor package AnnotationDbi (94). Multiple testing corrections were performed by false discovery rate (FDR) (95) (Fig. 2A).

Pathway Analyses:

Pathway analyses consisted of Ingenuity Pathway Analysis (IPA; Qiagen) (96, 97), Kyoto Encyclopedia of Genes and Genomes (KEGG) (98), Gene Ontology (GO) (99, 100) and STRING (101) in Cytoscape (cutoff 0.4) (102). GO analysis was filtered in excel utilizing key word targets to identify classes of processes affected by genotype and diet. STRING analysis was performed on Ts compared to 2N DEGs. Select genes including cholinergic receptor muscarinic 2 (Chrm2), glutamate ionotropic receptor NMDA type subunit 2A (Grin2a) and GABAA subunit gamma2 (Gabrg2) were filtered to isolate direct significant interactions and reanalyzed in STRING for each geneset to determine interactomes dysregulated in Ts mice. Once interactomes were identified, each gene was compared for disease diet effect (Ts+ versus Ts). This was performed to compare whether genes were still significantly different in the disease state. Genes significantly affected in disease diet drop out of the secondary STRING interactome, with those genes not rescued by diet in disease represented for each interactome.

qRT-PCR:

Validation was performed by qPCR utilizing the TaqMan Gene Expression Cell to Ct kit (Life Technologies, Grand Island, NY, USA) as described previously (52). Briefly, ~200 MSN neurons were isolated via LCM based on morphology from adjacent tissue sections in the same animals as utilized for RNA-seq (n=6 per genotype) after Nissl staining {0.1% thionin in sodium acetate (49.44 mM)/acetic acid (3.6 mM) buffer}(52, 103105). qRT-PCR was performed utilizing 2 μl cDNA from 50 μl reaction mixture with 22.5 μl input RNA. Taqman qPCR primers (Life Technologies) were selected from a subset of DEGs (52) that were significantly MCS responsive as evidenced in multiple pathways (e.g., IPA and KEGG). Probes included calcium/calmodulin dependent protein kinase II alpha (Camk2a; Mm01258148_m1), Chrm2 (Mm01167087_m1), Grin2a (Mm00433802_m1), mitogen-activated protein kinase 8 (Mapk8; Mm01218941_m1), and nerve growth factor receptor (Ngfr aka p75NTR; Mm01309638_m1). Samples were assayed in triplicate on a real-time qPCR cycler (PikoReal, ThermoFisher). 2N qPCR products were evaluated from previously published data (52) due to lack of additional sample material. The ddCT method was used to determine relative gene level differences between groups (106108). Glucuronidase Beta (GusB; Mm01197698_m1) qRT-PCR products was utilized as a control, as GusB did not show significant changes in RNA-seq data obtained from BFCNs herein or in previous analyses (52, 104, 105). Negative controls consisted of the reaction mixture without input RNA. Sample data was compared with respect to PCR product synthesis for each gene tested. qRT-PCR log-fold changes (LFCs) were utilized in concert with the Cell to Ct protocol as described previously (52). Violin plots were generated in Graphpad Prism (9.3.1, GraphPad, Boston, MA) normalizing qPCR product synthesis to 2N expression levels.

Results

Effects of MCS on MSN BFCN gene expression at 6 MO

RNA-seq was performed on MSN BFCNs from Ts, Ts+, 2N and 2N+ mice (Fig. 1 and Supplemental Fig. 1). RNA-seq reads from BFCNs were mapped and normalization and covariate analyses were performed using the Dream pipeline (92) (Fig. 2A). Variance analysis showed RNA input levels, intronic, and intergenic percentages were covariates and were adjusted accordingly. Principal component analysis (PCA) revealed 2N and 2N+ BFCN profiles cluster closely together while greater variability was observed in Ts and Ts+ BFCN profiles, indicating a stronger genotype and diet effect in Ts mice, with Ts+ profiles showing the most distinct gene expression pattern (Fig. 2B). Genotype and diet effects are interrogated by examining DEGs identified by genotype (Ts versus 2N) and disease diet (Ts+ versus Ts; Fig. 2C). PCA plots and heatmaps of DEGs likely reflect the onset of BFCN degeneration and indicate perinatal MCS has a profound beneficial effect on gene expression within BFCNs. A total of 2,510 genes were differentially expressed at p<0.05, with 511 genes reaching significance at FDR <0.05, by genotype (e.g., Ts compared to 2N). The Dream pipeline revealed 138 additional genes at FDR 0.05 and 1,067 additional DEGs at p<0.05 compared to the limma-voom pipeline (1,443 genes; (52)), indicating the Dream pipeline enabled us to differentiate small, but significant, differences in gene expression in the current paradigm. Therefore, we demonstrate the Dream pipeline is of high utility when determining DEGs from single population RNA-seq paradigms. Comparing disease diet (e.g., Ts+ versus Ts), Dream identified 2,098 DEGs at p<0.05, with 150 genes reaching significance at FDR 0.05. Comparisons for disomic diet (e.g., 2N+ versus 2N), supplemented genotype (e.g., Ts+ versus 2N+) and diet plus genotype (Ts+ versus 2N) revealed fewer DEGs (Fig. 3A). Since we are principally examining the effects genotype and diet in the trisomic model, we concentrated our investigation on genotype (Ts versus 2N) and disease diet (Ts+ versus Ts). The utility of the Dream pipeline is especially evident when querying DEGs by genotype. For example, in trisomic mice ~2.7-fold more downregulated genes were found compared to upregulated genes, with the majority of these DEGs showing small LFCs (< 2.5; 57.8%) indicating significant DEGs in trisomic mice, even those with low expression levels, can be readily identified using the Dream pipeline. This observation is confirmed when with the disease diet comparison, as Ts+ mice displayed approximately equal numbers of upregulated and downregulated genes, but approximately the same percentage (58.3%) of DEGs with small LFCs (<2.5) compared to genotype (Fig. 3B). Over 90% of DEGs were protein coding. The remainder of DEGs were noncoding RNAs (ncRNAs), pseudogenes, and microRNAs (miRNAs) (Fig. 3C and Supplemental Fig. 2). Volcano plots demonstrate individual DEGs by diet and genotype via LFC (Fig. 3D and Supplemental Fig. 2C). DEGs for each comparison (genotype x diet) are presented in Supplemental Tables 15.

Figure 3:

Figure 3:

DEGs are presented by genotype (Ts and 2N) and maternal diet (MCS and normal choline). A. Table showing the DEGs at each FDR and p-value cutoff from the total number of analyzed genes. B. Bar graphs highlighting genes per LFC bin upregulated (pink) and downregulated (green) by genotype and disease diet. C. Pie charts show percentage of DEGs for protein coding and ncRNAs. D. Volcano plots show upregulated and downregulated genes with individual genes noted per dot. Light green dots indicate p<0.01 downregulated, dark green dots indicate p<0.05 downregulated, light pink dots indicate p<0.01 upregulated, and dark pink dots indicate p<0.05 upregulated.

Impact of MCS on triplicated genes

We interrogated triplicated genes dysregulated in BFCNs by genotype and found 14 (of 73 with detectable expression) dysregulated (11 upregulated and 3 downregulated). We queried whether these triplicated genes were impacted in disease diet and found 7 genes significantly rescued in terms of gene expression by MCS. These included DnaJ heat shock protein family (Hsp40) member C28 (Dnajc28), E26 avian leukemia oncogene 2, 3’ domain (Ets2), SH3 domain protein 1A intersectin 1(Itsn1), junction adhesion molecule 2 (Jam2), MX dynamin-like GTPase 2 (Mx2), PAX3 and PAX7 binding protein 1 (Paxbp1), and T cell lymphoma invasion and metastasis 1 (Tiam1), (p<0.05, Fig. 4A) and two trend-level (bromodomain and WD repeat domain containing 1 (Brwd1) and dual-specificity tyrosine-(Y)-phosphorylation regulated kinase 1a (Dyrk1a); 0.05<p<0.1, Fig 4B). Only 5 triplicated genes, including DOP1 leucine zipper like protein B (Dop1b), receptor-interacting serine-threonine kinase 4 (Ripk4), Son DNA binding protein (Son), synaptojanin1 (Synj1), and tetratricopeptide repeat domain 3 (Ttc3) of the 14 genes significantly dysregulated in Ts BFCNs were not MCS responsive in trisomic mice (Fig. 4B). Another 4 genes triplicated in Ts65Dn mice that did not show significant genotype differences were disease diet responsive, namely ATP synthase, H+ transporting, mitochondrial F1 complex, O subunit (Atp5o), chaperonin containing Tcp1, subunit 8 (theta) (Cct8), potassium inwardly-rectifying channel, subfamily J, member 6 (Kcnj6), and solute carrier family 5 (inositol transporters), member 3 (Slc5a3) (Fig. 4C). Summating these results, DS triplicated gene expression within BFCNs is significantly decreased in the disease diet (e.g., Ts+ versus Ts) paradigm (Fig. 4B).

Figure 4:

Figure 4:

Trisomic protein coding genes do not necessarily match copy number within vulnerable MSN BFCNs. A total of 73 genes (of 88) show quantifiable expression levels with 7 DEGs attaining statistical significance at 6 MO by genotype were rescued in disease diet (A), an additional 7 genes were only affected by genotype (B), and an additional 4 genes were only affected by disease diet (C). Grey vertical dashes delineate DEGs affected by genotype and rescued in disease diet, those significant only by genotype, and those significantly affected by disease diet. D. Violin plot of 73 triplicated HSA21 orthologs expressed in BFCNs via LFC by genotype and disease diet. Each triangle represents individual gene and LFC amount is represented by thickness of the violin. Key: *** p<0.001, ** p<0.01, * p<0.05, t = 0.05>p<0.1. ns= not significant.

Pathway analysis of DEGs

To determine whether BFCN DEGs linked to biological processes and pathways are selective and/or specific to disease pathogenesis and if MCS ameliorates any of these deficits, we conducted bioinformatic analyses using IPA, KEGG and GO. By IPA, we found 128 neuronal pathways were dysregulated by genotype (Supplemental Table 7) and 148 neuronal pathways were impacted in disease diet (Supplemental Table 8). We queried pathways germane to neurological function (Fig. 5A). MCS effectively reversed dysregulation in several key pathways (seen by z-score) including oxidative phosphorylation, sirtuin signaling, TCA cycle II, synaptic long-term potentiation (LTP), glutamate receptor signaling, superpathway of cholesterol biosynthesis, and synaptic long-term depression (LTD). Not all investigated pathways generated a z-score in IPA to determine directionality (e.g., upregulation and/or downregulation), including mitochondrial dysfunction and GABA receptor signaling (Fig. 5A, Supplemental Tables 78).

Figure 5:

Figure 5:

Bioinformatic assessment of vulnerable pathways in trisomic BFCNs by IPA and KEGG. A. Bar charts show select pathways identified by IPA dysregulated by genotype and treatment, with LFC represented by each bar (2N versus Ts white; Ts versus Ts+ dots), with z-score reflected in coloring (pink upregulated, green downregulated) B. Pathways uniquely identified by KEGG analysis are shown. C. Bar graph showing percent gene expression changes within pathways affected by genotype and diet; red = gene changes unique to genotype, purple = gene changes affected by genotype and diet, blue = genes modified by diet in disease. D. Bar graph shows the number of genes are affected by genotype, diet, or both. E. Cholinergic pathway is dysregulated by genotype and partially rescued by diet, highlighting specific cholinergic receptor subunits and associated proteins. F. Glutamate receptor signaling is dysregulated by genotype and partially rescued by disease diet, with NMDA, AMPA, kainite, and metabotropic receptors showing individual subunit gene expression changes. G. GABA receptor signaling is dysregulated by genotype and modulated by MCS. (C-G, colored red for genotype, blue for MCS, and purple for genes impacted by both genotype and diet).

DEGs by genotype and disease diet were also assessed utilizing KEGG. A total of 43 neuronal pathways were identified by genotype and 37 neuronal pathways by disease diet (Supplemental Tables 9 and 10). Several additional pathways not found by IPA were identified by genotype and disease diet including Thermogenesis, Alzheimer’s disease and Cholinergic synapse (Fig. 5B). Interestingly, the Phosphatidylinositol Signaling system was not altered by genotype, but was significantly affected in disease diet in BFCNs (Fig. 5B). We demonstrate DEGs by genotype (red) or disease diet (blue), with a subset of DEGs affected by both genotype and disease diet (purple; Fig. 5 C, D). DEGs regulated by both genotype and disease diet in specific pathways range from 27–86%, with average gene overlap at 41% (genotype) and 45% (disease diet) (Fig. 5C). Importantly, of the DEGs that are differentially regulated by both genotype and disease diet, MCS reverses the genotype effect in >95% of the genes in these pathways (Table 1). We examined pathways where DEGs differ between genotype and disease diet by individual subunit expression within complexes, which can have profound effects on the determination of upregulation or downregulation. This is especially evident in the cholinergic (Fig. 5E), glutamatergic (Fig. 5F), and GABAergic (Fig. 5G) signaling pathways. Within the cholinergic signaling pathway, in addition to downstream effectors that are modulated independently by genotype or disease diet, the choline transporter (Cht) and Chrm2 are rescued in Ts+ BFCNs, while the cholinergic receptor muscarinic 1 (Chrm1), potassium inwardly rectifying channel subfamily J member 4 (Kir2.3 aka Kcnj4), and potassium inwardly rectifying channel subfamily J member 6 (Kir3.2 aka Kcnj6) receptors, along with the presynaptic Chat and vesicular acetylcholine transporter (VAChT aka Slc18a3) are only significantly changed in disease diet. Conversely, the potassium voltage-gated channel subfamily Q member 5 (Kv7.5 aka Kcnq5) receptor is significantly affected by genotype but not rescued in disease diet (Fig. 5E). Within the Glutamate receptor signaling pathway, NMDA, AMPA, Kainate and mGluRs are regulated by genotype, disease diet, or both, with a numerous DEGs being genotype and disease diet-dependent or genotype-independent and diet dependent (Fig. 5F). GABAergic receptor signaling shows similar findings, with specific changes in extrasynaptic subunits regulating tonic inhibition verses synaptic subunits mediating phasic inhibition of the GABAA pentameric complex differentially regulated by genotype {synaptic alpha1 (Gabra1), alpha3 (Gabra3) and extrasynaptic/synaptic beta2 (Gabrb2) subunits} and disease diet {extrasynaptic delta subunit (Gabrd) or both synaptic (Gabrg2)} (Fig. 5G).

Table 1:

Significant IPA canonical pathways identified by DEGs at (p<0.05).

IPA Canonical Pathway Modulated by genotype alone Genotype and disese diet modulated Modulated in disease diet alone % modulated in genotype reversed in disease diet
Mitochondrial Dysfunction 22 21 17 100.0%
Oxidative Phosphorylation 18 14 7 100.0%
Protein Kinase A Signaling 51 18 37 94.4%
Sirtuin Signaling Pathway 33 20 20 90.0%
TCA Cycle II (Eukaryotic) 7 3 3 100.0%
Synaptogenesis Signaling Pathway 29 23 37 100.0%
Calcium Signaling 27 12 23 100.0%
cAMP-mediated signaling 27 13 23 100.0%
Synaptic Long Term Potentiation 17 8 18 100.0%
14-3-3-mediated Signaling 16 8 15 100.0%
Glutamate Receptor Signaling 11 4 8 100.0%
GABA Receptor Signaling 16 8 11 100.0%
G-Protein Coupled Receptor Signaling 40 29 52 86.2%
Endocannabinoid Neuronal Synapse Pathway 17 9 17 100.0%
Insulin Secretion Signaling Pathway 24 16 31 93.8%
ERK/MAPK Signaling 22 9 20 100.0%
Superpathway of Cholesterol Biosynthesis 4 3 5 66.7%
CREB Signaling in Neurons 51 24 54 92.0%
Synaptic Long Term Depression 22 6 21 100.0%
Type II Diabetes Mellitus Signaling 16 6 17 100.0%

Cholinergic, Glutamatergic, and GABAergic interactomes modulated by MCS

Analysis by genotype and diet was performed on the 2,510 BFCN DEGs which resulted in 2,350 proteins identified by STRING in Cytoscape. Interactions were filtered by key receptors or receptor subunits significantly attenuated by MCS. For the cholinergic pathway, a total of 26 DEGs were found in the Chrm2 interactome (Fig. 6A). Of these 26 protein coding genes, 9 were significantly attenuated by disease diet (34.4%), while 18 (66.6%) DEGs were not significantly attenuated (Fig. 6B). For the glutamatergic pathway, a total of 67 DEGs were found in the Grin2a interactome (Fig. 6C). Of these 67 protein coding genes, 25 (37.3%) were significantly attenuated by disease diet (e.g., phenotypic rescue), while 42 (62.7%) DEGs remained significantly dysregulated (Fig. 6D). For the GABAergic pathway, a total of 54 DEGs were found in the Gabrg2 interactome (Fig. 6E), in which disease diet rescued 20 (37%) of the DEGs, with 34 (63%) not significantly altered in Ts+ (Fig. 6F). When comparing DEGs for supplemented genotype (Ts+ versus 2N+) and diet plus genotype (Ts+ versus 2N), we found very few DEGs were still significantly dysregulated in the Ts+ condition within these interactomes, as Chrm2 had 4 DEGs (15.4%), Grin2a had 16 (24.2%) and Gabrg2 had 16 (29.6%; Supplemental Fig. 3). Therefore, MCS has a profound effect in rescuing aberrant gene expression within key interactomes in multiple pathways relevant to cognition in trisomic mice.

Figure 6:

Figure 6:

STRING analysis of select DEGs in Cytoscape A. Chrm2 was selected from the Cholinergic pathway to identify genotype DEGs within the interactome, with 26 DEGs directly linked the Chmr2 interactome. B. A total of 9 trisomic interactome genes are significantly modulated with 18 DEGs within the interactome not significantly modulated in disease diet. C. Grin2A was selected from the glutamatergic neurotransmission pathway to identify genotype DEGs within the interactome. D. A total of 25 trisomic interactome genes are significantly rescued by disease diet and 42 (of 67) genes were not significantly attenuated by disease diet in the Grin2A glutamatergic interactome. E. From the GABAergic pathway, the Gabrg2 interactome contains 54 DEGs. F. A total of 20 trisomic interactome genes are rescued by MCS in Ts BFCNs and 34 of the DEGs within the Gabrg2 interactome are not significantly attenuated following MCS exposure. Key: pink DEGs, upregulated; green DEGs, downregulated.

MCS effects biological processes beyond pathway analysis

GO analysis examined biological process (BP) dynamics in relation to genotype and disease diet (Table 2). MCS strongly impacted BPs in the categories of RNA modifications, receptor signaling and synaptic functions (Supplemental Tables 1112). Examining GO BPs by genotype and disease diet indicated strong modulation by disease diet on dendritic processes (90% overlap), neurotransmitter processes (89% overlap), and synaptic localization and transport (100% of genotype modulated by disease diet) (Table 2; Supplemental Tables 1112). In contrast, receptor signaling, activity and localization, and signaling pathways have the most processes that are specifically modified by genotype, with MCS having a more moderate effect (Table 2; Supplemental Tables 1112).

Table 2:

Significant Biological Processes linked to DEGs (p<0.05) in GO filtered by keywords.

Filter Term Ts vs 2N unique processes regulated Ts vs 2N overlapping processes regulated Ts+ vs Ts unique processes regulated Ts+ vs Ts overlapping processes regulated
RNA 15 (44%) 19 (56%) 29 (60%) 19 (40%)
Protein Posttranslational modifications 12 (36%) 21 (64%) 17 (45%) 21 (55%)
Protein localization and transport 24 (46%) 28 (54%) 16 (36%) 28 (64%)
Signaling pathways 23 (49%) 24 (51%) 33 (58%) 24 (42%)
Signal transduction 7 (41%) 10 (59%) 7 (41%) 10 (59%)
Receptor signaling, activity and localization 15 (38%) 25 (63%) 37 (60%) 25 (40%)
Dendritic processes 10 (53%) 9 (47%) 1 (10%) 9 (90%)
Axonal processes 4 (21%) 15 (79%) 4 (21%) 15 (79%)
Endo- and Exocytosis processes 5 (29%) 12 (71%) 5 (29%) 12 (71%)
Neurodevelopmental processes 8 (26%) 23 (74%) 7 (23%) 23 (77%)
Neurotransmitter processes 15 (65%) 8 (35%) 1 (11%) 8 (89%)
Synaptic transmission and function 10 (34%) 19 (66%) 20 (51%) 19 (49%)
Synaptic vesicle processes 19 (54%) 16 (46%) 5 (24%) 16 (76%)
Synaptic localization and transport 0 (0%) 10 (100%) 11 (52%) 10 (48%)

MCS benefits memory circuits in trisomic mice

IPA analysis of disease and functions associated with BFCN DEGs revealed several key neurological functions dysregulated in trisomic mice which are rescued by MCS (e.g., disease diet; Fig. 7A). Disease diet shows attenuation of dysregulated gene expression within several IPA-designated neurological functions, including motor dysfunction or movement disorders, ataxia, cognitive impairment and early onset neurological disorders (Fig. 7B). Significant beneficial effects with disease diet are not found for spatial memory impairment or cell death of cortical neurons disrupted by genotype (Fig. 7A). Neurodegeneration is lowered in the disease diet comparison (e.g., Ts+ versus Ts) but does not reach significance by genotype (e.g., Ts versus 2N). In the motor dysfunction or movement disorder category, of the >300 curated genes, only 132 have known functional changes, with 30 trisomic DEGs reversed in disease diet and 1 gene showing additive effects (Fig. 7C, Table 3). Similarly, in the Ataxia category, of the >75 curated genes, 61 have known functional changes, with 14 trisomic DEGs reversed in disease diet and 2 genes showing additive effects in the Cognitive impairment category, of the >250 curated genes, 30 have known functional changes, with 7 trisomic DEGs rescued in disease diet (Fig. 7C, Table 3). Together, these findings suggest MCS rescues some, but not all, circuits or interactomes involved in memory retention in the Ts+ BFCNs.

Figure 7:

Figure 7:

DEGs by genotype and diet were interrogated in IPA for disease and functional correlations. A. Bar chart identifies key disease and behavioral functions that have underlying molecular changes due to genotype (no pattern) or disease diet (striped pattern). Z-scores of changes for genotype and disease diet are colored with green indicating downregulation and pink representing upregulation. B. Violin plots show LFC of DEGs unique to genotype (* red), genotype changes that overlap with disease diet (genotype** purple), disease diet that overlap with genotype (disease diet** purple), and disease diet (* blue) in four representative pathways of neurological function. C. Venn diagrams illustrate within motor dysfunction and movement disorders, cognitive impairment, and ataxia neurological functions for DEGs upregulated or downregulated (up arrow or down arrow) by disease in genotype (red) which are modulated by MCS (purple) and those that are only MCS responsive (blue).

Table 3:

DEGs in Select Disease and Functions (IPA) that were modulated by MCS.

Select Disease and Functions significantly changed in disease and diet p-value Ts vs 2N Activation z-score: Ts vs 2N # Molecules Ts vs 2N p-value Ts+ vs Ts Activation z-score Ts+ vs Ts # MoleculesTs+ vs Ts
Abnormal morphology of neurons 2.54E-18 155 3.84E-14 127
Abnormality of cerebral cortex 1.2E-07 1.663 66 1.92E-07 −1.39 58
Apoptosis of neurons 1.89E-10 2.561 116 2.91E-07 0.908 92
Ataxia 1.56E-09 2.921 85 2.65E-08 −1.568 72
Cellular homeostasis 9.65E-09 −3.875 370 6.7E-12 1.552 336
Cognitive impairment 1.88E-22 −0.232 263 2.28E-26 −0.758 243
Congenital neurological disorder 5.3E-13 0.928 219 1.13E-15 −0.447 201
Dementia 2.42E-08 158 5.82E-07 132
Disorder of basal ganglia 5.81E-31 1.165 283 3.58E-19 −0.482 219
Dominant mental retardation 1.38E-09 51 7.64E-16 57
Dyskinesia 6.88E-26 230 6.95E-18 183
Early-onset neurological disorder 5.86E-09 75 2.86E-07 62
Epilepsy or neurodevelopmental disorder 2.01E-16 1.212 208 4.38E-20 0.377 194
Excitatory postsynaptic potential 3.76E-12 2.268 52 1.72E-11 −0.793 46
Exocytosis 9.86E-08 −1.133 53 1.31E-07 0.268 47
Familial mental retardation 6.73E-12 115 2.03E-15 111
Long-term potentiation 2.64E-15 −0.055 87 1.11E-14 −1.031 77
Mental retardation 2.79E-14 183 1.21E-18 174
Morphogenesis of neurons 2.38E-25 −0.009 204 7.4E-30 −2.774 192
Motor dysfunction or movement disorder 3.82E-36 2.91 390 2.14E-26 −0.849 317
Movement Disorders 1.39E-35 2.53 385 5.18E-26 −0.869 313
Neurodevelopmental disorder 4.31E-15 139 1.69E-16 127
Neurological signs 7.58E-32 1.727 275 2.71E-21 −1.083 217
Neuronal cell death 3.33E-15 1.273 202 1.42E-12 0.693 169
Neurotransmission 7.25E-21 0.603 138 4.96E-21 −0.936 124
Plasticity of synapse 1.64E-12 −0.767 50 3.03E-12 −0.58 45
Potentiation of synapse 1.49E-15 −0.121 88 1.81E-15 −0.885 79
Seizures 1.98E-14 3.084 147 2.68E-12 0.038 124
Synaptic depression 2.42E-09 −0.292 46 2.01E-12 −0.882 47
Synaptic transmission 8.89E-18 1.2 112 1.6E-21 −1.362 108
Cell death of cerebral cortex cells 6.36E-08 1.738 62 ns
Size of cells 1.06E-07 135 ns
Action potential of neurons 1.99E-08 −0.636 43 ns
Excitatory postsynaptic potential of neurons 1.63E-10 28 ns
Endocytosis 3.41E-09 −3.104 140 ns
Memory deficits 6.08E-08 0.88 37 ns
Progressive neurological disorder 6.98E-10 1.219 263 ns
Spatial memory impairment 7.99E-09 28 ns
Alzheimer disease or frontotemporal dementia 3.59E-07 140 ns
Degenerative dementia 3.12E-07 141 ns
Tauopathy 2.67E-07 146 ns
Autophagy ns 1.23E-07 1.005 122
Cell viability of nervous tissue cell lines ns 6.3E-08 2.773 30
Long term synaptic depression of neurons ns 1.38E-07 −1.917 27
Long-term potentiation of hippocampus ns 8.1E-09 0.403 35
Miniature excitatory postsynaptic currents ns 2.04E-07 21
Neurodegeneration ns 4.69E-07 −1.338 72
Polarity of cells ns 1.54E-07 −1.036 23
Size of neurons ns 1.34E-07 30
Transport of vesicles ns 1.52E-07 0.209 35

Validation of MCS responsiveness of select DEGs by qRT-PCR

qPCR shows positive correlations with genes validated previously to be dysregulated by genotype. When re-examined in Ts and Ts+ MSN neurons, the Ts versus 2N DEGs correlate to RNA-seq gene expression LFCs (R2= 0.8673, Fig 8A). Moreover, the disease diet comparison revelated an extremely high correlation for MCS responsive DEGs (R2= 0.9363, Fig. 8A). For example, Chrm2 and Mapk8 were significantly downregulated by genotype (e.g., Ts versus 2N) using the Dream pipeline and correlate with qPCR results, while these genes were positively upregulated by disease diet (e.g., Ts+ versus to Ts) MSNs, which also correlate with qPCR results (Fig 8A, B). Similarly, Grin2a and Camk2a were significantly upregulated by RNA-seq by genotype and significantly downregulated in disease diet, which also correlated with qPCR (Fig. 8A, B). Furthermore, Ngfr was trend level upregulated by Dream pipeline analysis by genotype (p=0.08), correlated with qPCR, and was significantly downregulated by disease diet which also correlated with qPCR product synthesis (Fig. 8A, B).

Figure 8:

Figure 8:

Select DEGs by genotype and diet were validated by qRT-PCR. A. LFC of the means for qPCR synthesis products were correlated with RNA-seq gene expression data for genotype (circles) and disease diet (diamonds). Trend lines were generated to show correlation between qRT-PCR and RNA-seq LFCs. Key: red dashes, genotype; blue dot/dash, disease diet. B. Violin plots show relative gene expression levels for the interrogated genes. Camk2a, Chrm2, Grin2a, and Mapk8 are significantly dysregulated via RNA-seq by genotype and significantly attenuated by MCS in Ts+ MSN BFCNs. Ngfr was trend level dysregulated via RNA-seq by genotype and significantly attenuated by MCS. qPCR validated the directionality of all these DEGs found by RNA-seq. Key: 2N, white; Ts, dark green; Ts+, light green; 2N+, gray.

Discussion

Gene expression profiles and pathway analyses were generated using MSN BFCNs in 6 MO mice testing whether early choline delivery beneficially impacted the vulnerable molecular fingerprint of these septohippocampal circuit neurons. Utilizing LCM, we were able to specifically identify and microisolate individual ChAT-ir neurons within the MSN, generating a specific transcriptomic profile of these BFCNs without confounding signals from glial or other neuronal populations. Previous studies showed BFCN degeneration in Ts65Dn mice initiates approximately at 6 MO (29, 38, 40, 51, 109). Relatively few genes from the ‘DS critical region’ are actually triplicated by expression level in trisomic BFCNs, indicating cell-type specificity for overexpression of triplicated genes. The Dream pipeline identified seven additional significantly upregulated triplicated genes, while three previously identified upregulated triplicated transcripts failed to reach statistical significance (Fig. 4A) (52). Interestingly, two DEGs, Kcnj6 (aka Kir3.2) and Slc5a3, from the DS critical region which have been identified as disease diet dependent, are involved potassium channel signaling (110), with Kcnj6 thought to be expressed and involved at the cholinergic postsynaptic membrane (111). Slc5a3 (encoding SMIT1) increases sensitivity of Kv7.2 and Kv7.3 potassium channels and is inversely affected by cholinergic M1 receptor firing (110). Overall, MCS had a profound beneficial effect on ameliorating the triplicated gene expression, including a trend level attenuation of Dyrk1a, known to be critically important for normal cognitive development, and altered in DS and AD (112, 113) and full reversals of Dnajc28, a part of the DnaJ family of heat shock proteins involved in protein folding and molecular chaperones (114), Brwd1, recently shown to be an epigenomic mediator in the Ts mouse model (115), Ets2, an apoptosis pathway mediator (116), and Itsn1, involved in synaptic vesicle recycling (117). Importantly, rescue of gene expression includes, but is not limited to the DS critical region, indicating MCS results in significant modifications within this vulnerable cell type during this key timepoint of cholinergic degeneration onset. We identified numerous DEGs and rescue of aberrant gene expression by MCS (disease diet) to canonical pathways via bioinformatic inquiry and validated several DEGs by qPCR.

Pathway analysis resulted in several key findings for receptor signaling that are MCS responsive in circuits critical for memory and attention. In the cholinergic system several key gene expression changes are ameliorated in disease diet, including Chrm2, which is dysregulated in AD (118, 119). The potassium voltage gated channels Kir2.3, Kir3.2, and Kv7.5 were impacted by genotype and disease diet, which are thought to enable synaptic plasticity at the postsynaptic membrane (111). Additionally, decreased expression of VAChT is modulated in disease diet BFCNs. Loss of VAChT in mouse models and postmortem human AD has been linked to increased AD pathology, including increased amyloid-beta (Aβ) peptides and tau phosphorylation (120). STRING analysis identified numerous direct interacting partners of Chrm2 that were dysregulated in trisomic mice and rescued or partially rescued by diet, further indicating that early choline delivery a profound effect on normalizing the dysregulation seen in cholinergic synaptic activity at the initiation of BFCN degeneration. While current FDA approved AD therapeutics modulate cholinergic function through inhibiting acetylcholinesterase (AchE), specific targeting of individual receptors for modulation has been difficult (121). Recently, the cholinergic M1 receptor has been examined for therapeutic intervention in schizophrenia (122) and has shown to be dysregulated at the protein level in AD (123). However, the M2 receptor and VAChT have not been targeted for therapeutic intervention, although recent evidence is supportive. Specifically, in a mouse model of AD, chemogenetic-induced increases in Chrm2 and VAChT in the septohippocampal circuit rescued memory impairment (124).

Dysregulation of glutamate receptor signaling has been shown in DS mouse models, with most studies concentrated on hippocampal dysfunction (125127). We show BFCN dysregulation of glutamate receptor signaling is rescued or partially rescued in disease diet. Upregulation of NMDA receptor subunits Grin2a and Grin2b is found in trisomic mice with Grin2a significantly reduced in disease diet, which was corroborated by qRT-PCR, and Grin2b partially attenuated. A recent study showed Grin2A protein expression significantly decreases in the absence of choline, which is further exacerbated in an AD mouse model, suggesting this defect is due to alterations of postsynaptic protein concentrations due to insufficient choline (128). Previous studies have shown differential synaptic and peri- or extra-synaptic localization of these subunits, which can be exchanged by lateral diffusion to finely modulate synaptic strength (129). Perhaps even more intriguing, ratio imbalances between Grin2a and Grin2b increased cognitive decline and activation of Grin2b containing receptors has been linked to AD due to glutamate excitotoxicity (130). AMPA receptor subunits also show differential expression in trisomic mice for glutamate ionotropic receptor AMPA type subunits (Gria1, Gria3, and Gria4) that are essentially not MCS responsive, indicating differential effects of MCS on select NMDA and AMPA subunits, which may partially explain the amelioration of some but not all memory circuits by dietary intervention. However, MCS does significantly decrease Gria2 expression independent of genotype, which may be relevant to AD pathobiology as postmortem human hippocampal neuron studies indicate upregulation of Gria2 in AD and pre-pathology ApoE4 carriers (131, 132). Moreover, gene expression for discs large MAGUK scaffold protein 4 (Dlg4 encoding PSD-95) and discs large MAGUK scaffold protein 3 (Dlg3 encoding SAP-102) were upregulated in trisomic BFCNs and significantly decreased in disease diet. Both of these postsynaptic binding proteins have been implicated in NMDA and AMPA synaptic scaffolding, trafficking and re-localizing for internalization (133). Additionally, attenuation of aberrant gene expression was seen in a multitude of glutamatergic scaffolding proteins (Fig. 6C), including Shank1/2/3, Dlgap3, and Sacm1l (134). The glutamate metabotropic receptor 5 (Grm5) is upregulated in trisomic BFCNs and downregulated by MCS (Fig. 5F), paralleling observations of increased Grm5 in individuals with DS (135). This finding may have translational implications as reduction in GRM5 protein levels induces normal protein synthesis and re-establishes correct levels of LTD, ameliorating learning deficits in a Fragile X mouse model (136). Taken together, these data suggest MCS has a beneficial effect on glutamatergic transmission. We postulate that early choline delivery may exert these effects by protecting against excitotoxicity and rebalancing overall glutamatergic signaling during onset of BFCN degeneration.

GABA-mediated over inhibition has also been observed in DS in mouse models and postmortem human brain (137139) with modulation of GABA receptor subunits proposed for clinical treatment of DS (140). We demonstrate the GABA receptor signaling pathway is vulnerable in trisomic mice and impacted by maternal diet in BFCNs. Previous work has shown choline in the diet is critical for GABAergic transmission with choline supplementation increasing density of benzodiazepine receptor sites, with the mechanism of action likely involving membrane phospholipids (141). Importantly, we found MCS is beneficial for select GABAA receptor subunits dysregulated in the context of DS. Either full or partial rescue was found for several key GABAA receptor subunits, specifically those known to affect subcellular localization and function. The gamma 2 subunit is critical for postsynaptic localization of GABAA receptors (142, 143) and was significantly downregulated in trisomic BFCNs and significantly upregulated by MCS, suggesting higher concentrations of synaptic GABAA receptors. The postsynaptic clustering protein gephyrin (Gphn) was also partially rescued in trisomic BFCNs by MCS. Gphn is the main scaffolding protein at GABAergic postsynaptic sites (144). Concomitantly, a significant decrease in the GABAA delta subunit was seen in the disease diet comparison. GABAA delta subunits are localized to extrasynaptic clusters, mediating tonic inhibition (137, 145). These concurrent changes likely have a significant beneficial effect on the input-output relationship within these vulnerable BFCNs, essentially increasing the ability of the GABAA receptors to facilitate an action potential threshold of synchronized inputs (phasic inhibition) and decrease the tonic inhibition, which reduces the magnitude duration and propagation of excitatory post-synaptic potentials (35).

A comprehensive strategy for treatment of cognitive decline in DS through targeting multiple neurotransmitter systems has been proposed (146). In support of this moderating multiple transmitter systems approach, we show MCS modulates BFCN gene expression not only for specific transcripts but beneficially impacts cholinergic, glutamatergic, and GABAergic pathways. Specifically, we identified and examined three potential receptor-mediated targets for intervention within these pathways, Chrm2, Grin2a, and Gabrg2. These key targets are modulated by MCS and may also reflect the central moderators of an excitatory/inhibitory (E/I) ratio imbalance that has been postulated to underlie BFCN degeneration and has been found in Ts65Dn mice within multiple brain regions (27, 28, 147, 148). We postulate that MCS, previously shown to modulate the PEMT pathway and increase phosphocholine metabolites (66), normalizes expression of these phospholipids in trisomic BFCNs, and may stabilize the postsynaptic membrane for proper synaptic function and expression of both excitatory and inhibitory receptors. We show MCS beneficially impacts these multiple synaptic and neurotransmitter pathways. Notably, multiple markers of synaptic LTP and LTD are normalized in the disease diet comparison, suggesting MCS rebalances the E/I ratio within trisomic BFCNs, which in turn may protect the septohippocampal circuit.

GO analysis identified key biological processes involved in neurodevelopment, synaptic localization and transport, protein post-translational modification and receptor signaling, activity and localization. IPA analysis identified neurological disease and function changes including motor dysfunction and memory deficits, cognitive impairment and ataxia all of which showed lifelong (e.g., up to 6 MO) modulation by MCS. These disease and functions also implicated the Chrm2, Grin2a, and Gabrg2 subunits and their interactomes (Figs, 6, 7). Therefore, benefits conferred by MCS may underlie well-established spatial and attentional behaviors seen in Ts65Dn offspring (41, 43, 46, 59, 67, 74). Aggregating pathway findings mechanistically, these data suggest MCS provides organizational changes that positively benefit attention, spatial memory, and both short and long-term recognition memory improvements in this DS mouse model which has been corroborated by previous behavioral studies by our collaborative group (4143, 46, 70). By identifying and targeting select gene expression and pathway changes, we may isolate novel targets for therapeutic intervention and modulation for translation to DS and possibly AD.

While we strive to eliminate extraneous variables in the data, there remain limitations that may affect outcomes. These include variability in RNA quality, although differences due to genotype is unlikely (75, 76, 78, 79) and RNA quantity was normalized during bioinformatic analysis. It is possible that choline supplementation may also have unintended negative effects on developing pups or on gene expression within the brain, although we find this unlikely as multiple studies from our lab and other independent groups demonstrated the benefits of MCS using a variety of paradigms including RNA-seq, qPCR, immunocytochemistry, and behavioral testing (45, 59, 62, 67, 75, 76). Additionally, choline is a water soluble, essential nutrient, meaning it is unlikely to lead to toxic levels within the body (45, 67, 149). We limited this study to male mice, and there may exist sex differences in BFCN degenerative programs (30). BFCN transcriptomic analyses in female trisomic mice are in progress. However, mixed sex studies have not previously revealed significant differences gene expression or MCS effects in hippocampal CA1 pyramidal neuron studies (75, 76, 78). With IPA analysis of canonical pathways and neurological disease and memory circuit changes, not all of the DEGs identified can be classified as upregulated or downregulated, due to a lack of, or confounding evidence, in the literature (150). However, we found BFCN gene expression level changes showed a direct correlation between the degenerating septohippocampal circuit and behavioral perturbations in the trisomic animal model (41, 42, 45, 46, 74). While we utilized the Ts65Dn mouse model, as previously done in MCS behavioral studies (4143, 45, 70), several additional DS mouse models exist that may reflect differences in gene expression or effects of MCS, as they vary in the numbers of triplicated HSA21 orthologs (151154). We will consider evaluating MCS in the context of septohippocampal circuit neuroprotection utilizing alternative models of DS and AD pathobiology as part of future studies.

Future assessments will also include comparing gene and pathway changes due to MCS on the entire septohippocampal connectome, including GABAergic projection neurons within the MSN that project to hippocampus (155, 156) and CA1 hippocampal neurons in the same animals. Interrogation of astrocyte, oligodendrocyte and microglial populations are beyond the scope of this work, but may show significant effects, as seen in an AD mouse model following early choline delivery (73).

Conclusions

Through single population RNA-seq we have gene and pathway evidence for concomitant cholinergic, glutamatergic, and GABAergic dysfunction within trisomic MSN BFCNs at the onset of degeneration. We demonstrate dysregulation of a complex interconnected signaling relationship between cholinergic, glutamatergic, and GABAergic signaling that is rescued or partially rescued by MCS in trisomic mice. We also identify novel gene expression changes within vulnerable BFCNs that could be used for therapeutic intervention and modulation for DS and AD pathology within the septohippocampal circuit that may have organizational neuroprotective effects on the brain in both neurodevelopmental and neurodegenerative contexts.

Supplementary Material

Fig S1
Fig S2
Fig S3
Tab S1
Tab S2
Tab S3
Tab S4
Tab S5
Tab S6
Tab S7
Tab S8
Tab S9
Tab S10
Tab S11
Tab S12

Highlights:

  • Single population LCM generates RNA-seq DEGs from spatially characterized cholinergic neurons

  • Dream analysis identifies significantly higher number of DEGs compared to limma-voom

  • MCS significantly attenuates many of the dysregulated DEGs in Ts BFCNs

  • DEGs attenuated by MCS underlie the regulation of multiple pathways and processes of disease, including behavioral functions

  • Cholinergic, Glutamatergic and GABAergic synaptic pathways show normalization by MCS, likely modulating E/I ratio in Ts BFCNs

Acknowledgments

We thank Arthur Saltzman, M.S., Gabriel E. Hoffman Ph.D., Amanda Labuza, Ph.D, and Paul Zappile, M.S. for expert technical assistance.

Funding Statement

Funding was provided by support from grants AG014449, AG017617, AG072599, AG074004, and AG077103 from the National Institutes of Health.

Declarations

Animal protocols were approved by the Nathan Kline Institute/NYUGSOM IACUC in accordance with NIH guidelines.

Conflict of Interest Statement

The authors reported no biomedical financial interests or potential conflicts of interest.

Data Availability statement

Included in article: The data that support the findings of this study are available in the Methods and/or Supplementary Material of this article. Any study data are available on request from the corresponding author. RNA-seq data analyzed within this study are available from the corresponding author upon request.

References

  • 1.Parker SE, Mai CT, Canfield MA, Rickard R, Wang Y, Meyer RE, Anderson P, Mason CA, Collins JS, Kirby RS, and Correa A (2010) Updated National Birth Prevalence estimates for selected birth defects in the United States, 2004–2006. Birth Defects Res A Clin Mol Teratol 88, 1008–1016 [DOI] [PubMed] [Google Scholar]
  • 2.Mai CT, Isenburg JL, Canfield MA, Meyer RE, Correa A, Alverson CJ, Lupo PJ, Riehle-Colarusso T, Cho SJ, Aggarwal D, Kirby RS, and National Birth Defects Prevention, N. (2019) National population-based estimates for major birth defects, 2010–2014. Birth Defects Res 111, 1420–1435 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Mann DM, Yates PO, and Marcyniuk B (1984) 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 10, 185–207 [DOI] [PubMed] [Google Scholar]
  • 4.Coyle J ML O-G;Reeves RH; Gearhart JD (1988) Down syndrome, Alzheimer’s disease and the trisomy16 mouse. Trends Neurosci. 11, 390–394 [DOI] [PubMed] [Google Scholar]
  • 5.Hook EB (1989) Issues pertaining to the impact and etiology of trisomy 21 and other aneuploidy in humans; a consideration of evolutionary implications, maternal age mechanisms, and other matters. Prog Clin Biol Res 311, 1–27 [PubMed] [Google Scholar]
  • 6.Hodgkins PS, Prasher V, Farrar G, Armstrong R, Sturman S, Corbett J, and Blair JA (1993) Reduced transferrin binding in Down syndrome: a route to senile plaque formation and dementia. Neuroreport 5, 21–24 [DOI] [PubMed] [Google Scholar]
  • 7.Roth GM, Sun B, Greensite FS, Lott IT, and Dietrich RB (1996) Premature aging in persons with Down syndrome: MR findings. AJNR Am J Neuroradiol 17, 1283–1289 [PMC free article] [PubMed] [Google Scholar]
  • 8.Chapman RS, and Hesketh LJ (2000) Behavioral phenotype of individuals with Down syndrome. Ment Retard Dev Disabil Res Rev 6, 84–95 [DOI] [PubMed] [Google Scholar]
  • 9.Cataldo AM, Peterhoff CM, Troncoso JC, Gomez-Isla T, Hyman BT, and Nixon RA (2000) 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 157, 277–286 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Hartley D, Blumenthal T, Carrillo M, DiPaolo G, Esralew L, Gardiner K, Granholm AC, Iqbal K, Krams M, Lemere C, Lott I, Mobley W, Ness S, Nixon R, Potter H, Reeves R, Sabbagh M, Silverman W, Tycko B, Whitten M, and Wisniewski T (2015) Down syndrome and Alzheimer’s disease: Common pathways, common goals. Alzheimers Dement 11, 700–709 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Hartley SL, Handen BL, Devenny DA, Hardison R, Mihaila I, Price JC, Cohen AD, Klunk WE, Mailick MR, Johnson SC, and Christian BT (2014) Cognitive functioning in relation to brain amyloid-beta in healthy adults with Down syndrome. Brain 137, 2556–2563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Leverenz JB, and Raskind MA (1998) Early amyloid deposition in the medial temporal lobe of young Down syndrome patients: a regional quantitative analysis. Exp. Neurol 150, 296–304 [DOI] [PubMed] [Google Scholar]
  • 13.Wisniewski KE, Dalton AJ, Crapper McLachlan DR, Wen GY, and Wisniewski HM (1985) Alzheimer’s disease in Down’s syndrome: clinicopathologic studies. Neurology 35, 957–961 [DOI] [PubMed] [Google Scholar]
  • 14.Lai F, and Williams RS (1989) A prospective study of Alzheimer disease in Down syndrome. Arch Neurol 46, 849–853 [DOI] [PubMed] [Google Scholar]
  • 15.Perez SE, Miguel JC, He B, Malek-Ahmadi M, Abrahamson EE, Ikonomovic MD, Lott I, Doran E, Alldred MJ, Ginsberg SD, and Mufson EJ (2019) Frontal cortex and striatal cellular and molecular pathobiology in individuals with Down syndrome with and without dementia. Acta Neuropathol 137, 413–436 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Videla L, Benejam B, Pegueroles J, Carmona-Iragui M, Padilla C, Fernández S, Barroeta I, Altuna M, Valldeneu S, Garzón D, Ribas L, Montal V, Arranz Martínez J, Rozalem Aranha M, Alcolea D, Bejanin A, Iulita MF, Videla Cés S, Blesa R, Lleó A, and Fortea J (2022) Longitudinal Clinical and Cognitive Changes Along the Alzheimer Disease Continuum in Down Syndrome. JAMA Netw Open 5, e2225573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hill DA, Gridley G, Cnattingius S, Mellemkjaer L, Linet M, Adami HO, Olsen JH, Nyren O, and Fraumeni JF Jr. (2003) Mortality and cancer incidence among individuals with Down syndrome. Arch Intern Med 163, 705–711 [DOI] [PubMed] [Google Scholar]
  • 18.Bittles AH, Bower C, Hussain R, and Glasson EJ (2007) The four ages of Down syndrome. Eur J Public Health 17, 221–225 [DOI] [PubMed] [Google Scholar]
  • 19.Presson AP, Partyka G, Jensen KM, Devine OJ, Rasmussen SA, McCabe LL, and McCabe ER (2013) Current estimate of Down Syndrome population prevalence in the United States. J Pediatr 163, 1163–1168 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dick MB, Doran E, Phelan M, and Lott IT (2015) Cognitive profiles on the severe impairment battery are similar in Alzheimer disease and Down syndrome with dementia. Alzheimer Dis Assoc Disord [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hithersay R, Startin CM, Hamburg S, Mok KY, Hardy J, Fisher EMC, Tybulewicz VLJ, Nizetic D, and Strydom A (2019) Association of Dementia With Mortality Among Adults With Down Syndrome Older Than 35 Years. JAMA Neurol 76, 152–160 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sendera TJ, Ma SY, Jaffar S, Kozlowski PB, Kordower JH, Mawal Y, Saragovi HU, and Mufson EJ (2000) Reduction in TrkA-immunoreactive neurons is not associated with an overexpression of galaninergic fibers within the nucleus basalis in Down’s syndrome. J Neurochem 74, 1185–1196. [DOI] [PubMed] [Google Scholar]
  • 23.Mufson EJ, Bothwell M, and Kordower JH (1989) Loss of nerve growth factor receptor-containing neurons in Alzheimer’s disease: a quantitative analysis across subregions of the basal forebrain. Exp Neurol 105, 221–232 [DOI] [PubMed] [Google Scholar]
  • 24.Mann DM, Yates PO, Marcyniuk B, and Ravindra CR (1986) The topography of plaques and tangles in Down’s syndrome patients of different ages. Neuropathol. App. Neurobiol 12, 447–457 [DOI] [PubMed] [Google Scholar]
  • 25.Wegiel J, Flory M, Kuchna I, Nowicki K, Wegiel J, Ma SY, Zhong N, Bobrowicz TW, de Leon M, Lai F, Silverman WP, and Wisniewski T (2022) Developmental deficits and staging of dynamics of age associated Alzheimer’s disease neurodegeneration and neuronal loss in subjects with Down syndrome. Acta Neuropathol Commun 10, 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Richter N, David LS, Grothe MJ, Teipel S, Dietlein M, Tittgemeyer M, Neumaier B, Fink GR, Onur OA, and Kukolja J (2022) Age and Anterior Basal Forebrain Volume Predict the Cholinergic Deficit in Patients with Mild Cognitive Impairment due to Alzheimer’s Disease. J Alzheimers Dis 86, 425–440 [DOI] [PubMed] [Google Scholar]
  • 27.Belichenko PV, Kleschevnikov AM, Masliah E, Wu C, Takimoto-Kimura R, Salehi A, and Mobley WC (2009) Excitatory-inhibitory relationship in the fascia dentata in the Ts65Dn mouse model of Down syndrome. J Comp Neurol 512, 453–466 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Belichenko PV, Masliah E, Kleschevnikov AM, Villar AJ, Epstein CJ, Salehi A, and Mobley WC (2004) Synaptic structural abnormalities in the Ts65Dn mouse model of Down Syndrome. J Comp Neurol 480, 281–298 [DOI] [PubMed] [Google Scholar]
  • 29.Granholm AC, Sanders LA, and Crnic LS (2000) Loss of cholinergic phenotype in basal forebrain coincides with cognitive decline in a mouse model of Down’s syndrome. Exp Neurol 161, 647–663 [DOI] [PubMed] [Google Scholar]
  • 30.Kelley CM, Powers BE, Velazquez R, Ash JA, Ginsberg SD, Strupp BJ, and Mufson EJ (2014) Sex differences in the cholinergic basal forebrain in the Ts65Dn mouse model of Down syndrome and Alzheimer’s disease. Brain Pathol 24, 33–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kelley CM, Powers BE, Velazquez R, Ash JA, Ginsberg SD, Strupp BJ, and Mufson EJ (2014) Maternal choline supplementation differentially alters the basal forebrain cholinergic system of young-adult Ts65Dn and disomic mice. J Comp Neurol 522, 1390–1410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cooper JD, Salehi A, Delcroix JD, Howe CL, Belichenko PV, Chua-Couzens J, Kilbridge JF, Carlson EJ, Epstein CJ, and Mobley WC (2001) 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 98, 10439–10444 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sans-Dublanc A, Razzauti A, Desikan S, Pascual M, Monyer H, and Sindreu C (2020) Septal GABAergic inputs to CA1 govern contextual memory retrieval. Sci Adv 6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fernandez F, Morishita W, Zuniga E, Nguyen J, Blank M, Malenka RC, and Garner CC (2007) Pharmacotherapy for cognitive impairment in a mouse model of Down syndrome. Nat Neurosci 10, 411–413 [DOI] [PubMed] [Google Scholar]
  • 35.Contestabile A, Magara S, and Cancedda L (2017) The GABAergic hypothesis for cognitive disabilities in Down syndrome. Front Cell Neurosci 11, 54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Givens BS, and Olton DS (1990) Cholinergic and GABAergic modulation of medial septal area: effect on working memory. Behav Neurosci 104, 849–855 [DOI] [PubMed] [Google Scholar]
  • 37.Mesulam MM, Mufson EJ, Wainer BH, and Levey AI (1983) Central cholinergic pathways in the rat: an overview based on an alternative nomenclature (Ch1-Ch6). Neuroscience 10, 1185–1201 [DOI] [PubMed] [Google Scholar]
  • 38.Lockrow J, Prakasam A, Huang P, Bimonte-Nelson H, Sambamurti K, and Granholm AC (2009) Cholinergic degeneration and memory loss delayed by vitamin E in a Down syndrome mouse model. Exp Neurol 216, 278–289 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Hunter CL, Bimonte-Nelson HA, Nelson M, Eckman CB, and Granholm AC (2004) Behavioral and neurobiological markers of Alzheimer’s disease in Ts65Dn mice: effects of estrogen. Neurobiol Aging 25, 873–884 [DOI] [PubMed] [Google Scholar]
  • 40.Holtzman DM, Santucci D, Kilbridge J, Chua-Couzens J, Fontana DJ, Daniels SE, Johnson RM, Chen K, Sun Y, Carlson E, Alleva E, Epstein CJ, and Mobley WC (1996) Developmental abnormalities and age-related neurodegeneration in a mouse model of Down syndrome. Proc Natl Acad Sci USA 93, 13333–13338 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Powers BE, Kelley CM, Velazquez R, Ash JA, Strawderman MS, Alldred MJ, Ginsberg SD, Mufson EJ, and Strupp BJ (2017) Maternal choline supplementation in a mouse model of Down syndrome: Effects on attention and nucleus basalis/substantia innominata neuron morphology in adult offspring. Neuroscience 340, 501–514 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Powers BE, Velazquez R, Kelley CM, Ash JA, Strawderman MS, Alldred MJ, Ginsberg SD, Mufson EJ, and Strupp BJ (2016) Attentional function and basal forebrain cholinergic neuron morphology during aging in the Ts65Dn mouse model of Down syndrome. Brain Struct Funct 221, 4337–4352 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Ash JA, Velazquez R, Kelley CM, Powers BE, Ginsberg SD, Mufson EJ, and Strupp BJ (2014) Maternal choline supplementation improves spatial mapping and increases basal forebrain cholinergic neuron number and size in aged Ts65Dn mice. Neurobiol Dis 70, 32–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kelley CM, Ash JA, Powers BE, Velazquez R, Alldred MJ, Ikonomovic MD, Ginsberg SD, Strupp BJ, and Mufson EJ (2016) Effects of maternal choline supplementation on the septohippocampal cholinergic system in the Ts65Dn mouse model of Down syndrome. Curr Alzheimer Res 13, 84–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Strupp BJ, Powers BE, Velazquez R, Ash JA, Kelley CM, Alldred MJ, Strawderman M, Caudill MA, Mufson EJ, and Ginsberg SD (2016) Maternal choline supplementation: a potential prenatal treatment for Down syndrome and Alzheimer’s disease. Curr Alzheimer Res 13, 97–106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Moon J, Chen M, Gandhy SU, Strawderman M, Levitsky DA, Maclean KN, and Strupp BJ (2010) Perinatal choline supplementation improves cognitive functioning and emotion regulation in the Ts65Dn mouse model of Down syndrome. Behav Neurosci 124, 346–361 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hunter CL, Bimonte HA, and Granholm A-CE (2003) Behavioral comparison of 4 and 6 month-old Ts65Dn mice: Age-related impairments in working and reference memory. Behavioural Brain Research 138, 121–131 [DOI] [PubMed] [Google Scholar]
  • 48.Kelley CM, Ginsberg SD, Alldred MJ, Strupp BJ, and Mufson EJ (2019) Maternal Choline Supplementation Alters Basal Forebrain Cholinergic Neuron Gene Expression in the Ts65Dn Mouse Model of Down Syndrome. Dev Neurobiol 79, 664–683 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Cataldo AM, Petanceska S, Peterhoff CM, Terio NB, Epstein CJ, Villar A, Carlson EJ, Staufenbiel M, and Nixon RA (2003) App gene dosage modulates endosomal abnormalities of Alzheimer’s disease in a segmental trisomy 16 mouse model of Down syndrome. J Neurosci 23, 6788–6792 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hunter CL, Isacson O, Nelson M, Bimonte-Nelson H, Seo H, Lin L, Ford K, Kindy MS, and Granholm A-C (2003) Regional alterations in amyloid precursor protein and nerve growth factor across age in a mouse model of Down’s syndrome. Neuroscience Research 45, 437–445 [DOI] [PubMed] [Google Scholar]
  • 51.Contestabile A, Fila T, Bartesaghi R, and Ciani E (2006) Choline acetyltransferase activity at different ages in brain of Ts65Dn mice, an animal model for Down’s syndrome and related neurodegenerative diseases. J Neurochem 97, 515–526 [DOI] [PubMed] [Google Scholar]
  • 52.Alldred MJ, Penikalapati SC, Lee SH, Heguy A, Roussos P, and Ginsberg SD (2021) Profiling Basal Forebrain Cholinergic Neurons Reveals a Molecular Basis for Vulnerability Within the Ts65Dn Model of Down Syndrome and Alzheimer’s Disease. Molecular neurobiology 58, 5141–5162 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Davenport C, Yan J, Taesuwan S, Shields K, West AA, Jiang X, Perry CA, Malysheva OV, Stabler SP, Allen RH, and Caudill MA (2015) Choline intakes exceeding recommendations during human lactation improve breast milk choline content by increasing PEMT pathway metabolites. J Nutr Biochem 26, 903–911 [DOI] [PubMed] [Google Scholar]
  • 54.Ganz AB, Cohen VV, Swersky CC, Stover J, Vitiello GA, Lovesky J, Chuang JC, Shields K, Fomin VG, Lopez YS, Mohan S, Ganti A, Carrier B, Malysheva OV, and Caudill MA (2017) Genetic variation in choline-metabolizing enzymes alters choline metabolism in young women consuming choline intakes meeting current recommendations. Int J Mol Sci 18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Caudill MA, Strupp BJ, Muscalu L, Nevins JEH, and Canfield RL (2018) Maternal choline supplementation during the third trimester of pregnancy improves infant information processing speed: a randomized, double-blind, controlled feeding study. FASEB J 32, 2172–2180 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Zeisel SH (2013) Nutrition in pregnancy: the argument for including a source of choline. Int J Womens Health 5, 193–199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Gwee MC, and Sim MK (1978) Free choline concentration and cephalin-N-methyltransferase activity in the maternal and foetal liver and placenta of pregnant rats. Clin Exp Pharmacol Physiol 5, 649–653 [DOI] [PubMed] [Google Scholar]
  • 58.Institute of Medicine, N. A. o. S. U. (1998) Dietary reference intakes for thiamin, riboflavin, niacin, vitamin B6, folate, vitamin B12, pantothenic acid, biotin and choline, National Academy Press, Washington, DC: [PubMed] [Google Scholar]
  • 59.Meck WH, Smith RA, and Williams CL (1989) Organizational changes in cholinergic activity and enhanced visuospatial memory as a function of choline administered prenatally or postnatally or both. Behav Neurosci 103, 1234–1241 [DOI] [PubMed] [Google Scholar]
  • 60.Zeisel SH, Klatt KC, and Caudill MA (2018) Choline. Adv Nutr 9, 58–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Blusztajn JK (1998) Choline, a vital amine. Science 281, 794–795 [DOI] [PubMed] [Google Scholar]
  • 62.Blusztajn JK, and Mellott TJ (2013) Neuroprotective actions of perinatal choline nutrition. Clin Chem Lab Med 51, 591–599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.McGowan PO, Meaney MJ, and Szyf M (2008) Diet and the epigenetic (re)programming of phenotypic differences in behavior. Brain Res 1237, 12–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Kwan STC, King JH, Grenier JK, Yan J, Jiang X, Roberson MS, and Caudill MA (2018) Maternal choline supplementation during normal murine pregnancy alters the placental epigenome: results of an exploratory study. Nutrients 10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Blusztajn JK, Slack BE, and Mellott TJ (2017) Neuroprotective actions of dietary choline. Nutrients 9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Yan J, Ginsberg SD, Powers B, Alldred MJ, Saltzman A, Strupp BJ, and Caudill MA (2014) Maternal choline supplementation programs greater activity of the phosphatidylethanolamine N-methyltransferase (PEMT) pathway in adult Ts65Dn trisomic mice. FASEB J 28, 4312–4323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Meck WH, Smith RA, and Williams CL (1988) Pre- and postnatal choline supplementation produces long-term facilitation of spatial memory. Dev Psychobiol 21, 339–353 [DOI] [PubMed] [Google Scholar]
  • 68.Jiang X, Yan J, West AA, Perry CA, Malysheva OV, Devapatla S, Pressman E, Vermeylen F, and Caudill MA (2012) Maternal choline intake alters the epigenetic state of fetal cortisol-regulating genes in humans. FASEB J 26, 3563–3574 [DOI] [PubMed] [Google Scholar]
  • 69.Korsmo HW, Jiang X, and Caudill MA (2019) Choline: Exploring the Growing Science on Its Benefits for Moms and Babies. Nutrients 11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Powers BE, Velazquez R, Strawderman MS, Ginsberg SD, Mufson EJ, and Strupp BJ (2021) Maternal Choline Supplementation as a Potential Therapy for Down Syndrome: Assessment of Effects Throughout the Lifespan. Front Aging Neurosci 13, 723046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Taesuwan S, McDougall MQ, Malysheva OV, Bender E, Nevins JEH, Devapatla S, Vidavalur R, Caudill MA, and Klatt KC (2021) Choline metabolome response to prenatal choline supplementation across pregnancy: A randomized controlled trial. Faseb j 35, e22063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Warton FL, Molteno CD, Warton CMR, Wintermark P, Lindinger NM, Dodge NC, Zöllei L, van der Kouwe AJW, Carter RC, Jacobson JL, Jacobson SW, and Meintjes EM (2021) Maternal choline supplementation mitigates alcohol exposure effects on neonatal brain volumes. Alcohol Clin Exp Res 45, 1762–1774 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Velazquez R, Ferreira E, Knowles S, Fux C, Rodin A, Winslow W, and Oddo S (2019) Lifelong choline supplementation ameliorates Alzheimer’s disease pathology and associated cognitive deficits by attenuating microglia activation. Aging Cell 18, e13037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Velazquez R, Ash JA, Powers BE, Kelley CM, Strawderman M, Luscher ZI, Ginsberg SD, Mufson EJ, and Strupp BJ (2013) Maternal choline supplementation improves spatial learning and adult hippocampal neurogenesis in the Ts65Dn mouse model of Down syndrome. Neurobiol Dis 58, 92–101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Alldred MJ, Chao HM, Lee SH, Beilin J, Powers BE, Petkova E, Strupp BJ, and Ginsberg SD (2018) CA1 pyramidal neuron gene expression mosaics in the Ts65Dn murine model of Down syndrome and Alzheimer’s disease following maternal choline supplementation. Hippocampus 28, 251–268 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Alldred MJ, Chao HM, Lee SH, Beilin J, Powers BE, Petkova E, Strupp BJ, and Ginsberg SD (2019) Long-term effects of maternal choline supplementation on CA1 pyramidal neuron gene expression in the Ts65Dn mouse model of Down syndrome and Alzheimer’s disease. Faseb j 33, 9871–9884 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Bahnfleth CL, Strupp BJ, Caudill MA, and Canfield RL (2022) Prenatal choline supplementation improves child sustained attention: A 7-year follow-up of a randomized controlled feeding trial. Faseb j 36, e22054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Alldred MJ, Lee SH, Petkova E, and Ginsberg SD (2015b) Expression profile analysis of hippocampal CA1 pyramidal neurons in aged Ts65Dn mice, a model of Down syndrome (DS) and Alzheimer’s disease (AD). Brain Struct Funct 220, 2983–2996 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Alldred MJ, Lee SH, Petkova E, and Ginsberg SD (2015a) Expression profile analysis of vulnerable CA1 pyramidal neurons in young-middle-aged Ts65Dn mice. J Comp Neurol 523, 61–74 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Detopoulou P, Panagiotakos DB, Antonopoulou S, Pitsavos C, and Stefanadis C (2008) Dietary choline and betaine intakes in relation to concentrations of inflammatory markers in healthy adults: the ATTICA study. Am J Clin Nutr 87, 424–430 [DOI] [PubMed] [Google Scholar]
  • 81.Duchon A, Raveau M, Chevalier C, Nalesso V, Sharp AJ, and Herault Y (2011) Identification of the translocation breakpoints in the Ts65Dn and Ts1Cje mouse lines: relevance for modeling Down syndrome. Mamm Genome 22, 674–684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Andrews S (2010) FastQC: A quality control tool for high throughput sequence data.
  • 83.Bolger AM, Lohse M, and Usadel B (2014) Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics (Oxford, England) 30, 2114–2120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, and Gingeras TR (2013) STAR: ultrafast universal RNA-seq aligner. Bioinformatics (Oxford, England) 29, 15–21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.(2019) Picard Toolkit. Broad Institute, Github Repository [Google Scholar]
  • 86.Li B, and Dewey CN (2011) RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12, 323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Robinson MD, Oshlack A A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol 11, R25 (2010). 10.1186/gb-2010-11-3-r25 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Wang L, Wang S, and Li W (2012) RSeQC: quality control of RNA-seq experiments. Bioinformatics (Oxford, England) 28, 2184–2185 [DOI] [PubMed] [Google Scholar]
  • 89.Robinson MD, McCarthy DJ, and Smyth GK (2010) edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics (Oxford, England) 26, 139–140 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Zehetmayer S, Posch M, and Graf A (2022) Impact of adaptive filtering on power and false discovery rate in RNA-seq experiments. BMC Bioinformatics 23, 388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Rau A, Gallopin M, Celeux G, and Jaffrézic F (2013) Data-based filtering for replicated high-throughput transcriptome sequencing experiments. Bioinformatics (Oxford, England) 29, 2146–2152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hoffman GE, and Roussos P (2021) Dream: powerful differential expression analysis for repeated measures designs. Bioinformatics (Oxford, England) 37, 192–201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hoffman GE, and Schadt EE (2016) variancePartition: interpreting drivers of variation in complex gene expression studies. BMC Bioinformatics 17, 483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pages HCM, Falcon S; Li N (2019) AnnotationDbi: Manipulation of SQLite-based annotations in bioconductor.
  • 95.Broberg P (2005) A comparative review of estimates of the proportion unchanged genes and the false discovery rate. BMC Bioinformatics 6, 199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Qiagen. (2020) https://www.qiagenbioinformatics.com/products/ingenuity-pathway-analysis.
  • 97.Krämer A, Green J, Pollard J Jr, and Tugendreich S (2013) Causal analysis approaches in Ingenuity Pathway Analysis. Bioinformatics (Oxford, England) 30, 523–530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kanehisa M, and Goto S (2000) KEGG: kyoto encyclopedia of genes and genomes. Nucleic acids research 28, 27–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski K, Dwight SS, Eppig JT, Harris MA, Hill DP, Issel-Tarver L, Kasarskis A, Lewis S, Matese JC, Richardson JE, Ringwald M, Rubin GM, and Sherlock G (2000) Gene ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 25, 25–29 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.(2021) The Gene Ontology resource: enriching a GOld mine. Nucleic acids research 49, D325–d334 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, Jensen LJ, and Mering Christian v. (2018) STRING v11: protein–protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic acids research 47, D607–D613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, and Ideker T (2003) Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome research 13, 2498–2504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ginsberg SD, and Che S (2004) Combined histochemical staining, RNA amplification, regional, and single cell cDNA analysis within the hippocampus. Lab Invest 84, 952–962 [DOI] [PubMed] [Google Scholar]
  • 104.Alldred MJ, Lee SH, and Ginsberg SD (2021) Adiponectin Modulation by Genotype and Maternal Choline Supplementation in a Mouse Model of Down Syndrome and Alzheimer’s Disease. J Clin Med 10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Alldred MJ, Lee SH, Stutzmann GE, and Ginsberg SD (2021) Oxidative Phosphorylation Is Dysregulated Within the Basocortical Circuit in a 6-month old Mouse Model of Down Syndrome and Alzheimer’s Disease. Front Aging Neurosci 13, 707950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ginsberg SD, Alldred MJ, Counts SE, Cataldo AM, Neve RL, Jiang Y, Wuu J, Chao MV, Mufson EJ, Nixon RA, and Che S (2010) Microarray analysis of hippocampal CA1 neurons implicates early endosomal dysfunction during Alzheimer’s disease progression. Biol Psychiatry 68, 885–893 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Jiang Y, Mullaney KA, Peterhoff CM, Che S, Schmidt SD, Boyer-Boiteau A, Ginsberg SD, Cataldo AM, Mathews PM, and Nixon RA (2010) Alzheimer’s-related endosome dysfunction in Down syndrome is Abeta-independent but requires APP and is reversed by BACE-1 inhibition. Proc Natl Acad Sci USA 107, 1630–1635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.ABI. (2004) Guide to Performing Relative Quantitation of Gene Expression Using Real-Time Quantitative PCR. Applied Biosystems Product Guide, 1–60 [Google Scholar]
  • 109.Chen Y, Dyakin VV, Branch CA, Ardekani B, Yang D, Guilfoyle DN, Peterson J, Peterhoff C, Ginsberg SD, Cataldo AM, and Nixon RA (2009) In vivo MRI identifies cholinergic circuitry deficits in a Down syndrome model. Neurobiol Aging 30, 1453–1465 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Dai G, Yu H, Kruse M, Traynor-Kaplan A, and Hille B (2016) Osmoregulatory inositol transporter SMIT1 modulates electrical activity by adjusting PI(4,5)P2 levels. Proc Natl Acad Sci U S A 113, E3290–3299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Grosse G, Draguhn A, Höhne L, Tapp R, Veh RW, and Ahnert-Hilger G (2000) Expression of Kv1 potassium channels in mouse hippocampal primary cultures: development and activity-dependent regulation. J Neurosci 20, 1869–1882 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Liu F, Liang Z, Wegiel J, Hwang YW, Iqbal K, Grundke-Iqbal I, Ramakrishna N, and Gong CX (2008) Overexpression of Dyrk1A contributes to neurofibrillary degeneration in Down syndrome. FASEB J 22, 3224–3233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Ferrer I, Barrachina M, Puig B, Martinez de Lagran M, Marti E, Avila J, and Dierssen M (2005) Constitutive Dyrk1A is abnormally expressed in Alzheimer disease, Down syndrome, Pick disease, and related transgenic models. Neurobiol Dis 20, 392–400 [DOI] [PubMed] [Google Scholar]
  • 114.Zarouchlioti C, Parfitt DA, Li W, Gittings LM, and Cheetham ME (2018) DNAJ Proteins in neurodegeneration: essential and protective factors. Philos Trans R Soc Lond B Biol Sci 373 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Fulton SL, Wenderski W, Lepack AE, Eagle AL, Fanutza T, Bastle RM, Ramakrishnan A, Hays EC, Neal A, Bendl J, Farrelly LA, Al-Kachak A, Lyu Y, Cetin B, Chan JC, Tran TN, Neve RL, Roper RJ, Brennand KJ, Roussos P, Schimenti JC, Friedman AK, Shen L, Blitzer RD, Robison AJ, Crabtree GR, and Maze I (2022) Rescue of deficits by Brwd1 copy number restoration in the Ts65Dn mouse model of Down syndrome. Nature communications 13, 6384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Wolvetang EJ, Bradfield OM, Hatzistavrou T, Crack PJ, Busciglio J, Kola I, and Hertzog PJ (2003) Overexpression of the chromosome 21 transcription factor Ets2 induces neuronal apoptosis. Neurobiology of Disease 14, 349–356 [DOI] [PubMed] [Google Scholar]
  • 117.Yu Y, Chu PY, Bowser DN, Keating DJ, Dubach D, Harper I, Tkalcevic J, Finkelstein DI, and Pritchard MA (2008) Mice deficient for the chromosome 21 ortholog Itsn1 exhibit vesicle-trafficking abnormalities. Hum Mol Genet 17, 3281–3290 [DOI] [PubMed] [Google Scholar]
  • 118.Schliebs R, and Arendt T (2011) The cholinergic system in aging and neuronal degeneration. Behav Brain Res 221, 555–563 [DOI] [PubMed] [Google Scholar]
  • 119.Perry EK, Perry RH, Smith CJ, Purohit D, Bonham J, Dick DJ, Candy JM, Edwardson JA, and Fairbairn A (1986) Cholinergic receptors in cognitive disorders. Can J Neurol Sci 13, 521–527 [DOI] [PubMed] [Google Scholar]
  • 120.Kolisnyk B, Al-Onaizi M, Soreq L, Barbash S, Bekenstein U, Haberman N, Hanin G, Kish MT, Souza da Silva J, Fahnestock M, Ule J, Soreq H, Prado VF, and Prado MAM (2017) Cholinergic Surveillance over Hippocampal RNA Metabolism and Alzheimer’s-Like Pathology. Cereb Cortex 27, 3553–3567 [DOI] [PubMed] [Google Scholar]
  • 121.Ferreira-Vieira TH, Guimaraes IM, Silva FR, and Ribeiro FM (2016) Alzheimer’s disease: Targeting the Cholinergic System. Curr Neuropharmacol 14, 101–115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kurimoto E, Yamada R, Hirakawa T, and Kimura H (2021) Therapeutic potential of TAK-071, a muscarinic M(1) receptor positive allosteric modulator with low cooperativity, for the treatment of cognitive deficits and negative symptoms associated with schizophrenia. Neurosci Lett 764, 136240. [DOI] [PubMed] [Google Scholar]
  • 123.Sabbir MG, Speth RC, and Albensi BC (2022) Loss of Cholinergic Receptor Muscarinic 1 (CHRM1) Protein in the Hippocampus and Temporal Cortex of a Subset of Individuals with Alzheimer’s Disease, Parkinson’s Disease, or Frontotemporal Dementia: Implications for Patient Survival. J Alzheimers Dis 90, 727–747 [DOI] [PubMed] [Google Scholar]
  • 124.Liu W, Li J, Yang M, Ke X, Dai Y, Lin H, Wang S, Chen L, and Tao J (2022) Chemical genetic activation of the cholinergic basal forebrain hippocampal circuit rescues memory loss in Alzheimer’s disease. Alzheimers Res Ther 14, 53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kaur G, Sharma A, Xu W, Gerum S, Alldred MJ, Subbanna S, Basavarajappa BS, Pawlik M, Ohno M, Ginsberg SD, Wilson DA, Guilfoyle DN, and Levy E (2014) Glutamatergic transmission aberration: a major cause of behavioral deficits in a murine model of Down’s syndrome. J Neurosci 34, 5099–5106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Rueda N, Llorens-Martin M, Florez J, Valdizan E, Banerjee P, Trejo JL, and Martinez-Cue C (2010) Memantine normalizes several phenotypic features in the Ts65Dn mouse model of Down syndrome. J Alzheimers Dis 21, 277–290 [DOI] [PubMed] [Google Scholar]
  • 127.Lockrow J, Boger H, Bimonte-Nelson H, and Granholm AC (2011) Effects of long-term memantine on memory and neuropathology in Ts65Dn mice, a model for Down syndrome. Behav Brain Res 221, 610–622 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Dave N, Judd JM, Decker A, Winslow W, Sarette P, Villarreal Espinosa O, Tallino S, Bartholomew SK, Bilal A, Sandler J, McDonough I, Winstone JK, Blackwood EA, Glembotski C, Karr T, and Velazquez R (2023) Dietary choline intake is necessary to prevent systems-wide organ pathology and reduce Alzheimer’s disease hallmarks. Aging Cell 22, e13775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Paoletti P, Bellone C, and Zhou Q (2013) NMDA receptor subunit diversity: impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci 14, 383–400 [DOI] [PubMed] [Google Scholar]
  • 130.Babaei P (2021) NMDA and AMPA receptors dysregulation in Alzheimer’s disease. Eur J Pharmacol 908, 174310. [DOI] [PubMed] [Google Scholar]
  • 131.Ginsberg SD, Hemby SE, Lee VM, Eberwine JH, and Trojanowski JQ (2000) Expression profile of transcripts in Alzheimer’s disease tangle-bearing CA1 neurons. Ann Neurol 48, 77–87 [PubMed] [Google Scholar]
  • 132.Conejero-Goldberg C, Hyde TM, Chen S, Dreses-Werringloer U, Herman MM, Kleinman JE, Davies P, and Goldberg TE (2011) Molecular signatures in post-mortem brain tissue of younger individuals at high risk for Alzheimer’s disease as based on APOE genotype. Mol Psychiatry 16, 836–847 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Moretto E, Murru L, Martano G, Sassone J, and Passafaro M (2018) Glutamatergic synapses in neurodevelopmental disorders. Progress in Neuro-Psychopharmacology and Biological Psychiatry 84, 328–342 [DOI] [PubMed] [Google Scholar]
  • 134.Schwenk J, Baehrens D, Haupt A, Bildl W, Boudkkazi S, Roeper J, Fakler B, and Schulte U (2014) Regional diversity and developmental dynamics of the AMPA-receptor proteome in the mammalian brain. Neuron 84, 41–54 [DOI] [PubMed] [Google Scholar]
  • 135.Oka A, and Takashima S (1999) The up-regulation of metabotropic glutamate receptor 5 (mGluR5) in Down’s syndrome brains. Acta Neuropathol 97, 275–278 [DOI] [PubMed] [Google Scholar]
  • 136.Dölen G, Osterweil E, Rao BS, Smith GB, Auerbach BD, Chattarji S, and Bear MF (2007) Correction of fragile X syndrome in mice. Neuron 56, 955–962 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Rudolph U, and Mohler H (2014) GABAA receptor subtypes: therapeutic potential in Down syndrome, affective disorders, schizophrenia, and autism. Annu Rev Pharmacol Toxicol 54, 483–507 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Milenkovic I, Stojanovic T, Aronica E, Fülöp L, Bozsó Z, Máté Z, Yanagawa Y, Adle-Biassette H, Lubec G, Szabó G, Harkany T, Kovács GG, and Keimpema E (2018) GABA(A) receptor subunit deregulation in the hippocampus of human foetuses with Down syndrome. Brain Struct Funct 223, 1501–1518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Rueda N, Florez J, and Martinez-Cue C (2012) Mouse models of Down syndrome as a tool to unravel the causes of mental disabilities. Neural Plast 2012, 584071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Rueda N, Flórez J, Dierssen M, and Martínez-Cué C (2020) Translational validity and implications of pharmacotherapies in preclinical models of Down syndrome. Prog Brain Res 251, 245–268 [DOI] [PubMed] [Google Scholar]
  • 141.Miller LG, Greenblatt DJ, Roy RB, Lopez F, and Wecker L (1989) Dietary choline intake modulates benzodiazepine receptor binding and gamma-aminobutyric acidA receptor function in mouse brain. J Pharmacol Exp Ther 248, 1–6 [PubMed] [Google Scholar]
  • 142.Schweizer C, Balsiger S, Bluethmann H, Mansuy IM, Fritschy JM, Mohler H, and Lüscher B (2003) The gamma 2 subunit of GABA(A) receptors is required for maintenance of receptors at mature synapses. Mol Cell Neurosci 24, 442–450 [DOI] [PubMed] [Google Scholar]
  • 143.Lorenz-Guertin JM, Bambino MJ, and Jacob TC (2018) γ2 GABA(A)R Trafficking and the Consequences of Human Genetic Variation. Front Cell Neurosci 12, 265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Tyagarajan SK, and Fritschy JM (2010) GABA(A) receptors, gephyrin and homeostatic synaptic plasticity. J Physiol 588, 101–106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Brickley SG, and Mody I (2012) Extrasynaptic GABA(A) receptors: their function in the CNS and implications for disease. Neuron 73, 23–34 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Das D, Phillips C, Hsieh W, Sumanth K, Dang V, and Salehi A (2014) Neurotransmitter-based strategies for the treatment of cognitive dysfunction in Down syndrome. Prog Neuropsychopharmacol Biol Psychiatry 54, 140–148 [DOI] [PubMed] [Google Scholar]
  • 147.Belichenko PV, Kleschevnikov AM, Salehi A, Epstein CJ, and Mobley WC (2007) Synaptic and cognitive abnormalities in mouse models of Down syndrome: exploring genotype-phenotype relationships. J Comp Neurol 504, 329–345 [DOI] [PubMed] [Google Scholar]
  • 148.Carbonell J, Blasco-Ibáñez JM, Crespo C, Nácher J, and Varea E (2020) Piriform cortex alterations in the Ts65Dn model for Down syndrome. Brain Res 1747, 147031. [DOI] [PubMed] [Google Scholar]
  • 149.Zeisel SH (2010) Choline: clinical nutrigenetic/nutrigenomic approaches for identification of functions and dietary requirements. J Nutrigenet Nutrigenomics 3, 209–219 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Cirillo E, Parnell LD, and Evelo CT (2017) A Review of Pathway-Based Analysis Tools That Visualize Genetic Variants. Front Genet 8, 174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Choong XY, Tosh JL, Pulford LJ, and Fisher EM (2015) Dissecting Alzheimer disease in Down syndrome using mouse models. Front Behav Neurosci 9, 268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Gupta M, Dhanasekaran AR, and Gardiner KJ (2016) Mouse models of Down syndrome: gene content and consequences. Mamm Genome 27, 538–555 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Gardiner K, Fortna A, Bechtel L, and Davisson MT (2003) 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 318, 137–147 [DOI] [PubMed] [Google Scholar]
  • 154.Sturgeon X, and Gardiner KJ (2011) Transcript catalogs of human chromosome 21 and orthologous chimpanzee and mouse regions. Mamm Genome 22, 261–271 [DOI] [PubMed] [Google Scholar]
  • 155.Freund TF, and Antal M (1988) GABA-containing neurons in the septum control inhibitory interneurons in the hippocampus. Nature 336, 170–173 [DOI] [PubMed] [Google Scholar]
  • 156.Gulyás AI, Hájos N, Katona I, and Freund TF (2003) Interneurons are the local targets of hippocampal inhibitory cells which project to the medial septum. Eur J Neurosci 17, 1861–1872 [DOI] [PubMed] [Google Scholar]

Associated Data

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

Supplementary Materials

Fig S1
Fig S2
Fig S3
Tab S1
Tab S2
Tab S3
Tab S4
Tab S5
Tab S6
Tab S7
Tab S8
Tab S9
Tab S10
Tab S11
Tab S12

Data Availability Statement

Included in article: The data that support the findings of this study are available in the Methods and/or Supplementary Material of this article. Any study data are available on request from the corresponding author. RNA-seq data analyzed within this study are available from the corresponding author upon request.

RESOURCES