Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Neurobiol Dis ; 168: 105694, 2022 06 15.
Article in English | MEDLINE | ID: mdl-35307513

ABSTRACT

Down syndrome (DS) is characterized by chronic neuroinflammation, peripheral inflammation, astrogliosis, imbalanced excitatory/inhibitory neuronal function, and cognitive deficits in both humans and mouse models. Suppression of inflammation has been proposed as a therapeutic approach to treating DS co-morbidities, including intellectual disability (DS/ID). Conversely, we discovered previously that treatment with the innate immune system stimulating cytokine granulocyte-macrophage colony-stimulating factor (GM-CSF), which has both pro- and anti-inflammatory activities, improved cognition and reduced brain pathology in a mouse model of Alzheimer's disease (AD), another inflammatory disorder, and improved cognition and reduced biomarkers of brain pathology in a phase II trial of humans with mild-to-moderate AD. To investigate the effects of GM-CSF treatment on DS/ID in the absence of AD, we assessed behavior and brain pathology in 12-14 month-old DS mice (Dp[16]1Yey) and their wild-type (WT) littermates, neither of which develop amyloid, and found that subcutaneous GM-CSF treatment (5 µg/day, five days/week, for five weeks) improved performance in the radial arm water maze in both Dp16 and WT mice compared to placebo. Dp16 mice also showed abnormal astrocyte morphology, increased percent area of GFAP staining in the hippocampus, clustering of astrocytes in the hippocampus, and reduced numbers of calretinin-positive interneurons in the entorhinal cortex and subiculum, and all of these brain pathologies were improved by GM-CSF treatment. These findings suggest that stimulating and/or modulating inflammation and the innate immune system with GM-CSF treatment may enhance cognition in both people with DS/ID and in the typical aging population.


Subject(s)
Alzheimer Disease , Down Syndrome , Aged , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Astrocytes/metabolism , Cognition , Cytokines/metabolism , Disease Models, Animal , Down Syndrome/drug therapy , Down Syndrome/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hippocampus/metabolism , Humans , Immune System/metabolism , Immune System/pathology , Inflammation/drug therapy , Inflammation/pathology , Interneurons/metabolism , Mice
2.
Genes (Basel) ; 12(11)2021 10 26.
Article in English | MEDLINE | ID: mdl-34828303

ABSTRACT

Down syndrome (DS), trisomy of human chromosome 21 (Hsa21), is the most common genetic cause of intellectual disability. The Dp10(1)Yey (Dp10) is a mouse model of DS that is trisomic for orthologs of 25% of the Hsa21 protein-coding genes, the entirety of the Hsa21 syntenic region on mouse chromosome 10. Trisomic genes include several involved in brain development and function, two that modify and regulate the activities of sex hormones, and two that produce sex-specific phenotypes as null mutants. These last four are the only Hsa21 genes with known sexually dimorphic properties. Relatively little is known about the potential contributions to the DS phenotype of segmental trisomy of Mmu10 orthologs. Here, we have tested separate cohorts of female and male Dp10 mice, at 3 and 9 months of age, in an open field elevated zero maze, rotarod, and balance beam, plus the learning and memory tasks, spontaneous alternation, puzzle box, double-H maze, context fear conditioning, and acoustic startle/prepulse inhibition, that depend upon the function of the prefrontal cortex, striatum, hippocampus, and cerebellum. We show that there are age and sex-specific differences in strengths and weaknesses, suggesting that genes within the telomere proximal region of Hsa21 influence the DS phenotype.


Subject(s)
Chromosomes, Mammalian/genetics , Down Syndrome , Learning/physiology , Mice/genetics , Age Factors , Animals , Behavior, Animal , Chromosome Mapping , Disease Models, Animal , Down Syndrome/genetics , Down Syndrome/pathology , Female , Humans , Male , Maze Learning/physiology , Mice, Inbred C57BL , Phenotype , Sex Characteristics
3.
Genes (Basel) ; 12(10)2021 09 28.
Article in English | MEDLINE | ID: mdl-34680922

ABSTRACT

Down syndrome (DS), trisomy of the long arm of human chromosome 21 (Hsa21), is the most common genetic cause of intellectual disability (ID). Currently, there are no effective pharmacotherapies. The success of clinical trials to improve cognition depends in part on the design of preclinical evaluations in mouse models. To broaden understanding of the common limitations of experiments in learning and memory, we report performance in context fear conditioning (CFC) in three mouse models of DS, the Dp(16)1Yey, Dp(17)1Yey and Dp(10)1Yey (abbreviated Dp16, Dp17 and Dp10), separately trisomic for the human Hsa21 orthologs mapping to mouse chromosomes 16, 17 and 10, respectively. We examined female and male mice of the three lines on the standard C57BL/6J background at 3 months of age and Dp17 and Dp10 at 18 months of age. We also examined female and male mice of Dp17 and Dp10 at 3 months of age as F1 hybrids obtained from a cross with the DBA/2J background. Results indicate that genotype, sex, age and genetic background affect CFC performance. These data support the need to use both female and male mice, trisomy of sets of all Hsa21 orthologs, and additional ages and genetic backgrounds to improve the reliability of preclinical evaluations of drugs for ID in DS.


Subject(s)
Conditioning, Classical , Down Syndrome/physiopathology , Genetic Background , Animals , Down Syndrome/genetics , Fear , Female , Hippocampus/growth & development , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Sex Characteristics
4.
Front Neurol ; 12: 654606, 2021.
Article in English | MEDLINE | ID: mdl-34122302

ABSTRACT

Epilepsy is characterized by recurrent, spontaneous seizures and is a major contributor to the global burden of neurological disease. Although epilepsy can result from a variety of brain insults, in many cases the cause is unknown and, in a significant proportion of cases, seizures cannot be controlled by available treatments. Understanding the molecular alterations that underlie or are triggered by epileptogenesis would help to identify therapeutics to prevent or control progression to epilepsy. To this end, the moderate throughput technique of Reverse Phase Protein Arrays (RPPA) was used to profile changes in protein expression in a pilocarpine mouse model of acquired epilepsy. Levels of 54 proteins, comprising phosphorylation-dependent and phosphorylation-independent components of major signaling pathways and cellular complexes, were measured in hippocampus, cortex and cerebellum of mice at six time points, spanning 15 min to 2 weeks after induction of status epilepticus. Results illustrate the time dependence of levels of the commonly studied MTOR pathway component, pS6, and show, for the first time, detailed responses during epileptogenesis of multiple components of the MTOR, MAPK, JAK/STAT and apoptosis pathways, NMDA receptors, and additional cellular complexes. Also noted are time- and brain region- specific changes in correlations among levels of functionally related proteins affecting both neurons and glia. While hippocampus and cortex are primary areas studied in pilocarpine-induced epilepsy, cerebellum also shows significant time-dependent molecular responses.

5.
Trends Genet ; 37(5): 444-459, 2021 05.
Article in English | MEDLINE | ID: mdl-33097276

ABSTRACT

Human chromosome 21 (Hsa21) contains more than 500 genes, making trisomy 21 one of the most complex genetic perturbations compatible with life. The ultimate goal of Down syndrome (DS) research is to design therapies that improve quality of life for individuals with DS by understanding which subsets of Hsa21 genes contribute to DS-associated phenotypes throughout the lifetime. However, the complexity of DS pathogenesis has made developing appropriate animal models an ongoing challenge. Here, we examine lessons learned from a variety of model systems, including yeast, nematode, fruit fly, and zebrafish, and discuss emerging methods for creating murine models that better reflect the genetic basis of trisomy 21.


Subject(s)
Chromosomes, Human, Pair 21/genetics , Down Syndrome/genetics , Genomics/methods , Aneuploidy , Animals , Disease Models, Animal , Drosophila/genetics , Humans , Mice , Pan troglodytes/genetics , Rats , Yeasts/genetics , Zebrafish/genetics
6.
Aging Cell ; 17(5): e12812, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30028071

ABSTRACT

Mounting evidence implicates chronic oxidative stress as a critical driver of the aging process. Down syndrome (DS) is characterized by a complex phenotype, including early senescence. DS cells display increased levels of reactive oxygen species (ROS) and mitochondrial structural and metabolic dysfunction, which are counterbalanced by sustained Nrf2-mediated transcription of cellular antioxidant response elements (ARE). Here, we show that caspase 3/PKCδdependent activation of the Nrf2 pathway in DS and Dp16 (a mouse model of DS) cells is necessary to protect against chronic oxidative damage and to preserve cellular functionality. Mitochondria-targeted catalase (mCAT) significantly reduced oxidative stress, restored mitochondrial structure and function, normalized replicative and wound healing capacity, and rendered the Nrf2-mediated antioxidant response dispensable. These results highlight the critical role of Nrf2/ARE in the maintenance of DS cell homeostasis and validate mitochondrial-specific interventions as a key aspect of antioxidant and antiaging therapies.


Subject(s)
Down Syndrome/metabolism , Down Syndrome/pathology , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Animals , Antioxidants/metabolism , Caspase 3/metabolism , Catalase/metabolism , Cell Proliferation , Cell Survival , Cytoprotection , Fibroblasts/metabolism , Fibroblasts/pathology , HEK293 Cells , Humans , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/pathology , Models, Biological , Protein Kinase C-delta/metabolism , Protein Stability , Signal Transduction , Wound Healing
7.
J Bioinform Comput Biol ; 16(3): 1850001, 2018 06.
Article in English | MEDLINE | ID: mdl-29478376

ABSTRACT

Reverse Phase Protein Arrays (RPPA) is a high-throughput technology used to profile levels of protein expression. Handling the large datasets generated by RPPA can be facilitated by appropriate software tools. Here, we describe RPPAware, a free and intuitive software suite that was developed specifically for analysis and visualization of RPPA data. RPPAware is a portable tool that requires no installation and was built using Java. Many modules of the tool invoke R to utilize the statistical features. To demonstrate the utility of RPPAware, data generated from screening brain regions of a mouse model of Down syndrome with 62 antibodies were used as a case study. The ease of use and efficiency of RPPAware can accelerate data analysis to facilitate biological discovery. RPPAware 1.0 is freely available under GNU General Public License from the project website at http://downsyndrome.ucdenver.edu/iddrc/rppaware/home.htm along with a full documentation of the tool.


Subject(s)
Brain/metabolism , Down Syndrome/metabolism , Protein Array Analysis/methods , Software , Animals , Antibodies/analysis , Brain-Derived Neurotrophic Factor/metabolism , Chromosomes, Human, Pair 21 , Disease Models, Animal , Humans , Mice , Protein Array Analysis/statistics & numerical data , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , User-Computer Interface , bcl-Associated Death Protein/metabolism , Dyrk Kinases
8.
Neurobiol Aging ; 57: 120-132, 2017 09.
Article in English | MEDLINE | ID: mdl-28641136

ABSTRACT

The Ts65Dn is a popular mouse model of Down syndrome (DS). It displays DS-relevant features of learning/memory deficits and age-related loss of functional markers in basal forebrain cholinergic neurons. Here we describe protein expression abnormalities in brain regions of 12-month-old male Ts65Dn mice. We show that the magnitudes of abnormalities of human chromosome 21 and non-human chromosome 21 orthologous proteins are greater at 12 months than at ∼6 months. Age-related exacerbations involve the number of components affected in the mechanistic target of rapamycin pathway, the levels of components of the mitogen-activated protein kinase pathway, and proteins associated with Alzheimer's disease. Among brain regions, the number of abnormalities in cerebellum decreased while the number in cortex greatly increased with age. The Ts65Dn is being used in preclinical evaluations of drugs for cognition in DS. Most commonly, drug evaluations are tested in ∼4- to 6-month-old mice. Data on age-related changes in magnitude and specificity of protein perturbations can be used to understand the molecular basis of changes in cognitive ability and to predict potential age-related specificities in drug efficacies.


Subject(s)
Aging/genetics , Cerebellum/metabolism , Down Syndrome/genetics , Gene Expression , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Aging/pathology , Aging/psychology , Animals , Basal Forebrain/pathology , Cholinergic Neurons/pathology , Chromosomes, Human, Pair 21/genetics , Disease Models, Animal , Down Syndrome/pathology , Down Syndrome/psychology , Female , Humans , Learning , Male , Memory , Mice, Inbred C3H , Mice, Inbred C57BL
9.
Prenat Diagn ; 37(1): 31-36, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27859447

ABSTRACT

Individuals with Down syndrome (DS) are diagnosed with autism spectrum disorder (ASD) at a significantly higher frequency than the typical population. The differentiation of ASD symptoms from those of severe intellectual disability presents diagnostic challenges, which have led to more refined methods in the clinical evaluation of ASD in DS. These improved phenotypic characterization methods not only provide better diagnosis of ASD in DS, but may also be useful in elucidating the etiology of the increased prevalence of ASD in DS. Because all individuals with the classic presentation of DS have trisomy 21, it is possible that those with co-occurring DS and ASD may have additional genetic variants which can act as modifiers of the phenotype, leading to the development of ASD. © 2016 John Wiley & Sons, Ltd.


Subject(s)
Autism Spectrum Disorder/complications , Down Syndrome/complications , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/genetics , Down Syndrome/genetics , Genetic Variation , Humans
10.
Mol Syndromol ; 7(5): 251-261, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27867340

ABSTRACT

Down syndrome (DS) is the most common genetic cause of intellectual disability (ID) in humans with an incidence of ∼1:1,000 live births worldwide. It is caused by the presence of an extra copy of all or a segment of the long arm of human chromosome 21 (trisomy 21). People with DS present with a constellation of phenotypic alterations involving most organs and organ systems. ID is present in all people with DS, albeit with variable severity. DS is also the most frequent genetic cause of Alzheimer's disease (AD), and ∼50% of those with DS will develop AD-related dementia. In the last few years, significant progress has been made in understanding the crucial genotype-phenotype relationships in DS, in identifying the alterations in molecular pathways leading to the various clinical conditions present in DS, and in preclinical evaluations of potential therapies to improve the overall health and well-being of individuals with DS. In June 2015, 230 scientists, advocates, patients, and family members met in Paris for the 1st International Conference of the Trisomy 21 Research Society. Here, we report some of the most relevant presentations that took place during the meeting.

11.
Mamm Genome ; 27(11-12): 538-555, 2016 12.
Article in English | MEDLINE | ID: mdl-27538963

ABSTRACT

Down syndrome (DS), trisomy of human chromosome 21 (Hsa21), is challenging to model in mice. Not only is it a contiguous gene syndrome spanning 35 Mb of the long arm of Hsa21, but orthologs of Hsa21 genes map to segments of three mouse chromosomes, Mmu16, Mmu17, and Mmu10. The Ts65Dn was the first viable segmental trisomy mouse model for DS; it is a partial trisomy currently popular in preclinical evaluations of drugs for cognition in DS. Limitations of the Ts65Dn are as follows: (i) it is trisomic for 125 human protein-coding orthologs, but only 90 of these are Hsa21 orthologs and (ii) it lacks trisomy for ~75 Hsa21 orthologs. In recent years, several additional mouse models of DS have been generated, each trisomic for a different subset of Hsa21 genes or their orthologs. To best exploit these models and interpret the results obtained with them, prior to proposing clinical trials, an understanding of their trisomic gene content, relative to full trisomy 21, is necessary. Here we first review the functional information on Hsa21 protein-coding genes and the more recent annotation of a large number of functional RNA genes. We then discuss the conservation and genomic distribution of Hsa21 orthologs in the mouse genome and the distribution of mouse-specific genes. Lastly, we consider the strengths and weaknesses of mouse models of DS based on the number and nature of the Hsa21 orthologs that are, and are not, trisomic in each, and discuss their validity for use in preclinical evaluations of drug responses.


Subject(s)
Disease Models, Animal , Down Syndrome/genetics , Genome/genetics , Trisomy/genetics , Animals , Chromosomes/genetics , Chromosomes, Human, Pair 21/genetics , Down Syndrome/drug therapy , Down Syndrome/pathology , Humans , Mice , Trisomy/pathology
12.
Biol Sex Differ ; 6: 24, 2015.
Article in English | MEDLINE | ID: mdl-26557979

ABSTRACT

BACKGROUND: While many sex differences in structure and function of the mammalian brain have been described, the molecular correlates of these differences are not broadly known. Also unknown is how sex differences at the protein level are perturbed by mutations that lead to intellectual disability (ID). Down syndrome (DS) is the most common genetic cause of ID and is due to trisomy of human chromosome 21 (Hsa21) and the resulting increased expression of Hsa21-encoded genes. The Dp(10)1Yey mouse model (Dp10) of DS is trisomic for orthologs of 39 Hsa21 protein-coding genes that map to mouse chromosome 10 (Mmu10), including four genes with known sex differences in functional properties. How these genes contribute to the DS cognitive phenotype is not known. METHODS: Using reverse phase protein arrays, levels of ~100 proteins/protein modifications were measured in the hippocampus, cerebellum, and cortex of female and male controls and their trisomic Dp10 littermates. Proteins were chosen for their known roles in learning/memory and synaptic plasticity and include components of the MAPK, MTOR, and apoptosis pathways, immediate early genes, and subunits of ionotropic glutamate receptors. Protein levels were compared between genotypes, sexes, and brain regions using a three-level mixed effects model and the Benjamini-Hochberg correction for multiple testing. RESULTS: In control mice, levels of approximately one half of the proteins differ significantly between females and males in at least one brain region; in the hippocampus alone, levels of 40 % of the proteins are significantly higher in females. Trisomy of the Mmu10 segment differentially affects female and male profiles, perturbing protein levels most in the cerebellum of female Dp10 and most in the hippocampus of male Dp10. Cortex is minimally affected by sex and genotype. Diverse pathways and processes are implicated in both sex and genotype differences. CONCLUSIONS: The extensive sex differences in control mice in levels of proteins involved in learning/memory illustrate the molecular complexity underlying sex differences in normal neurological processes. The sex-specific abnormalities in the Dp10 suggest the possibility of sex-specific phenotypic features in DS and reinforce the need to use female as well as male mice, in particular in preclinical evaluations of drug responses.

13.
PLoS One ; 10(6): e0129126, 2015.
Article in English | MEDLINE | ID: mdl-26111164

ABSTRACT

Down syndrome (DS) is a chromosomal abnormality (trisomy of human chromosome 21) associated with intellectual disability and affecting approximately one in 1000 live births worldwide. The overexpression of genes encoded by the extra copy of a normal chromosome in DS is believed to be sufficient to perturb normal pathways and normal responses to stimulation, causing learning and memory deficits. In this work, we have designed a strategy based on the unsupervised clustering method, Self Organizing Maps (SOM), to identify biologically important differences in protein levels in mice exposed to context fear conditioning (CFC). We analyzed expression levels of 77 proteins obtained from normal genotype control mice and from their trisomic littermates (Ts65Dn) both with and without treatment with the drug memantine. Control mice learn successfully while the trisomic mice fail, unless they are first treated with the drug, which rescues their learning ability. The SOM approach identified reduced subsets of proteins predicted to make the most critical contributions to normal learning, to failed learning and rescued learning, and provides a visual representation of the data that allows the user to extract patterns that may underlie novel biological responses to the different kinds of learning and the response to memantine. Results suggest that the application of SOM to new experimental data sets of complex protein profiles can be used to identify common critical protein responses, which in turn may aid in identifying potentially more effective drug targets.


Subject(s)
Down Syndrome/metabolism , Learning , Memantine/pharmacology , Protein Interaction Maps , Animals , Cluster Analysis , Disease Models, Animal , Down Syndrome/drug therapy , Gene Expression Regulation/drug effects , Humans , Learning/drug effects , Memantine/therapeutic use , Memory Disorders/drug therapy , Memory Disorders/metabolism , Mice , Protein Interaction Maps/drug effects
14.
PLoS One ; 10(3): e0119491, 2015.
Article in English | MEDLINE | ID: mdl-25793384

ABSTRACT

Down syndrome (DS) is caused by an extra copy of human chromosome 21 (Hsa21). Although it is the most common genetic cause of intellectual disability (ID), there are, as yet, no effective pharmacotherapies. The Ts65Dn mouse model of DS is trisomic for orthologs of ∼55% of Hsa21 classical protein coding genes. These mice display many features relevant to those seen in DS, including deficits in learning and memory (L/M) tasks requiring a functional hippocampus. Recently, the N-methyl-D-aspartate (NMDA) receptor antagonist, memantine, was shown to rescue performance of the Ts65Dn in several L/M tasks. These studies, however, have not been accompanied by molecular analyses. In previous work, we described changes in protein expression induced in hippocampus and cortex in control mice after exposure to context fear conditioning (CFC), with and without memantine treatment. Here, we extend this analysis to Ts65Dn mice, measuring levels of 85 proteins/protein modifications, including components of MAP kinase and MTOR pathways, and subunits of NMDA receptors, in cortex and hippocampus of Ts65Dn mice after failed learning in CFC and after learning was rescued by memantine. We show that, compared with wild type littermate controls, (i) of the dynamic responses seen in control mice in normal learning, >40% also occur in Ts65Dn in failed learning or are compensated by baseline abnormalities, and thus are considered necessary but not sufficient for successful learning, and (ii) treatment with memantine does not in general normalize the initial protein levels but instead induces direct and indirect responses in approximately half the proteins measured and results in normalization of the endpoint protein levels. Together, these datasets provide a first view of the complexities associated with pharmacological rescue of learning in the Ts65Dn. Extending such studies to additional drugs and mouse models of DS will aid in identifying pharmacotherapies for effective clinical trials.


Subject(s)
Down Syndrome/metabolism , Learning , Proteome , Proteomics , Animals , Disease Models, Animal , Down Syndrome/drug therapy , Down Syndrome/genetics , Learning/drug effects , Memantine/pharmacology , Mice , Protein Subunits/metabolism , Proteomics/methods , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism
15.
Rev. Síndr. Down ; 32(124): 25-34, mar. 2015. tab, ilus
Article in Spanish | IBECS | ID: ibc-134718

ABSTRACT

Es creciente, y a veces acuciante, el interés de familias y profesionales relacionados con el síndrome de Down por conocer los posible beneficios que algunos fármacos pueden reportar sobre la cognición y conducta de las personas con síndrome de Down. Los primeros ensayos clínicos que se están realizando en las personas, y los resultados de los cada vez más frecuentes estudios sobre los modelos animales, han creado una atmósfera de esperanza y, por qué no decirlo, también de ansiedad. A principios del presente mes de enero fue publicada una exhaustiva revisión escrita por la investigadora norteamericana Katheleen J. Gardiner, en la que analiza con detalle todos los resultados obtenidos por diversos fármacos en el modelo de ratón más universalmente utilizado, el ratón Ts65Dn, y explica las posibilidades de que algunos de estos fármacos puedan ser probados en la clínica humana. Ofrecemos un muy amplio resumen en español que, esperamos, dé cumplida respuesta a las actuales inquietudes. Quien desee disponer del trabajo original en inglés y la bibliografía, puede solicitarlos a la dirección de la revista


Over the last several years, very promising results have been obtained with a mouse model of Down syndrome, the Ts65Dn. A diverse array of drugs has been shown to rescue, or partially rescue relevant deficits in learning and memory and abnormalities in cellular and electrophysiological features seen in the Ts65Dn. These results suggest that some levels of amelioration or prevention of cognitive deficits in people with Down syndrome may be possible. We review information from the preclinical evaluations on the efficacy of these drugs, and advance possible applications of some of the studied drugs to clinical trials


Subject(s)
Animals , Mice , Drug Combinations , Neurogenesis/genetics , Down Syndrome/complications , Basal Ganglia/abnormalities , Basal Ganglia/metabolism , Neurogenesis/physiology , Down Syndrome/genetics , Down Syndrome/history
16.
Drug Des Devel Ther ; 9: 103-25, 2015.
Article in English | MEDLINE | ID: mdl-25552901

ABSTRACT

Down syndrome (DS), also known as trisomy 21, is the most common genetic cause of intellectual disability (ID). Although ID can be mild, the average intelligence quotient is in the range of 40-50. All individuals with DS will also develop the neuropathology of Alzheimer's disease (AD) by the age of 30-40 years, and approximately half will display an AD-like dementia by the age of 60 years. DS is caused by an extra copy of the long arm of human chromosome 21 (Hsa21) and the consequent elevated levels of expression, due to dosage, of trisomic genes. Despite a worldwide incidence of one in 700-1,000 live births, there are currently no pharmacological treatments available for ID or AD in DS. However, over the last several years, very promising results have been obtained with a mouse model of DS, the Ts65Dn. A diverse array of drugs has been shown to rescue, or partially rescue, DS-relevant deficits in learning and memory and abnormalities in cellular and electrophysiological features seen in the Ts65Dn. These results suggest that some level of amelioration or prevention of cognitive deficits in people with DS may be possible. Here, we review information from the preclinical evaluations in the Ts65Dn, how drugs were selected, how efficacy was judged, and how outcomes differ, or not, among studies. We also summarize the current state of human clinical trials for ID and AD in DS. Lastly, we describe the genetic limitations of the Ts65Dn as a model of DS, and in the preclinical testing of pharmacotherapeutics, and suggest additional targets to be considered for potential pharmacotherapies.


Subject(s)
Cognition Disorders/drug therapy , Down Syndrome/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Cognition Disorders/genetics , Down Syndrome/genetics , Humans
17.
Alzheimers Dement ; 11(6): 700-9, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25510383

ABSTRACT

In the United States, estimates indicate there are between 250,000 and 400,000 individuals with Down syndrome (DS), and nearly all will develop Alzheimer's disease (AD) pathology starting in their 30s. With the current lifespan being 55 to 60 years, approximately 70% will develop dementia, and if their life expectancy continues to increase, the number of individuals developing AD will concomitantly increase. Pathogenic and mechanistic links between DS and Alzheimer's prompted the Alzheimer's Association to partner with the Linda Crnic Institute for Down Syndrome and the Global Down Syndrome Foundation at a workshop of AD and DS experts to discuss similarities and differences, challenges, and future directions for this field. The workshop articulated a set of research priorities: (1) target identification and drug development, (2) clinical and pathological staging, (3) cognitive assessment and clinical trials, and (4) partnerships and collaborations with the ultimate goal to deliver effective disease-modifying treatments.


Subject(s)
Alzheimer Disease/physiopathology , Down Syndrome/physiopathology , Alzheimer Disease/diagnosis , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Animals , Clinical Trials as Topic , Congresses as Topic , Disease Models, Animal , Down Syndrome/diagnosis , Down Syndrome/drug therapy , Down Syndrome/pathology , Drug Discovery , Humans , Neuropsychological Tests
18.
Mol Cell Proteomics ; 13(4): 919-37, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24469516

ABSTRACT

Analysis of the molecular basis of learning and memory has revealed details of the roles played by many genes and the proteins they encode. Because most individual studies focus on a small number of proteins, many complexities of the relationships among proteins and their dynamic responses to stimulation are not known. We have used the technique of reverse phase protein arrays (RPPA) to assess the levels of more than 80 proteins/protein modifications in subcellular fractions from hippocampus and cortex of mice trained in Context Fear Conditioning (CFC). Proteins include components of signaling pathways, several encoded by immediate early genes or involved in apoptosis and inflammation, and subunits of glutamate receptors. At one hour after training, levels of more than half the proteins had changed in one or more fractions, among them multiple components of the Mitogen-activated protein kinase, MAPK, and Mechanistic Target of Rapamycin, MTOR, pathways, subunits of glutamate receptors, and the NOTCH pathway modulator, NUMB homolog (Drosophila). Levels of 37 proteins changed in the nuclear fraction of hippocampus alone. Abnormalities in levels of thirteen proteins analyzed have been reported in brains of patients with Alzheimer's Disease. We therefore further investigated the protein profiles of mice treated with memantine, a drug approved for treatment of AD. In hippocampus, memantine alone induced many changes similar to those seen after CFC and altered the levels of seven proteins associated with Alzheimer's Disease abnormalities. Lastly, to further explore the relevance of these datasets, we superimposed responses to CFC and memantine onto components of the long term potentiation pathway, a process subserving learning and memory formation. Fourteen components of the long term potentiation pathway and 26 proteins interacting with components responded to CFC and/or memantine. Together, these datasets provide a novel view of the diversity and complexity in protein responses and interactions following normal learning.


Subject(s)
Cerebral Cortex/metabolism , Conditioning, Classical/physiology , Fear/physiology , Hippocampus/metabolism , Signal Transduction , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cerebral Cortex/drug effects , Conditioning, Classical/drug effects , Cytosol/drug effects , Cytosol/metabolism , Hippocampus/drug effects , Humans , Memantine/pharmacology , Mice , Mice, Inbred C57BL , Protein Array Analysis , Proteomics , Signal Transduction/drug effects
19.
Hum Mol Genet ; 22(9): 1709-24, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23349361

ABSTRACT

Tc1 mouse model of Down syndrome (DS) is functionally trisomic for ∼120 human chromosome 21 (HSA21) classical protein-coding genes. Tc1 mice display features relevant to the DS phenotype, including abnormalities in learning and memory and synaptic plasticity. To determine the molecular basis for the phenotypic features, the levels of 90 phosphorylation-specific and phosphorylation-independent proteins were measured by Reverse Phase Protein Arrays in hippocampus and cortex, and 64 in cerebellum, of Tc1 mice and littermate controls. Abnormal levels of proteins involved in MAP kinase, mTOR, GSK3B and neuregulin signaling were identified in trisomic mice. In addition, altered correlations among the levels of N-methyl-D-aspartate (NMDA) receptor subunits and the HSA21 proteins amyloid beta (A4) precursor protein (APP) and TIAM1, and between immediate early gene (IEG) proteins and the HSA21 protein superoxide dismutase-1 (SOD1) were found in the hippocampus of Tc1 mice, suggesting altered stoichiometry among these sets of functionally interacting proteins. Protein abnormalities in Tc1 mice were compared with the results of a similar analysis of Ts65Dn mice, a DS mouse model that is trisomic for orthologs of 50 genes trisomic in the Tc1 plus an additional 38 HSA21 orthologs. While there are similarities, abnormalities unique to the Tc1 include increased levels of the S100B calcium-binding protein, mTOR proteins RAPTOR and P70S6, the AMP-kinase catalytic subunit AMPKA, the IEG proteins FBJ murine osteosarcoma viral oncogene homolog (CFOS) and activity-regulated cytoskeleton-associated protein (ARC), and the neuregulin 1 receptor ERBB4. These data identify novel perturbations, relevant to neurological function and to some seen in Alzheimer's disease, that may occur in the DS brain, potentially contributing to phenotypic features and influencing drug responses.


Subject(s)
Brain/physiopathology , Down Syndrome/genetics , Gene Expression Profiling , Neoplasm Proteins/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Down Syndrome/metabolism , Down Syndrome/physiopathology , Female , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Phosphorylation , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Regulatory-Associated Protein of mTOR , S100 Calcium Binding Protein beta Subunit , S100 Proteins/genetics , S100 Proteins/metabolism , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
20.
Gene ; 512(2): 219-25, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23103828

ABSTRACT

Down syndrome (DS) is the most common genetic aberration leading to intellectual disability. DS results from an extra copy of the long arm of human chromosome 21 (HSA21) and the increased expression of trisomic genes due to gene dosage. While expression in DS and DS models has been studied extensively at the RNA level, much less is known about expression of trisomic genes at the protein level. We have used quantitative Western blotting with antibodies to 20 proteins encoded by HSA21 to assess trisomic protein expression in lymphoblastoid cell lines (LCLs) from patients with DS and in brains from two mouse models of DS. These antibodies have recently become available and the 20 proteins largely have not been investigated previously for their potential contributions to the phenotypic features of DS. Twelve proteins had detectable expression in LCLs and three, CCT8, MX1 and PWP2, showed elevated levels in LCLs derived from patients with DS compared with controls. Antibodies against 15 proteins detected bands of appropriate sizes in lysates from mouse brain cortex. Genes for 12 of these proteins are trisomic in the Tc1 mouse model of DS, but only SIM2 and ZNF295 showed elevated expression in Tc1 cortex when compared with controls. Genes for eight of the 15 proteins are trisomic in the Ts65Dn mouse model of DS, but only ZNF294 was over expressed in cortex. Comparison of trisomic gene expression at the protein level with previous reports at the mRNA level showed many inconsistencies. These may be caused by natural inter-individual variability, differences in the age of mice analyzed, or post-transcriptional regulation of gene dosage effects. These antibodies provide resources for further investigation of the molecular basis of intellectual disability in DS.


Subject(s)
Cerebral Cortex/metabolism , Chromosomes, Human, Pair 21/metabolism , Down Syndrome/metabolism , Gene Dosage , Gene Expression Regulation , Nerve Tissue Proteins/biosynthesis , Adult , Animals , Cell Line , Cerebral Cortex/pathology , Chromosomes, Human, Pair 21/genetics , Down Syndrome/genetics , Female , Humans , Male , Mice , Nerve Tissue Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...