Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Alzheimers Res Ther ; 14(1): 88, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35768831

ABSTRACT

BACKGROUND: The apolipoprotein E (APOE) ε4 allele confers the strongest risk for late-onset Alzheimer's disease (AD) besides age itself, but the mechanisms underlying this risk are debated. One hypothesis supported by evidence from multiple labs is that apoE4 binds to the amyloid-ß (Aß) peptide and catalyzes its polymerization into neurotoxic oligomers and fibrils. Inhibiting this early step in the amyloid cascade may thereby reduce or prevent neurodegeneration and AD. METHODS: Using a design of experiments (DOE) approach, we developed a high-throughput assay to identify inhibitors of apoE4-catalyzed polymerization of Aß into oligomers and fibrils. We used it to screen the NIH Clinical Collection of small molecule drugs tested previously in human clinical trials. We then evaluated the efficacy and cytotoxicity of the hit compounds in primary neuron models of apoE4-induced Aß and phosphorylated tau aggregation. Finally, we performed retrospective analyses of the National Alzheimer's Coordinating Center (NACC) clinical dataset, using Cox regression and Cox proportional hazards models to determine if the use of two FDA-approved hit compounds was associated with better cognitive scores (Mini-Mental State Exam), or improved AD clinical diagnosis, when compared with other medications of the same clinical indication. RESULTS: Our high-throughput screen identified eight blood-brain barrier (BBB)-permeable hit compounds that reduced apoE4-catalyzed Aß oligomer and fibril formation in a dose-dependent manner. Five hit compounds were non-toxic toward cultured neurons and also reduced apoE4-promoted Aß and tau neuropathology in a dose-dependent manner. Three of the five compounds were determined to be specific inhibitors of apoE4, whereas the other two compounds were Aß or tau aggregation inhibitors. When prescribed to AD patients for their normal clinical indications, two of the apoE4 inhibitors, imipramine and olanzapine, but not other (non-hit) antipsychotic or antidepressant medications, were associated with improvements in cognition and clinical diagnosis, especially among APOE4 carriers. CONCLUSIONS: The critical test of any proposed AD mechanism is whether it leads to effective treatments. Our high-throughput screen identified two promising FDA-approved drugs, imipramine and olanzapine, which have no structural, functional, or clinical similarities other than their shared ability to inhibit apoE4-catalyzed Aß polymerization, thus identifying this mechanism as an essential contribution of apoE4 to AD.


Subject(s)
Alzheimer Disease , Apolipoprotein E4 , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E4/metabolism , Catalysis , Cognition , Humans , Imipramine/therapeutic use , Olanzapine/therapeutic use , Polymerization , Retrospective Studies
2.
Appl Radiat Isot ; 181: 110115, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35063868

ABSTRACT

The paper describes the preparation and characterization methodology of thermal neutron sensitive inorganic compound loaded polystyrene based thin-film Plastic Scintillators (PS). Inorganic compounds with thermal neutron sensitive elements such as B2O3, LiF, Gd2O2S:Tb, Gd2O3, Gd(BO2)3:Tb is loaded in plastic scintillators, at predetermined weight percentage in a solvent-based fabrication method. The PS base solution is prepared by dissolving required quantities of polystyrene, 2,5-Diphenyloxazole (PPO), 1,4-bis(5-Phenyloxazol-2-yl) Benzene (POPOP) in the p-xylene solvent. The loading material is mixed with the scintillator solution and coated over cellulose acetate-based transparency sheet. Thin sheets of 200 × 150 mm size and 250 ± 50 µm thickness PS are prepared through multiple coating. Photoluminescence (PL), Radioluminescence (RL) and pulse height spectra (PHS) of α, ß, γ and neutron sources of the composite scintillators are studied. The selection of loading compounds is decided on their physical properties such as refractive index, the atomic percentage of neutron sensitive elements in the inorganic compound, band gap, and cost and availability of material. Gadolinium based compounds as a thermal neutron absorber showed higher detection capability for low-energy neutrons. Among the gadolinium loaded PS, Gd(BO2)3:Tb (14.3 wt%) composite scintillator showed the highest RL. A comprehensive analysis of PL, RL, PHS of α, pure ß and γ sources, un-moderated and moderated Am-Be neutron source is discussed in the paper.

3.
Sleep ; 44(10)2021 10 11.
Article in English | MEDLINE | ID: mdl-33999194

ABSTRACT

STUDY OBJECTIVES: Slow wave and spindle coupling supports memory consolidation, and loss of coupling is linked with cognitive decline and neurodegeneration. Coupling is proposed to be a possible biomarker of neurological disease, yet little is known about the different subtypes of coupling that normally occur throughout human development and aging. Here we identify distinct subtypes of spindles within slow wave upstates and describe their relationships with sleep stage across the human lifespan. METHODS: Coupling within a cross-sectional cohort of 582 subjects was quantified from stages N2 and N3 sleep across ages 6-88 years old. Results were analyzed across the study population via mixed model regression. Within a subset of subjects, we further utilized coupling to identify discrete subtypes of slow waves by their coupled spindles. RESULTS: Two different subtypes of spindles were identified during the upstates of (distinct) slow waves: an "early-fast" spindle, more common in stage N2 sleep, and a "late-fast" spindle, more common in stage N3. We further found stages N2 and N3 sleep contain a mixture of discrete subtypes of slow waves, each identified by their unique coupled-spindle timing and frequency. The relative contribution of coupling subtypes shifts across the human lifespan, and a deeper sleep phenotype prevails with increasing age. CONCLUSIONS: Distinct subtypes of slow waves and coupled spindles form the composite of slow wave sleep. Our findings support a model of sleep-dependent synaptic regulation via discrete slow wave/spindle coupling subtypes and advance a conceptual framework for the development of coupling-based biomarkers in age-associated neurological disease.


Subject(s)
Memory Consolidation , Sleep, Slow-Wave , Adolescent , Adult , Aged , Aged, 80 and over , Aging , Child , Cross-Sectional Studies , Electroencephalography , Humans , Longevity , Middle Aged , Sleep , Young Adult
5.
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
6.
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
7.
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.

8.
BMC Bioinformatics ; 16: 62, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25887958

ABSTRACT

BACKGROUND: Mass spectrometric analysis of microbial metabolism provides a long list of possible compounds. Restricting the identification of the possible compounds to those produced by the specific organism would benefit the identification process. Currently, identification of mass spectrometry (MS) data is commonly done using empirically derived compound databases. Unfortunately, most databases contain relatively few compounds, leaving long lists of unidentified molecules. Incorporating genome-encoded metabolism enables MS output identification that may not be included in databases. Using an organism's genome as a database restricts metabolite identification to only those compounds that the organism can produce. RESULTS: To address the challenge of metabolomic analysis from MS data, a web-based application to directly search genome-constructed metabolic databases was developed. The user query returns a genome-restricted list of possible compound identifications along with the putative metabolic pathways based on the name, formula, SMILES structure, and the compound mass as defined by the user. Multiple queries can be done simultaneously by submitting a text file created by the user or obtained from the MS analysis software. The user can also provide parameters specific to the experiment's MS analysis conditions, such as mass deviation, adducts, and detection mode during the query so as to provide additional levels of evidence to produce the tentative identification. The query results are provided as an HTML page and downloadable text file of possible compounds that are restricted to a specific genome. Hyperlinks provided in the HTML file connect the user to the curated metabolic databases housed in ProCyc, a Pathway Tools platform, as well as the KEGG Pathway database for visualization and metabolic pathway analysis. CONCLUSIONS: Metabolome Searcher, a web-based tool, facilitates putative compound identification of MS output based on genome-restricted metabolic capability. This enables researchers to rapidly extend the possible identifications of large data sets for metabolites that are not in compound databases. Putative compound names with their associated metabolic pathways from metabolomics data sets are returned to the user for additional biological interpretation and visualization. This novel approach enables compound identification by restricting the possible masses to those encoded in the genome.


Subject(s)
Bacteria/metabolism , Databases, Factual , Genome, Bacterial , Metabolic Networks and Pathways , Metabolome , Metabolomics/methods , Software , Bacteria/classification , Mass Spectrometry/methods
9.
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
10.
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
11.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...