Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 17029, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34426604

ABSTRACT

Mutations in MAPT (microtubule-associated protein tau) cause frontotemporal dementia (FTD). MAPT mutations are associated with abnormal tau phosphorylation levels and accumulation of misfolded tau protein that can propagate between neurons ultimately leading to cell death (tauopathy). Recently, a p.A152T tau variant was identified as a risk factor for FTD, Alzheimer's disease, and synucleinopathies. Here we used induced pluripotent stem cells (iPSC) from a patient carrying this p.A152T variant to create a robust, functional cellular assay system for probing pathophysiological tau accumulation and phosphorylation. Using stably transduced iPSC-derived neural progenitor cells engineered to enable inducible expression of the pro-neural transcription factor Neurogenin 2 (Ngn2), we generated disease-relevant, cortical-like glutamatergic neurons in a scalable, high-throughput screening compatible format. Utilizing automated confocal microscopy, and an advanced image-processing pipeline optimized for analysis of morphologically complex human neuronal cultures, we report quantitative, subcellular localization-specific effects of multiple kinase inhibitors on tau, including ones under clinical investigation not previously reported to affect tau phosphorylation. These results demonstrate the potential for using patient iPSC-derived ex vivo models of tauopathy as genetically accurate, disease-relevant systems to probe tau biochemistry and support the discovery of novel therapeutics for tauopathies.


Subject(s)
Glutamates/metabolism , Image Processing, Computer-Assisted , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Neurons/pathology , Proteomics , Tauopathies/pathology , tau Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Line , Humans , Induced Pluripotent Stem Cells/drug effects , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Phosphorylation/drug effects , Protein Kinases/metabolism , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology
2.
J Med Chem ; 62(21): 9600-9617, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31535859

ABSTRACT

Using structure-guided design, several cell based assays, and microdosed positron emission tomography (PET) imaging, we identified a series of highly potent, selective, and brain-penetrant oxazole-4-carboxamide-based inhibitors of glycogen synthase kinase-3 (GSK-3). An isotopologue of our first-generation lead, [3H]PF-367, demonstrates selective and specific target engagement in vitro, irrespective of the activation state. We discovered substantial ubiquitous GSK-3-specific radioligand binding in Tg2576 Alzheimer's disease (AD), suggesting application for these compounds in AD diagnosis and identified [11C]OCM-44 as our lead GSK-3 radiotracer, with optimized brain uptake by PET imaging in nonhuman primates. GSK-3ß-isozyme selectivity was assessed to reveal OCM-51, the most potent (IC50 = 0.030 nM) and selective (>10-fold GSK-3ß/GSK-3α) GSK-3ß inhibitor known to date. Inhibition of CRMP2T514 and tau phosphorylation, as well as favorable therapeutic window against WNT/ß-catenin signaling activation, was observed in cells.


Subject(s)
Brain/metabolism , Drug Discovery , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Positron-Emission Tomography/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Animals , Blood-Brain Barrier/metabolism , Brain/diagnostic imaging , Catalytic Domain , Glycogen Synthase Kinase 3 beta/chemistry , HEK293 Cells , Humans , Mice , Models, Molecular , Neuroimaging , Oxazoles/chemistry , Oxazoles/metabolism , Oxazoles/pharmacology , Protein Kinase Inhibitors/metabolism , Triazoles/chemistry , Triazoles/metabolism , Triazoles/pharmacology
3.
ACS Chem Neurosci ; 10(8): 3769-3777, 2019 08 21.
Article in English | MEDLINE | ID: mdl-31330099

ABSTRACT

Histone deacetylases (HDACs) are enzymes involved in the epigenetic control of gene expression. A handful of HDAC inhibitors have been approved for the treatment of cancer, and HDAC inhibition has also been proposed as a novel therapeutic strategy for neurodegenerative disorders. These disorders include progranulin (PGRN)-deficient forms of frontotemporal dementia caused by mutations in the GRN gene that lead to haploinsufficiency. Hydroxamic-acid-based inhibitors of HDACs 1-3, reported to have fast-on/fast-off binding kinetics, induce increased expression of PGRN in human neuronal models, while the benzamide class of slow-binding HDAC inhibitors does not produce this effect. These observations indicate that the kinetics of HDAC inhibitor binding can be tuned for optimal induction of human PGRN expression in neurons. Here, we further expand on these findings using human cortical-like, glutamatergic neurons. We provide evidence that two prototypical, potent hydroxamic acid HDAC inhibitors that induce PGRN (panobinostat and trichostatin A) exhibit an initial fast-binding step followed by a second, slower step, referred to as mechanism B of slow binding, rather than simpler fast-on/fast-off binding kinetics. In addition, we show that trapoxin A, a macrocyclic, epoxyketone-containing class I HDAC inhibitor, exhibits slow binding with high, picomolar potency and also induces PGRN expression in human neurons. Finally, we demonstrate induction of PGRN expression by fast-on/fast-off, highly potent, macrocyclic HDAC inhibitors with ethyl ketone or ethyl ester Zn2+ binding groups. Taken together, these data expand our understanding of HDAC1-3 inhibitor binding kinetics, and further delineate the specific combinations of structural and kinetic features of HDAC inhibitors that are optimal for upregulating PGRN expression in human neurons and thus may have translational relevance in neurodegenerative disease.


Subject(s)
Histone Deacetylase Inhibitors/pharmacokinetics , Hydroxamic Acids/pharmacokinetics , Neurons/drug effects , Panobinostat/pharmacokinetics , Progranulins/metabolism , Frontotemporal Dementia/metabolism , Gene Expression/drug effects , Glutamic Acid/metabolism , Histone Deacetylases/metabolism , Humans , Neural Stem Cells , Neurons/metabolism , Peptides/pharmacokinetics
4.
Nat Med ; 25(1): 152-164, 2019 01.
Article in English | MEDLINE | ID: mdl-30510257

ABSTRACT

Identifying the mechanisms through which genetic risk causes dementia is an imperative for new therapeutic development. Here, we apply a multistage, systems biology approach to elucidate the disease mechanisms in frontotemporal dementia. We identify two gene coexpression modules that are preserved in mice harboring mutations in MAPT, GRN and other dementia mutations on diverse genetic backgrounds. We bridge the species divide via integration with proteomic and transcriptomic data from the human brain to identify evolutionarily conserved, disease-relevant networks. We find that overexpression of miR-203, a hub of a putative regulatory microRNA (miRNA) module, recapitulates mRNA coexpression patterns associated with disease state and induces neuronal cell death, establishing this miRNA as a regulator of neurodegeneration. Using a database of drug-mediated gene expression changes, we identify small molecules that can normalize the disease-associated modules and validate this experimentally. Our results highlight the utility of an integrative, cross-species network approach to drug discovery.


Subject(s)
Dementia/genetics , Evolution, Molecular , Gene Regulatory Networks , Neurodegenerative Diseases/genetics , Animals , Cell Death/genetics , Disease Models, Animal , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Gene Expression Regulation , Genetic Predisposition to Disease , Genetic Vectors/metabolism , Humans , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , MicroRNAs/metabolism , Proteomics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Transcriptome/genetics , tau Proteins/metabolism
5.
J Biol Chem ; 292(47): 19209-19225, 2017 11 24.
Article in English | MEDLINE | ID: mdl-28972160

ABSTRACT

The ubiquitin-proteasome system (UPS) is responsible for most selective protein degradation in eukaryotes and regulates numerous cellular processes, including cell cycle control and protein quality control. A component of this system, the deubiquitinating enzyme USP14, associates with the proteasome where it can rescue substrates from degradation by removal of the ubiquitin tag. We previously found that a small-molecule inhibitor of USP14, known as IU1, can increase the rate of degradation of a subset of proteasome substrates. We report here the synthesis and characterization of 87 variants of IU1, which resulted in the identification of a 10-fold more potent USP14 inhibitor that retains specificity for USP14. The capacity of this compound, IU1-47, to enhance protein degradation in cells was tested using as a reporter the microtubule-associated protein tau, which has been implicated in many neurodegenerative diseases. Using primary neuronal cultures, IU1-47 was found to accelerate the rate of degradation of wild-type tau, the pathological tau mutants P301L and P301S, and the A152T tau variant. We also report that a specific residue in tau, lysine 174, is critical for the IU1-47-mediated tau degradation by the proteasome. Finally, we show that IU1-47 stimulates autophagic flux in primary neurons. In summary, these findings provide a powerful research tool for investigating the complex biology of USP14.


Subject(s)
Embryo, Mammalian/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Neurons/metabolism , Pyrroles/pharmacology , Ubiquitin Thiolesterase/physiology , tau Proteins/metabolism , Animals , Cells, Cultured , Cytoplasm/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Enzyme Inhibitors/chemical synthesis , Fibroblasts/cytology , Fibroblasts/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Pyrroles/chemical synthesis , Rats, Sprague-Dawley , Ubiquitin/metabolism , Ubiquitination
6.
Curr Protoc Hum Genet ; 92: 21.8.1-21.8.21, 2017 01 11.
Article in English | MEDLINE | ID: mdl-28075486

ABSTRACT

Reprogramming of human somatic cells into induced pluripotent stem (iPS) cells has greatly expanded the set of research tools available to investigate the molecular and cellular mechanisms underlying central nervous system (CNS) disorders. Realizing the promise of iPS cell technology for the identification of novel therapeutic targets and for high-throughput drug screening requires implementation of methods for the large-scale production of defined CNS cell types. Here we describe a protocol for generating stable, highly expandable, iPS cell-derived CNS neural progenitor cells (NPC) using multi-dimensional fluorescence activated cell sorting (FACS) to purify NPC defined by cell surface markers. In addition, we describe a rapid, efficient, and reproducible method for generating excitatory cortical-like neurons from these NPC through inducible expression of the pro-neural transcription factor Neurogenin 2 (iNgn2-NPC). Finally, we describe methodology for the use of iNgn2-NPC for probing human neuroplasticity and mechanisms underlying CNS disorders using high-content, single-cell-level automated microscopy assays. © 2017 by John Wiley & Sons, Inc.


Subject(s)
Cell Differentiation , Central Nervous System Diseases/drug therapy , Central Nervous System Diseases/pathology , High-Throughput Screening Assays , Induced Pluripotent Stem Cells/cytology , Models, Biological , Neural Stem Cells/cytology , Neurons/cytology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Cell Proliferation , Humans , Induced Pluripotent Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neuronal Plasticity , Neurons/metabolism , Single-Cell Analysis
7.
Stem Cell Reports ; 7(3): 325-340, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27594585

ABSTRACT

Frontotemporal dementia (FTD) and other tauopathies characterized by focal brain neurodegeneration and pathological accumulation of proteins are commonly associated with tau mutations. However, the mechanism of neuronal loss is not fully understood. To identify molecular events associated with tauopathy, we studied induced pluripotent stem cell (iPSC)-derived neurons from individuals carrying the tau-A152T variant. We highlight the potential of in-depth phenotyping of human neuronal cell models for pre-clinical studies and identification of modulators of endogenous tau toxicity. Through a panel of biochemical and cellular assays, A152T neurons showed accumulation, redistribution, and decreased solubility of tau. Upregulation of tau was coupled to enhanced stress-inducible markers and cell vulnerability to proteotoxic, excitotoxic, and mitochondrial stressors, which was rescued upon CRISPR/Cas9-mediated targeting of tau or by pharmacological activation of autophagy. Our findings unmask tau-mediated perturbations of specific pathways associated with neuronal vulnerability, revealing potential early disease biomarkers and therapeutic targets for FTD and other tauopathies.


Subject(s)
Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Induced Pluripotent Stem Cells/metabolism , Mutation , Neurons/metabolism , tau Proteins/genetics , Amino Acid Substitution , Autophagy/genetics , Biomarkers , Cell Differentiation , Cell Line , Codon , Frontotemporal Dementia/pathology , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Protein Isoforms , Protein Processing, Post-Translational , Stress, Physiological , tau Proteins/metabolism
8.
J Biomol Screen ; 17(9): 1252-63, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22923789

ABSTRACT

Wnt/ß-catenin signaling has emerged as a central player in pathways implicated in the pathophysiology and treatment of neuropsychiatric disorders. To identify potential novel therapeutics for these disorders, high-throughput screening (HTS) assays reporting on Wnt/ß-catenin signaling in disease-relevant contexts are needed. The use of human patient-derived induced pluripotent stem cell (iPSC) models provides ideal disease-relevant context if these stem cell cultures can be adapted for HTS-compatible formats. Here, we describe a sensitive, HTS-compatible Wnt/ß-catenin signaling reporter system generated in homogeneous, expandable neural progenitor cells (NPCs) derived from human iPSCs. We validated this system by demonstrating dose-responsive stimulation by several known Wnt/ß-catenin signaling pathway modulators, including Wnt3a, a glycogen synthase kinase-3 (GSK3) inhibitor, and the bipolar disorder therapeutic lithium. These responses were robust and reproducible over time across many repeated assays. We then conducted a screen of ~1500 compounds from a library of Food and Drug Administration-approved drugs and known bioactives and confirmed the HTS hits, revealing multiple chemical and biological classes of novel small-molecule probes of Wnt/ß-catenin signaling. Generating these type of pathway-selective, cell-based phenotypic assays in human iPSC-derived neural cells will advance the field of human experimental neurobiology toward the goal of identifying and validating targets for neuropsychiatric disorders.


Subject(s)
High-Throughput Screening Assays/methods , Neural Stem Cells/drug effects , Wnt Signaling Pathway/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Lithium/pharmacology , Mental Disorders/drug therapy , Mental Disorders/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Reproducibility of Results , Small Molecule Libraries/pharmacology , Wnt3A Protein/metabolism , beta Catenin/metabolism
9.
Diabetes Res Clin Pract ; 90(3): e85-7, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20970867

ABSTRACT

The objective of this study was to elucidate the role of fat distribution in predicting insulin resistance in peri- or post-menopausal women. The results demonstrated that insulin resistance increases with waist circumference and subscapular skinfold thickness but decreases with higher thigh circumflex in the peri- and post-menopausal women.


Subject(s)
Body Constitution , Insulin Resistance , Postmenopause , Adipose Tissue/anatomy & histology , Aged , Arm/anatomy & histology , Female , Humans , Middle Aged , Skinfold Thickness , Thigh/anatomy & histology , Waist Circumference
10.
J Neurosci ; 22(24): 10643-52, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12486157

ABSTRACT

The glutamate transporter excitatory amino acid transporter 3 (EAAT3) is polarized to the apical surface in epithelial cells and localized to the dendritic compartment in hippocampal neurons, where it is clustered adjacent to postsynaptic sites. In this study, we analyzed the sequences in EAAT3 that are responsible for its polarized localization in Madin-Darby canine kidney (MDCK) cells and neurons. Confocal microscopy and cell surface biotinylation assays demonstrated that deletion of the EAAT3 C terminus or replacement of the C terminus of EAAT3 with the analogous region in EAAT1 eliminated apical localization in MDCK cells. The C terminus of EAAT3 was sufficient to redirect the basolateral-preferring EAAT1 and the nonpolarized EAAT2 to the apical surface. Using alanine substitution mutants, we identified a short peptide motif in the cytoplasmic C-terminal region of EAAT3 that directs its apical localization in MDCK cells. Mutation of this sequence also impairs dendritic targeting of EAAT3 in hippocampal neurons but does not interfere with the clustering of EAAT3 on dendritic spines and filopodia. These data provide the first evidence that an identical cytoplasmic motif can direct apical targeting in epithelia and somatodendritic targeting in neurons. Moreover, our results demonstrate that the two fundamental features of the localization of EAAT3 in neurons, its restriction to the somatodendritic domain and its clustering near postsynaptic sites, are mediated by distinct molecular mechanisms.


Subject(s)
Amino Acid Transport System X-AG/analysis , Amino Acid Transport System X-AG/chemistry , Hippocampus/chemistry , Neurons/chemistry , Symporters/analysis , Symporters/chemistry , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Transport System X-AG/genetics , Amino Acid Transport System X-AG/metabolism , Animals , Cell Line , Cell Polarity , Cells, Cultured , Dendrites/chemistry , Dogs , Excitatory Amino Acid Transporter 1 , Excitatory Amino Acid Transporter 2 , Excitatory Amino Acid Transporter 3 , Glutamate Plasma Membrane Transport Proteins , Hippocampus/cytology , Hippocampus/metabolism , Molecular Sequence Data , Mutation , Neurons/metabolism , Protein Sorting Signals , Protein Transport , Rats , Symporters/genetics , Symporters/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...