Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Lipid Res ; 65(2): 100497, 2024 02.
Article in English | MEDLINE | ID: mdl-38216056

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of disease burden in the world and is highly correlated with chronic elevations of LDL-C. LDL-C-lowering drugs, such as statins or monoclonal antibodies against proprotein convertase subtilisin/kexin type 9 (PCSK9), are known to reduce the risk of cardiovascular diseases; however, statins are associated with limited efficacy and poor adherence to treatment, whereas PCSK9 inhibitors are only prescribed to a "high-risk" patient population or those who have failed other therapies. Based on the proven efficacy and safety profile of existing monoclonal antibodies, we have developed a peptide-based vaccine against PCSK9, VXX-401, as an alternative option to treat hypercholesterolemia and prevent ASCVD. VXX-401 is designed to trigger a safe humoral immune response against PCSK9, resulting in the production of endogenous antibodies and a subsequent 30-40% reduction in blood LDL-C. In this article, VXX-401 demonstrates robust immunogenicity and sustained serum LDL-C-lowering effects in nonhuman primates. In addition, antibodies induced by VXX-401 bind to human PCSK9 with high affinity and block the inhibitory effect of PCSK9 on LDL-C uptake in a hepatic cell model. A repeat-dose toxicity study conducted in nonhuman primates under good laboratory practices toxicity indicated a suitable safety and tolerability profile, with injection site reactions being the main findings. As a promising safe and effective LDL-C-lowering therapy, VXX-401 may represent a broadly accessible and convenient option to treat hypercholesterolemia and prevent ASCVD.


Subject(s)
Anticholesteremic Agents , Atherosclerosis , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hypercholesterolemia , Animals , Humans , Proprotein Convertase 9 , Hypercholesterolemia/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Cholesterol, LDL , Macaca fascicularis , Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Atherosclerosis/metabolism
2.
Neurobiol Dis ; 111: 26-35, 2018 03.
Article in English | MEDLINE | ID: mdl-29246723

ABSTRACT

Missense mutations in the multi-domain kinase LRRK2 cause late onset familial Parkinson's disease. They most commonly with classic proteinopathy in the form of Lewy bodies and Lewy neurites comprised of insoluble α-synuclein, but in rare cases can also manifest tauopathy. The normal function of LRRK2 has remained elusive, as have the cellular consequences of its mutation. Data from LRRK2 null model organisms and LRRK2-inhibitor treated animals support a physiological role for LRRK2 in regulating lysosome function. Since idiopathic and LRRK2-linked PD are associated with the intraneuronal accumulation of protein aggregates, a series of critical questions emerge. First, how do pathogenic mutations that increase LRRK2 kinase activity affect lysosome biology in neurons? Second, are mutation-induced changes in lysosome function sufficient to alter the metabolism of α-synuclein? Lastly, are changes caused by pathogenic mutation sensitive to reversal with LRRK2 kinase inhibitors? Here, we report that mutation of LRRK2 induces modest but significant changes in lysosomal morphology and acidification, and decreased basal autophagic flux when compared to WT neurons. These changes were associated with an accumulation of detergent-insoluble α-synuclein and increased neuronal release of α-synuclein and were reversed by pharmacologic inhibition of LRRK2 kinase activity. These data demonstrate a critical and disease-relevant influence of native neuronal LRRK2 kinase activity on lysosome function and α-synuclein homeostasis. Furthermore, they also suggest that lysosome dysfunction, altered neuronal α-synuclein metabolism, and the insidious accumulation of aggregated protein over decades may contribute to pathogenesis in this late-onset form of familial PD.


Subject(s)
Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Lysosomes/metabolism , Mutation , Neurons/metabolism , alpha-Synuclein/metabolism , Animals , Autophagy , Cells, Cultured , Humans , Hydrogen-Ion Concentration , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Lysosomes/pathology , Mice, Transgenic , Neurons/pathology , Parkinson Disease/genetics , Parkinson Disease/metabolism
3.
Nat Med ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902546

ABSTRACT

Investigational therapeutics that target toxic species of α-synuclein (αSyn) aim to slow down or halt disease progression in patients with Parkinson's disease (PD). Here this 44-week, randomized, placebo-controlled, double-blind, single-center phase 1 study investigated safety, tolerability and immunogenicity of UB-312, an active immunotherapeutic targeting pathological αSyn, in patients with PD. The primary outcome measures were adverse event frequency and change in anti-αSyn antibody titers in blood and cerebrospinal fluid (CSF). Exploratory outcomes were changes in clinical scales and biomarker-based target engagement as measured by seed amplification assays. Twenty patients were randomized 7:3 (UB-312:placebo) into 300/100/100 µg or 300/300/300 µg (weeks 1, 5 and 13) intramuscular prime-boost dose groups. Safety was similar across groups; adverse events were mostly mild and transient. Two patients experienced three serious adverse events in total, one possibly treatment related; all resolved without sequalae. Anti-αSyn antibodies in serum from 12/13 and CSF from 5/13 patients who received three UB-312 doses confirmed immunogenicity. Mean serum titers (in log-dilution factor) increased from baseline by 1.398 and 1.354, and peaked at week 29 at 2.520 and 2.133, for 300/100/100 µg and 300/300/300 µg, respectively. CSF titers were 0 at baseline and were 0.182 and 0.032 at week 21, respectively. Exploratory analyses showed no statistical differences in clinical scales but a significant reduction of αSyn seeds in CSF of a subset of UB-312-treated patients. These data support further UB-312 development. ClinicalTrials.gov: NCT04075318 .

4.
Nat Protoc ; 18(7): 1981-2013, 2023 07.
Article in English | MEDLINE | ID: mdl-37344608

ABSTRACT

In image-based profiling, software extracts thousands of morphological features of cells from multi-channel fluorescence microscopy images, yielding single-cell profiles that can be used for basic research and drug discovery. Powerful applications have been proven, including clustering chemical and genetic perturbations on the basis of their similar morphological impact, identifying disease phenotypes by observing differences in profiles between healthy and diseased cells and predicting assay outcomes by using machine learning, among many others. Here, we provide an updated protocol for the most popular assay for image-based profiling, Cell Painting. Introduced in 2013, it uses six stains imaged in five channels and labels eight diverse components of the cell: DNA, cytoplasmic RNA, nucleoli, actin, Golgi apparatus, plasma membrane, endoplasmic reticulum and mitochondria. The original protocol was updated in 2016 on the basis of several years' experience running it at two sites, after optimizing it by visual stain quality. Here, we describe the work of the Joint Undertaking for Morphological Profiling Cell Painting Consortium, to improve upon the assay via quantitative optimization by measuring the assay's ability to detect morphological phenotypes and group similar perturbations together. The assay gives very robust outputs despite various changes to the protocol, and two vendors' dyes work equivalently well. We present Cell Painting version 3, in which some steps are simplified and several stain concentrations can be reduced, saving costs. Cell culture and image acquisition take 1-2 weeks for typically sized batches of ≤20 plates; feature extraction and data analysis take an additional 1-2 weeks.This protocol is an update to Nat. Protoc. 11, 1757-1774 (2016): https://doi.org/10.1038/nprot.2016.105.


Subject(s)
Cell Culture Techniques , Image Processing, Computer-Assisted , Image Processing, Computer-Assisted/methods , Microscopy, Fluorescence , Mitochondria , Software
5.
J Biol Chem ; 286(40): 34914-22, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21835916

ABSTRACT

A pathological hallmark of Alzheimer disease (AD) is deposition of amyloid ß (Aß) in the brain. Aß binds to microglia via a receptor complex that includes CD36 leading to production of proinflammatory cytokines and neurotoxic reactive oxygen species and subsequent neurodegeneration. Interruption of Aß binding to CD36 is a potential therapeutic strategy for AD. To identify pharmacologic inhibitors of Aß binding to CD36, we developed a 384-well plate assay for binding of fluorescently labeled Aß to Chinese hamster ovary cells stably expressing human CD36 (CHO-CD36) and screened an Food and Drug Administration-approved compound library. The assay was optimized based on the cells' tolerance to dimethyl sulfoxide, Aß concentration, time required for Aß binding, reproducibility, and signal-to-background ratio. Using this assay, we identified four compounds as potential inhibitors of Aß binding to CD36. These compounds were ursolic acid, ellipticine, zoxazolamine, and homomoschatoline. Of these compounds, only ursolic acid, a naturally occurring pentacyclic triterpenoid, successfully inhibited binding of Aß to CHO-CD36 cells in a dose-dependent manner. The ursolic acid effect reached a plateau at ~20 µm, with a maximal inhibition of 64%. Ursolic acid also blocked binding of Aß to microglial cells and subsequent ROS production. Our data indicate that cell-based high-content screening of small molecule libraries for their ability to block binding of Aß to its receptors is a useful tool to identify novel inhibitors of receptors involved in AD pathogenesis. Our data also suggest that ursolic acid is a potential therapeutic agent for AD via its ability to block Aß-CD36 interactions.


Subject(s)
Amyloid beta-Peptides/metabolism , CD36 Antigens/biosynthesis , Triterpenes/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Drug Evaluation, Preclinical/methods , Humans , Mice , Microglia/metabolism , Neurodegenerative Diseases/metabolism , Neurotoxins , Plasmids/metabolism , Receptors, Scavenger/metabolism , Ursolic Acid
6.
Front Cell Neurosci ; 16: 894601, 2022.
Article in English | MEDLINE | ID: mdl-35677758

ABSTRACT

Microglia, the resident immune cells of the central nervous system (CNS), are responsible for maintaining homeostasis in the brain by clearing debris and are suggested to be inefficient in Alzheimer's Disease (AD), a progressive neurodegenerative disorder for which there is no disease-modifying drug. Besides pathological approaches, unbiased evidence from genome-wide association studies (GWAS) and gene network analysis implicate genes expressed in microglia that reduce phagocytic ability as susceptibility genes for AD. Thus, a central feature toward AD therapy is to increase the microglial phagocytic activities while maintaining synaptic integrity. Here, we developed a robust unbiased high content screening assay to identify potential therapeutics which can reduce the amyloid-beta (Aß1-42) load by increasing microglial uptake ability. Our screen identified the small-molecule GW5074, an inhibitor of c-RAF, a serine/threonine kinase, which significantly increased the Aß1-42 clearance activities in human monocyte-derived microglia-like (MDMi) cells, a microglia culture model that recapitulates many genetic and phenotypic aspects of human microglia. Notably, GW5074 was previously reported to be neuroprotective for cerebellar granule cells and cortical neurons. We found that GW5074 significantly increased the expression of key AD-associated microglial molecules known to modulate phagocytosis: TYROBP, SIRPß1, and TREM2. Our results demonstrated that GW5074 is a potential therapeutic for AD, by targeting microglia.

7.
Anesthesiology ; 115(4): 754-63, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21666433

ABSTRACT

BACKGROUND: In models, isoflurane produces neural and behavioral deficits in vitro and in vivo. This study tested the hypothesis that neural stem cells are adversely affected by isoflurane such that it inhibits proliferation and kills these cells. METHODS: Sprague-Dawley rat embryonic neural stem cells were plated onto 96-well plates and treated with isoflurane, 0.7, 1.4, or 2.8%, in 21% oxygen for 6 h and fixed either at the end of treatment or 6 or 24 h later. Control plates received 21% oxygen under identical conditions. Cell proliferation was assessed immunocytochemically using 5-ethynyl-2'-deoxyuridine incorporation and death by propidium iodide staining, lactate dehydrogenase release, and nuclear expression of cleaved caspase 3. Data were analyzed at each concentration using an ANOVA; P < 0.05 was considered significant. RESULTS: Isoflurane did not kill neural stem cells by any measure at any time. Isoflurane, 1.4 and 2.8%, reduced cell proliferation based upon 5-ethynyl-2'-deoxyuridine incorporation, whereas isoflurane, 0.7%, had no effect. At 24 h after treatment, the net effect was a 20-30% decrease in the number of cells in culture. CONCLUSIONS: Isoflurane does not kill neural stem cells in vitro. At concentrations at and above the minimum alveolar concentrations required for general anesthesia (1.4 and 2.8%), isoflurane inhibits proliferation of these cells but has no such effect at a subminimum alveolar concentration (0.7%). These data imply that dosages of isoflurane at and above minimum alveolar concentrations may reduce the pool of neural stem cells in vivo but that lower dosages may be devoid of such effects.


Subject(s)
Anesthetics, Inhalation/pharmacology , Isoflurane/pharmacology , Neural Stem Cells/drug effects , Animals , Carcinogens/pharmacology , Cell Death/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Coloring Agents , Culture Media , Deoxyuridine/analogs & derivatives , Female , Image Processing, Computer-Assisted , Immunohistochemistry , Intermediate Filament Proteins/metabolism , L-Lactate Dehydrogenase/metabolism , Nerve Tissue Proteins/metabolism , Nestin , Pregnancy , Propidium , Rats , Rats, Sprague-Dawley , SOXB1 Transcription Factors/metabolism , Staurosporine/pharmacology
8.
Nat Rev Drug Discov ; 20(2): 145-159, 2021 02.
Article in English | MEDLINE | ID: mdl-33353986

ABSTRACT

Image-based profiling is a maturing strategy by which the rich information present in biological images is reduced to a multidimensional profile, a collection of extracted image-based features. These profiles can be mined for relevant patterns, revealing unexpected biological activity that is useful for many steps in the drug discovery process. Such applications include identifying disease-associated screenable phenotypes, understanding disease mechanisms and predicting a drug's activity, toxicity or mechanism of action. Several of these applications have been recently validated and have moved into production mode within academia and the pharmaceutical industry. Some of these have yielded disappointing results in practice but are now of renewed interest due to improved machine-learning strategies that better leverage image-based information. Although challenges remain, novel computational technologies such as deep learning and single-cell methods that better capture the biological information in images hold promise for accelerating drug discovery.


Subject(s)
Drug Discovery/methods , Drug Industry/methods , Image Processing, Computer-Assisted/methods , Machine Learning , Animals , Computational Biology/methods , Computational Biology/trends , Drug Discovery/trends , Drug Industry/trends , High-Throughput Screening Assays/methods , High-Throughput Screening Assays/trends , Humans , Image Processing, Computer-Assisted/trends , Machine Learning/trends
9.
Methods Mol Biol ; 438: 31-8, 2008.
Article in English | MEDLINE | ID: mdl-18369747

ABSTRACT

Recent work shows that major developmental and clinical processes such as central nervous system regeneration and carcinogenesis involve stem cells (SCs) in the brain. In spite of this importance, the requirements of these SCs and their differentiated offspring (neurons, astrocytes, and oligodendrocytes) for survival and proper function are little understood. In vivo, the SCs themselves interact with their environment. This "SC niche" may be complex because it likely includes cells of the vascular and immune systems. The ability to maintain (1) and differentiate (1 -4) central nervous system (CNS) SCs in tissue culture where they can be pharmacologically or genetically (5) manipulated provides a powerful starting point for understanding their behavior. We present detailed information on the methods that permit CNS SCs to differentiate into functional neurons in tissue culture. Important aspects of the culture systems include (1) homogeneity, so that the input and output of a manipulation is known to involve the SC itself; (2) growth in monolayer to visualize and study individual SCs and their offspring; and (3) the use of fully defined culture components to exclude unknown factors from the culture. These conditions support the differentiation of functional, electrically active neurons. These methods allow cell growth and differentiation from normal adult and diseased tissue derived from both animal models and clinical samples. Ultimate validation of such a system comes from accurate prediction of in vivo effects, and the methods we present for CNS SC culture have also successfully predicted regenerative responses in the injured adult nervous system.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Neurons/cytology , Stem Cells/cytology , Animals , Cell Survival , Cryopreservation , Dissection , Immunohistochemistry , Mice , Rats
10.
PLoS One ; 13(1): e0191160, 2018.
Article in English | MEDLINE | ID: mdl-29346405

ABSTRACT

Synthetic oxytocin (sOT) is widely used during labor, yet little is known about its effects on fetal brain development despite evidence that it reaches the fetal circulation. Here, we tested the hypothesis that sOT would affect early neurodevelopment by investigating its effects on neural progenitor cells (NPC) from embryonic day 14 rat pups. NPCs expressed the oxytocin receptor (OXTR), which was downregulated by 45% upon prolonged treatment with sOT. Next, we examined the effects of sOT on NPC death, apoptosis, proliferation, and differentiation using antibodies to NeuN (neurons), Olig2 (oligodendrocytes), and GFAP (astrocytes). Treated NPCs were analysed with unbiased high-throughput immunocytochemistry. Neither 6 nor 24 h exposure to 100 pM or 100 nM sOT had an effect on viability as assessed by PI or CC-3 immunocytochemistry. Similarly, sOT had negligible effect on NPC proliferation, except that the overall rate of NPC proliferation was higher in the 24 h compared to the 6 h group regardless of sOT exposure. The most significant finding was that sOT exposure caused NPCs to select a predominantly neuronal lineage, along with a concomitant decrease in glial cells. Collectively, our data suggest that perinatal exposure to sOT can have neurodevelopmental consequences for the fetus, and support the need for in vivo anatomical and behavioral studies in offspring exposed to sOT in utero.


Subject(s)
Neural Stem Cells/drug effects , Oxytocin/toxicity , Animals , Astrocytes/cytology , Astrocytes/drug effects , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Female , Humans , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neuroglia/cytology , Neuroglia/drug effects , Neurons/cytology , Neurons/drug effects , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oxytocin/administration & dosage , Oxytocin/metabolism , Placenta/metabolism , Pregnancy , Prenatal Exposure Delayed Effects , Rats , Rats, Sprague-Dawley , Receptors, Oxytocin/metabolism
11.
Brain Res ; 1175: 107-16, 2007 Oct 17.
Article in English | MEDLINE | ID: mdl-17884023

ABSTRACT

Parkinson's disease (PD) is a neurodegenerative disease whose hallmark pathological features include a selective loss of dopaminergic neurons in the midbrain. Recent studies have described the activation of a stress-induced signal cascade, c-Jun N-terminal kinase (JNK)-mediated activation of c-Jun, and an increase in the expression of a downstream effector, cyclooxygenase 2 (COX-2), in postmortem PD brains. The neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), which induces selective neuronal loss in the midbrain similar to that seen in PD, also induces JNK-mediated activation of c-Jun and generates a COX-2 response in C57BL/6J mice. However, mice exhibit a strain-dependent susceptibility to MPTP. Identifying the point(s) of molecular divergence in the MPTP-induced response may provide insight into the cause of PD or a means to identify susceptibility to PD in humans. Here we examined JNK signaling and COX-2 induction in two strains of mice, the MPTP-sensitive C57BL/6J and the MPTP-resistant Swiss Webster (SW). We show that C57BL/6J and SW strains differ in JNK and c-Jun activation in response to MPTP. In addition, the MPTP-induced COX-2 response occurs exclusively in C57BL/6J mice. Furthermore, strain-specific responses to MPTP are not due to differences in MPP(+) levels and are not secondary to cell death. These results provide evidence toward a mechanism of strain-dependent sensitivity to MPTP.


Subject(s)
Cyclooxygenase 2/metabolism , Drug Resistance/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Nerve Degeneration/enzymology , Parkinsonian Disorders/enzymology , Substantia Nigra/enzymology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/enzymology , Neurons/pathology , Neurotoxins/pharmacology , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Species Specificity , Substantia Nigra/drug effects , Substantia Nigra/physiopathology
12.
Sci Transl Med ; 9(421)2017 Dec 20.
Article in English | MEDLINE | ID: mdl-29263232

ABSTRACT

Microglia are emerging as a key cell type in neurodegenerative diseases, yet human microglia are challenging to study in vitro. We developed an in vitro cell model system composed of human monocyte-derived microglia-like (MDMi) cells that recapitulated key aspects of microglia phenotype and function. We then used this model system to perform an expression quantitative trait locus (eQTL) study examining 94 genes from loci associated with Alzheimer's disease, Parkinson's disease, and multiple sclerosis. We found six loci (CD33, PILRB, NUP160, LRRK2, RGS1, and METTL21B) in which the risk haplotype drives the association with both disease susceptibility and altered expression of a nearby gene (cis-eQTL). In the PILRB and LRRK2 loci, the cis-eQTL was found in the MDMi cells but not in human peripheral blood monocytes, suggesting that differentiation of monocytes into microglia-like cells led to the acquisition of a cellular state that could reveal the functional consequences of certain genetic variants. We further validated the effect of risk haplotypes at the protein level for PILRB and CD33, and we confirmed that the CD33 risk haplotype altered phagocytosis by the MDMi cells. We propose that increased LRRK2 gene expression by MDMi cells could be a functional outcome of rs76904798, a single-nucleotide polymorphism in the LRKK2 locus that is associated with Parkinson's disease.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Microglia/pathology , Models, Biological , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Cell Polarity , Gene Expression Regulation , Genome-Wide Association Study , Genotype , Humans , Monocytes/pathology , Phenotype , Polymorphism, Single Nucleotide/genetics , Protein Isoforms/metabolism , Quantitative Trait Loci/genetics , Sialic Acid Binding Ig-like Lectin 3/metabolism
14.
PLoS One ; 11(7): e0158058, 2016.
Article in English | MEDLINE | ID: mdl-27379684

ABSTRACT

Neurocognitive dysfunction is common in survivors of intensive care. Prolonged sedation has been implicated but the mechanisms are unclear. Neurogenesis continues into adulthood and is implicated in learning. The neural progenitor cells (NPC) that drive neurogenesis have receptors for the major classes of sedatives used clinically, suggesting that interruption of neurogenesis may partly contribute to cognitive decline in ICU survivors. Using an in vitro system, we tested the hypothesis that prolonged exposure to propofol concentration- and duration-dependently kills or markedly decreases the proliferation of NPCs. NPCs isolated from embryonic day 14 Sprague-Dawley rat pups were exposed to 0, 2.5, or 5.0 µg/mL of propofol, concentrations consistent with deep clinical anesthesia, for either 4 or 24 hours. Cells were assayed for cell death and proliferation either immediately following propofol exposure or 24 hours later. NPC death and apoptosis were measured by propidium iodine staining and cleaved caspase-3 immunocytochemistry, respectively, while proliferation was measured by EdU incorporation. Staurosporine (1µM for 6h) was used as a positive control for cell death. Cells were analyzed with unbiased high-throughput immunocytochemistry. There was no cell death at either concentration of propofol or duration of exposure. Neither concentration of propofol impaired NPC proliferation when exposure lasted 4 h, but when exposure lasted 24 h, propofol had an anti-proliferative effect at both concentrations (P < 0.0001, propofol vs. control). However, this effect was transient; proliferation returned to baseline 24 h after discontinuation of propofol (P = 0.37, propofol vs. control). The transient but reversible suppression of NPC proliferation, absence of cytotoxicity, and negligible effect on the neural stem cell pool pool suggest that propofol, even in concentrations used for clinical anesthesia, has limited impact on neural progenitor cell biology.


Subject(s)
Cell Proliferation/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Propofol/pharmacology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Hypnotics and Sedatives/pharmacology , Immunohistochemistry , Nestin/metabolism , Neural Stem Cells/metabolism , Rats, Sprague-Dawley , Staurosporine/pharmacology , Time Factors
15.
Cell Rep ; 12(9): 1414-29, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26299970

ABSTRACT

Genetic and clinical association studies have identified disrupted in schizophrenia 1 (DISC1) as a candidate risk gene for major mental illness. DISC1 is interrupted by a balanced chr(1;11) translocation in a Scottish family in which the translocation predisposes to psychiatric disorders. We investigate the consequences of DISC1 interruption in human neural cells using TALENs or CRISPR-Cas9 to target the DISC1 locus. We show that disruption of DISC1 near the site of the translocation results in decreased DISC1 protein levels because of nonsense-mediated decay of long splice variants. This results in an increased level of canonical Wnt signaling in neural progenitor cells and altered expression of fate markers such as Foxg1 and Tbr2. These gene expression changes are rescued by antagonizing Wnt signaling in a critical developmental window, supporting the hypothesis that DISC1-dependent suppression of basal Wnt signaling influences the distribution of cell types generated during cortical development.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Neural Stem Cells/metabolism , Neurogenesis , Wnt Signaling Pathway , Cells, Cultured , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Genome, Human , Humans , Induced Pluripotent Stem Cells/cytology , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Nonsense Mediated mRNA Decay , Protein Isoforms/genetics , Protein Isoforms/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Translocation, Genetic
17.
J Biomol Screen ; 19(1): 44-56, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24019256

ABSTRACT

TDP-43 is an RNA binding protein found to accumulate in the cytoplasm of brain and spinal cord from patients affected with amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). Nuclear TDP-43 protein regulates transcription through several mechanisms, and under stressed conditions, it forms cytoplasmic aggregates that co-localize with stress granule (SG) proteins in cell culture. These granules are also found in the brain and spinal cord of patients affected with ALS and FTLD. The mechanism through which TDP-43 might contribute to neurodegenerative diseases is poorly understood. To investigate the pathophysiology of TDP-43 aggregation and to isolate potential therapeutic targets, we screened a chemical library of 75,000 compounds using high-content analysis with PC12 cells that inducibly express human TDP-43 tagged with green fluorescent protein (GFP). The screen identified 16 compounds that dose-dependently decreased the TDP-43 inclusions without significant cellular toxicity or changes in total TDP-43 expression levels. To validate the effect, we tested compounds by Western blot analysis and in a Caenorhabditis elegans model that replicates some of the relevant disease phenotypes. The hits from this assay will be useful for elucidating regulation of TDP-43, stress granule response, and possible ALS therapeutics.


Subject(s)
DNA-Binding Proteins/metabolism , Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays , Stress, Physiological/drug effects , Animals , Animals, Genetically Modified , Arsenites/pharmacology , Caenorhabditis elegans , Cell Line , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Drug Discovery/methods , Gene Expression , Genes, Reporter , Humans , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neuroprotective Agents/pharmacology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Small Molecule Libraries , Sodium Compounds/pharmacology
18.
mBio ; 3(1)2012.
Article in English | MEDLINE | ID: mdl-22334517

ABSTRACT

UNLABELLED: Many Gram-negative bacteria utilize specialized secretion systems to inject proteins (effectors) directly into host cells. Little is known regarding how bacteria ensure that only small subsets of the thousands of proteins they encode are recognized as substrates of the secretion systems, limiting their identification through bioinformatic analyses. Many of these proteins require chaperones to direct their secretion. Here, using the newly described protein interaction platform assay, we demonstrate that type 3 secretion system class IB chaperones from one bacterium directly bind their own effectors as well as those from other species. In addition, we observe that expression of class IB homologs from seven species, including pathogens and endosymbionts, mediate the translocation of effectors from Shigella directly into host cells, demonstrating that class IB chaperones are often functionally interchangeable. Notably, class IB chaperones bind numerous effectors. However, as previously proposed, they are not promiscuous; rather they recognize a defined sequence that we designate the conserved chaperone-binding domain (CCBD) sequence [(LMIF)(1)XXX(IV)(5)XX(IV)(8)X(N)(10)]. This sequence is the first defined amino acid sequence to be identified for any interspecies bacterial secretion system, i.e., a system that delivers proteins directly into eukaryotic cells. This sequence provides a new means to identify substrates of type III secretion systems. Indeed, using a pattern search algorithm for the CCBD sequence, we have identified the first two probable effectors from an endosymbiont, Sodalis glossinidius. IMPORTANCE: Many Gram-negative pathogens utilize type 3 secretion systems to deliver tens of effectors into host cells. In order to understand the diverse ways that these organisms cause disease, it is necessary to identify their effectors, many of which require chaperones to be secreted. Here we establish that class IB chaperones are not promiscuous, as previously proposed, but rather recognize a conserved effector sequence. We demonstrate that pattern search algorithms based on this defined sequence can be used to identify previously unknown effectors. Furthermore, we observe that class IB chaperones from at least seven bacterial species are functionally interchangeable. Not only do they bind and mediate the delivery of their own set of effectors into host cells but they also bind to type 3 substrates from other bacteria, suggesting that inhibitors that block chaperone-effector interactions could provide a novel means to effectively treat infections due to Gram-negative pathogens, including organisms resistant to currently available antibiotics.


Subject(s)
Bacterial Secretion Systems , Enterobacteriaceae/metabolism , Molecular Chaperones/metabolism , Protein Interaction Mapping/methods , Shigella/physiology , Algorithms , Amino Acid Sequence , Bacterial Proteins/metabolism , Computational Biology , Conserved Sequence , Enterobacteriaceae/physiology , HeLa Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Salmonella typhimurium/metabolism , Salmonella typhimurium/physiology , Shigella/metabolism , Species Specificity , Substrate Specificity , Symbiosis
19.
PLoS One ; 5(2): e9414, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20195471

ABSTRACT

BACKGROUND: The ability to grow a uniform cell type from the adult central nervous system (CNS) is valuable for developing cell therapies and new strategies for drug discovery. The adult mammalian brain is a source of neural stem cells (NSC) found in both neurogenic and non-neurogenic zones but difficulties in culturing these hinders their use as research tools. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that NSCs can be efficiently grown in adherent cell cultures when angiogenic signals are included in the medium. These signals include both anti-angiogenic factors (the soluble form of the Notch receptor ligand, Dll4) and pro-angiogenic factors (the Tie-2 receptor ligand, Angiopoietin 2). These treatments support the self renewal state of cultured NSCs and expression of the transcription factor Hes3, which also identifies the cancer stem cell population in human tumors. In an organotypic slice model, angiogenic factors maintain vascular structure and increase the density of dopamine neuron processes. CONCLUSIONS/SIGNIFICANCE: We demonstrate new properties of adult NSCs and a method to generate efficient adult NSC cultures from various central nervous system areas. These findings will help establish cellular models relevant to cancer and regeneration.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Cell Proliferation/drug effects , Neurons/cytology , Stem Cells/cytology , Adult , Adult Stem Cells/cytology , Angiopoietin-2/pharmacology , Animals , Brain/cytology , Cell Culture Techniques , Cell Differentiation/drug effects , Cells, Cultured , DNA-Binding Proteins/metabolism , Fibroblast Growth Factors/pharmacology , Haplorhini , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Membrane Proteins/pharmacology , Mice , Rats , Repressor Proteins , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL