Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Neurobiol Dis ; 146: 105092, 2020 12.
Article in English | MEDLINE | ID: mdl-32979507

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in exon 1 of the huntingtin gene. Emerging evidence shows that additional epigenetic factors can modify disease phenotypes. Harnessing the ability of the epigenome to modify the disease for therapeutic purposes is therefore of interest. Epigenome modifiers, such as histone deacetylase inhibitors (HDACi), have improved pathology in a range of HD models. Yet in clinical trials, HDACi have failed to alleviate HD symptoms in patients. This study investigated potential reasons for the lack of translation of the therapeutic benefits of HDACi from lab to clinic. We analysed histone acetylation patterns of immuno-positive nuclei from brain sections and tissue microarrays from post-mortem human control and HD cases alongside several well-established HD models (OVT73 transgenic HD sheep, YAC128 mice, and an in vitro cell model expressing 97Q mutant huntingtin). Significant increases in histone H4 acetylation were observed in post-mortem HD cases, OVT73 transgenic HD sheep and in vitro models; these changes were absent in YAC128 mice. In addition, nuclear labelling for acetyl-histone H4 levels were inversely proportional to mutant huntingtin aggregate load in HD human cortex. Our data raise concerns regarding the utility of HDACi for the treatment of HD when regions of pathology exhibit already elevated histone acetylation patterns and emphasize the importance of searching for alternative epigenetic targets in future therapeutic strategies aiming to rescue HD phenotypes.


Subject(s)
Brain/metabolism , Histone Deacetylase Inhibitors/pharmacology , Huntington Disease/genetics , Huntington Disease/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Brain/drug effects , Brain/physiopathology , Disease Models, Animal , Histone Deacetylases/drug effects , Histone Deacetylases/metabolism , Humans , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Sheep/genetics , Sheep/physiology
2.
Adv Exp Med Biol ; 978: 321-336, 2017.
Article in English | MEDLINE | ID: mdl-28523554

ABSTRACT

Substantial progress has been made in identifying Alzheimer's disease (AD) risk-associated variants using genome-wide association studies (GWAS). The majority of these risk variants reside in noncoding regions of the genome making their functional evaluation difficult; however, they also infer the presence of unconventional regulatory regions that may reside at these locations. We know from these studies that rare familial cases of AD account for less than 5% of all AD cases and autosomal dominant mutations in APP, PSEN1 and PSEN2 account for less than 10% of the genetic basis of these familial cases [1]. The sporadic form of AD, while more complex, still has a substantial genetic component evidenced by observational studies where 30-48% of AD patients have a first degree relative who is also affected [2]. In addition, the strongest risk factor after age is the APOE E4 polymorphism, and more than 20 other risk variants have been identified to date, reviewed in two recent papers [3, 4]. Monozygotic twin studies have revealed a discordance for AD, implicating that a combination of epigenetic and genetic factors are likely involved in the development of AD [5].


Subject(s)
Alzheimer Disease/genetics , Epigenesis, Genetic/genetics , Histone Code/genetics , Nerve Tissue Proteins/metabolism , Acetylation , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Apolipoprotein E4/genetics , Brain Chemistry , Disease Models, Animal , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Methylation , Mice , Mutation , Nerve Tissue Proteins/genetics , Phosphorylation , Protein Processing, Post-Translational/genetics , Risk Factors
3.
Neurobiol Dis ; 74: 281-94, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25484284

ABSTRACT

Histone acetylation is an epigenetic modification that plays a critical role in chromatin remodelling and transcriptional regulation. There is increasing evidence that epigenetic modifications may become compromised in aging and increase susceptibility to the development of neurodegenerative disorders such as Alzheimer's disease. Immunohistochemical labelling of free-floating sections from the inferior temporal gyrus (Alzheimer's disease, n=14; control, n=17) and paraffin-embedded tissue microarrays containing tissue from the middle temporal gyrus (Alzheimer's disease, n=29; control, n=28) demonstrated that acetyl histone H3 and acetyl histone H4 levels, as well as total histone H3 and total histone H4 protein levels, were significantly increased in post-mortem Alzheimer's disease brain tissue compared to age- and sex-matched neurologically normal control brain tissue. Changes in acetyl histone levels were proportional to changes in total histone levels. The increase in acetyl histone H3 and H4 was observed in Neuronal N immunopositive pyramidal neurons in Alzheimer's disease brain. Using immunolabelling, histone markers correlated significantly with the level of glial fibrillary acidic protein and HLA-DP, -DQ and -DR immunopositive cells and with the pathological hallmarks of Alzheimer's disease (hyperphosphorylated tau load and ß-amyloid plaques). Given that histone acetylation changes were correlated with changes in total histone protein, it was important to evaluate if protein degradation pathways may be compromised in Alzheimer's disease. Consequently, significant positive correlations were also found between ubiquitin load and histone modifications. The relationship between histone acetylation and ubiquitin levels was further investigated in an in vitro model of SK-N-SH cells treated with the proteasome inhibitor Mg132 and the histone deacetylase inhibitor valproic acid. In this model, compromised protein degradation caused by Mg132 lead to elevated histone labelling. In addition, valproic acid worked synergistically with Mg132 in elevating ubiquitin load and causing cell death. These findings highlight important pathological relationships linking a compromise in protein turnover with the histone changes observed in Alzheimer's disease post-mortem human brain.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Histones/metabolism , Aged , Alzheimer Disease/pathology , Amyloid/metabolism , Benzothiazoles , Blotting, Western , Brain/pathology , Cell Line, Tumor , Female , Fluorescent Antibody Technique , Humans , Male , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Thiazoles/metabolism , Tissue Array Analysis , Ubiquitin/metabolism , tau Proteins/metabolism
4.
Commun Biol ; 5(1): 235, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35301433

ABSTRACT

Platelet-derived growth factor-BB (PDGF-BB):PDGF receptor-ß (PDGFRß) signalling in brain pericytes is critical to the development, maintenance and function of a healthy blood-brain barrier (BBB). Furthermore, BBB impairment and pericyte loss in Alzheimer's disease (AD) is well documented. We found that PDGF-BB:PDGFRß signalling components were altered in human AD brains, with a marked reduction in vascular PDGFB. We hypothesised that reduced PDGF-BB:PDGFRß signalling in pericytes may impact on the BBB. We therefore tested the effects of PDGF-BB on primary human brain pericytes in vitro to define pathways related to BBB function. Using pharmacological inhibitors, we dissected distinct aspects of the PDGF-BB response that are controlled by extracellular signal-regulated kinase (ERK) and Akt pathways. PDGF-BB promotes the proliferation of pericytes and protection from apoptosis through ERK signalling. In contrast, PDGF-BB:PDGFRß signalling through Akt augments pericyte-derived inflammatory secretions. It may therefore be possible to supplement PDGF-BB signalling to stabilise the cerebrovasculature in AD.


Subject(s)
Alzheimer Disease , Pericytes , Alzheimer Disease/metabolism , Becaplermin/metabolism , Becaplermin/pharmacology , Brain/metabolism , Humans , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/pharmacology
5.
Elife ; 102021 07 06.
Article in English | MEDLINE | ID: mdl-34227937

ABSTRACT

In mammals, females generally live longer than males. Nevertheless, the mechanisms underpinning sex-dependent longevity are currently unclear. Epigenetic clocks are powerful biological biomarkers capable of precisely estimating chronological age and identifying novel factors influencing the aging rate using only DNA methylation data. In this study, we developed the first epigenetic clock for domesticated sheep (Ovis aries), which can predict chronological age with a median absolute error of 5.1 months. We have discovered that castrated male sheep have a decelerated aging rate compared to intact males, mediated at least in part by the removal of androgens. Furthermore, we identified several androgen-sensitive CpG dinucleotides that become progressively hypomethylated with age in intact males, but remain stable in castrated males and females. Comparable sex-specific methylation differences in MKLN1 also exist in bat skin and a range of mouse tissues that have high androgen receptor expression, indicating that it may drive androgen-dependent hypomethylation in divergent mammalian species. In characterizing these sites, we identify biologically plausible mechanisms explaining how androgens drive male-accelerated aging.


Subject(s)
Aging/genetics , Androgens/deficiency , DNA Methylation , Epigenesis, Genetic , Feminization/veterinary , Orchiectomy/veterinary , Sheep, Domestic/physiology , Animals , Biological Clocks , Female , Feminization/metabolism , Male , Sheep, Domestic/surgery
6.
Nat Commun ; 11(1): 4529, 2020 09 10.
Article in English | MEDLINE | ID: mdl-32913184

ABSTRACT

Although Huntington's disease (HD) is a well studied Mendelian genetic disorder, less is known about its associated epigenetic changes. Here, we characterize DNA methylation levels in six different tissues from 3 species: a mouse huntingtin (Htt) gene knock-in model, a transgenic HTT sheep model, and humans. Our epigenome-wide association study (EWAS) of human blood reveals that HD mutation status is significantly (p < 10-7) associated with 33 CpG sites, including the HTT gene (p = 6.5 × 10-26). These Htt/HTT associations were replicated in the Q175 Htt knock-in mouse model (p = 6.0 × 10-8) and in the transgenic sheep model (p = 2.4 × 10-88). We define a measure of HD motor score progression among manifest HD cases based on multiple clinical assessments. EWAS of motor progression in manifest HD cases exhibits significant (p < 10-7) associations with methylation levels at three loci: near PEX14 (p = 9.3 × 10-9), GRIK4 (p = 3.0 × 10-8), and COX4I2 (p = 6.5 × 10-8). We conclude that HD is accompanied by profound changes of DNA methylation levels in three mammalian species.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Huntingtin Protein/genetics , Huntington Disease/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Animals, Genetically Modified , Behavior, Animal , CpG Islands/genetics , Cross-Sectional Studies , Disease Models, Animal , Disease Progression , Female , Follow-Up Studies , Gene Knock-In Techniques , Genetic Loci , Genome-Wide Association Study , Global Burden of Disease , Humans , Huntington Disease/blood , Huntington Disease/diagnosis , Huntington Disease/epidemiology , Longitudinal Studies , Male , Mice , Middle Aged , Mutation , Prospective Studies , Recombinant Proteins/genetics , Registries/statistics & numerical data , Severity of Illness Index , Sheep , Young Adult
7.
J Neurosci Methods ; 171(1): 174-9, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18367250

ABSTRACT

Mutant protein aggregates are an important biomarker in Huntington's and other neurodegenerative diseases however their quantification has typically relied on manual imaging and counting, or cell-free assays, which do not allow for concurrent analysis of cell viability. Here we describe four automated high throughput image analysis methods, developed using Metamorph software, to quantify mutant huntingtin aggregates in a cellular context. Imaging of aggregate-forming cells was also automated, using a Discovery-1 automated fluorescence microscope. All four analysis methods measured aggregate formation accurately in relation to manual counting, but with differing throughput. Our in-house PolyQ assay gave the highest throughput, processing images at 0.31 s per image. The Cell Scoring assay gave lower throughput, at 19.5s per image, but offered accurate quantification of the proportion of cells which formed aggregates, without bias from cell death. These image analysis tools provide rapid and objective alternatives to manual counting in studies of aggregate formation, to facilitate the discovery of drugs to treat Huntington's and related neurodegenerative diseases.


Subject(s)
Diagnostic Imaging/methods , Image Processing, Computer-Assisted/methods , Mutation/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion/genetics , Animals , Cell Aggregation/genetics , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression/drug effects , Gene Expression/genetics , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Huntingtin Protein , Hydrazines/pharmacology , PC12 Cells , Protein Transport/drug effects , Protein Transport/genetics , Rats , Time Factors , Transfection/methods
8.
Clin Exp Pharmacol Physiol ; 35(11): 1377-82, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18565191

ABSTRACT

1. Receptor transport between intracellular compartments has important consequences for receptor function and is an exciting area of current study. Existing methods for studying receptor trafficking often require labour-intensive techniques or are difficult to quantify reliably. We report a novel high-throughput method that uses automated imaging and analysis tools to accurately quantify cannabinoid CB1 receptor trafficking. 2. Haemagglutinin (HA)-tagged CB1 was stably expressed in HEK-293 cells and cell surface or total receptors were detected immunocytochemically. Images of receptor and nuclear staining were acquired with an automated fluorescent microscope (Discovery-1; Molecular Devices, Sunnyvale, CA, USA) and quantified at high throughput with MetaMorph (Molecular Devices) software. The 'Granularity' assay measured internalization by counting receptor clusters that appear during receptor endocytosis, a well-established approach. Our assay, referred to as 'Total Grey Value per Cell' (TGVC), measures the total fluorescence above background, normalized to cell count. 3. Incubation with the cannabinoid agonist HU-210 (100 nmol/L) resulted in rapid CB1 internalization, reaching a maximum within 20 min. Whether quantified by Granularity or TGVC, the time-course of endocytosis could be modelled with exponentially derived curves and with similar half-lives. We demonstrate the sensitivity of our TGVC method by measuring the concentration dependence of CB1 internalization and its versatility by measuring downregulation following chronic agonist exposure, whereby total CB1 was reduced to approximately 55% of basal after 3 h. 4. The TGVC quantification method described is efficient, accurate and versatile and is likely to provide a valuable tool in receptor trafficking studies.


Subject(s)
Cell Count/methods , Receptor, Cannabinoid, CB1/metabolism , Animals , Cell Line , Dronabinol/analogs & derivatives , Dronabinol/pharmacology , Endocytosis/drug effects , Endocytosis/physiology , Humans , Microscopy, Fluorescence/methods , Protein Transport/drug effects , Protein Transport/physiology , Rats , Receptor, Cannabinoid, CB1/analysis
9.
J Vis Exp ; (133)2018 03 31.
Article in English | MEDLINE | ID: mdl-29658914

ABSTRACT

Several dyes are currently available for use in detecting differentiation of mesenchymal cells into adipocytes. Dyes, such as Oil Red O, are cheap, easy to use and widely utilized by laboratories analyzing the adipogenic potential of mesenchymal cells. However, they are not specific to changes in gene transcription. We have developed a gene-specific differentiation assay to analyze when a mesenchymal cell has switched its fate to an adipogenic lineage. Immuno-labelling against fatty acid binding protein-4 (FABP4), a lineage-specific marker of adipogenic differentiation, enabled visualization and quantification of differentiated cells. The ability to quantify adipogenic differentiation potential of mesenchymal cells in a 96 well microplate format has promising implications for a number of applications. Hundreds of clinical trials involve the use of adult mesenchymal stromal cells and it is currently difficult to correlate therapeutic outcomes within and especially between such clinical trials. This simple high-throughput FABP4 assay provides a quantitative assay for assessing the differentiation potential of patient-derived cells and is a robust tool for comparing different isolation and expansion methods. This is particularly important given the increasing recognition of the heterogeneity of the cells being administered to patients in mesenchymal cell products. The assay also has potential utility in high throughput drug screening, particularly in obesity and pre-diabetes research.


Subject(s)
Adipogenesis/genetics , Biomarkers/metabolism , Cell Lineage/genetics , Immunohistochemistry/methods , Mesenchymal Stem Cells/metabolism , Cell Differentiation , Cells, Cultured , Humans , Mesenchymal Stem Cells/cytology
10.
Mol Neurodegener ; 13(1): 44, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30124174

ABSTRACT

BACKGROUND: Microglia play critical roles in the brain during homeostasis and pathological conditions. Understanding the molecular events underpinning microglial functions and activation states will further enable us to target these cells for the treatment of neurological disorders. The transcription factor PU.1 is critical in the development of myeloid cells and a major regulator of microglial gene expression. In the brain, PU.1 is specifically expressed in microglia and recent evidence from genome-wide association studies suggests that reductions in PU.1 contribute to a delayed onset of Alzheimer's disease (AD), possibly through limiting neuroinflammatory responses. METHODS: To investigate how PU.1 contributes to immune activation in human microglia, microarray analysis was performed on primary human mixed glial cultures subjected to siRNA-mediated knockdown of PU.1. Microarray hits were confirmed by qRT-PCR and immunocytochemistry in both mixed glial cultures and isolated microglia following PU.1 knockdown. To identify attenuators of PU.1 expression in microglia, high throughput drug screening was undertaken using a compound library containing FDA-approved drugs. NanoString and immunohistochemistry was utilised to investigate the expression of PU.1 itself and PU.1-regulated mediators in primary human brain tissue derived from neurologically normal and clinically and pathologically confirmed cases of AD. RESULTS: Bioinformatic analysis of gene expression upon PU.1 silencing in mixed glial cultures revealed a network of modified AD-associated microglial genes involved in the innate and adaptive immune systems, particularly those involved in antigen presentation and phagocytosis. These gene changes were confirmed using isolated microglial cultures. Utilising high throughput screening of FDA-approved compounds in mixed glial cultures we identified the histone deacetylase inhibitor vorinostat as an effective attenuator of PU.1 expression in human microglia. Further characterisation of vorinostat in isolated microglial cultures revealed gene and protein changes partially recapitulating those seen following siRNA-mediated PU.1 knockdown. Lastly, we demonstrate that several of these PU.1-regulated genes are expressed by microglia in the human AD brain in situ. CONCLUSIONS: Collectively, these results suggest that attenuating PU.1 may be a valid therapeutic approach to limit microglial-mediated inflammatory responses in AD and demonstrate utility of vorinostat for this purpose.


Subject(s)
Alzheimer Disease/metabolism , Gene Expression Regulation/physiology , Microglia/metabolism , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Gene Expression Regulation/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Microglia/drug effects , Vorinostat/pharmacology
12.
J Biomol Screen ; 12(7): 999-1005, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17942793

ABSTRACT

A number of biochemical methods are available for measuring fat accumulation in cell culture. The authors report a simple image-based method for measuring fat accumulation in adipocytes using a combination of high-throughput brightfield microscopy and image analysis, which was validated biochemically using Oil-Red-O. The quickest and most accurate method of analysis was one based on thresholding brightfield images and determining the area of fat droplets per image. Thus, the authors have developed a simple high-throughput, label-free method for measuring fat accumulation that is applicable to any cell or tissue type where fat droplets are visible under light microscopy.


Subject(s)
Fats/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Mice
13.
J Neurosci Methods ; 164(2): 339-49, 2007 Aug 30.
Article in English | MEDLINE | ID: mdl-17559941

ABSTRACT

Automated image-based and biochemical assays have greatly increased throughput for quantifying cell numbers in in vitro studies. However, it has been more difficult to automate the counting of specific cell types with complex morphologies in mixed cell cultures. We have developed a fully automated, fast, accurate and objective method for the quantification of primary human GFAP-positive astrocytes and CD45-positive microglia from images of mixed cell populations. This method, called the complex cell count (CCC) assay, utilizes a combination of image processing and analysis operations from MetaMorph (Version 6.2.6, Molecular Devices). The CCC assay consists of four main aspects: image processing with a unique combination of morphology filters; digital thresholding; integrated morphometry analysis; and a configuration of object standards. The time needed to analyze each image is 1.82s. Significant correlations have been consistently achieved between the data obtained from CCC analysis and manual cell counts. This assay can quickly and accurately quantify the number of human astrocytes and microglia in mixed cell culture and can be applied to quantifying a range of other cells/objects with complex morphology in neuroscience research.


Subject(s)
Brain/cytology , Cell Count/methods , Diagnostic Imaging/methods , Image Processing, Computer-Assisted/methods , Neurons/cytology , Neurons/physiology , Adult , Cells, Cultured , Female , Humans , Male , Middle Aged , Statistics as Topic
14.
J Neurosci Methods ; 166(1): 89-98, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17719090

ABSTRACT

Microglia and astrocytes play vital roles in normal human brain function and in neurological disorders. To study their physiological and pathological roles it is desirable to establish in vitro systems that are derived from the adult human brain. Although several groups have successfully cultured cells from the human brain, the composition of these cultures remains controversial. Using morphological criteria, immunocytochemical analysis and a BrdU incorporation assay we demonstrate the presence of poorly proliferative microglia and astrocytes in cultures derived from epilepsy biopsy tissue. In addition, we characterized a third cell type as fibronectin and prolyl 4-hydroxylase immunopositive fibroblast-like cells, which are highly proliferative and become the predominant cell type after successive sub-culturing. Therefore, although cultures from adult human brain tissue provide an excellent resource for studying human glial cells, careful consideration must be given to their cellular composition when performing studies using these methods.


Subject(s)
Brain/cytology , Cell Proliferation , Neuroglia/cytology , Adult , Astrocytes/cytology , Astrocytes/metabolism , Biomarkers/metabolism , Biopsy , Brain/metabolism , Bromodeoxyuridine , Cell Culture Techniques/methods , Cell Shape/physiology , Cell Survival/physiology , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Fibronectins/metabolism , Humans , Immunohistochemistry , Microglia/cytology , Microglia/metabolism , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Procollagen-Proline Dioxygenase/metabolism
15.
Brain Res ; 1164: 1-13, 2007 Aug 20.
Article in English | MEDLINE | ID: mdl-17644078

ABSTRACT

Glial scar formation occurs after virtually any injury to the brain. The migration of astrocytes into regions of brain injury underlies the formation of the glial scar. The exact role of the glial scar has yet to be elucidated, although it is likely to impair brain recovery. Understanding astrocyte migration is fundamental to understanding the formation of the glial scar. We have used human astrocytes (NT2A cells), derived from human NT2/D1 precursor cells to study astrocyte migration using an in vitro scratch wound model. Time-lapse microscopy and bromodeoxyuridine labeling revealed that the astrocytes migrated rather than proliferated across the scratch. Time course immunocytochemical studies showed that scratching human astrocytes induced the activation (phosphorylation) of ERK 1/2 at 10 min after scratch. The MEK 1/2 inhibitor U0126 inhibited both the ERK 1/2 phosphorylation and the migration of the astrocytes across the wound after scratch. Thus, the migration of human astrocytes after injury is partly initiated by activation of the MEK-ERK signalling pathway.


Subject(s)
Astrocytes/metabolism , Cell Movement/physiology , Cicatrix/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Gliosis/metabolism , Brain Injuries/metabolism , Brain Injuries/physiopathology , Cell Line, Tumor , Cell Proliferation , Cicatrix/physiopathology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Gliosis/physiopathology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Nerve Regeneration/physiology , Phosphorylation/drug effects
16.
J Neurosci Methods ; 247: 41-9, 2015 May 30.
Article in English | MEDLINE | ID: mdl-25813427

ABSTRACT

BACKGROUND: Tissue microarrays are commonly used to evaluate disease pathology however methods to automate and quantify pathological changes are limited. NEW METHOD: This article demonstrates the utility of the VSlide scanner (MetaSystems) for automated image acquisition from immunolabelled tissue microarray slides, and subsequent automated image analysis with MetaXpress (Molecular Devices) software to obtain objective, efficient and reproducible data from immunolabelled tissue microarray sections. RESULTS: Significant increases in fibrinogen immunolabelling were observed in 29 Alzheimer's disease cases compared to 28 control cases analysed from a single tissue microarray slide. Western blot analysis also demonstrated significant increases in fibrinogen immunolabelling in 6 Alzheimer's cases compared to 6 control cases. The observed changes were also validated with gold standard blinded manual H-scoring. COMPARISON WITH EXISTING METHOD: VSlide Metafer software offers a 'tissue microarray acquisition' plugin for easy mapping of tissue cores with their original position on the tissue microarray map. High resolution VSlide images are compatible with MetaXpress image analysis software. This article details the coupling of these two technologies to accurately and reproducibly analyse immunolabelled tissue microarrays within minutes, compared to the gold standard method of manual counting using H-scores which is significantly slower and prone to inter-observer variation. CONCLUSIONS: Here, we couple brain tissue microarray technology with high-content screening and automated image analysis as a powerful way to address bottle necks in data generation and improve throughput, as well as sensitivity to study biological/pathological changes in brain disease.


Subject(s)
Alzheimer Disease/pathology , Brain/pathology , Fibrinogen/analysis , Image Processing, Computer-Assisted/methods , Tissue Array Analysis/methods , Aged , Female , Humans , Male
17.
J Neurosci Methods ; 193(1): 54-61, 2010 Oct 30.
Article in English | MEDLINE | ID: mdl-20817035

ABSTRACT

There is increasing demand for automated image analysis of cell nuclei to be fast, objective and informative. Here, we have developed a high content analysis method for quantifying histone acetylation within any given population of cells. To demonstrate the utility of this method we quantified the effect of valproic acid (VPA) on histone H3 acetylation levels in SK-N-SH cells, a human neuroblastomal cell line. VPA, commonly used for treatment of bipolar disorder and epilepsy, has also been shown to act as a histone deacetylase inhibitor (HDACi), and to maintain the N-terminals of susceptible histones in an acetylated and transcriptionally active state. The Discovery-1™ (Molecular Devices) platform was used for automated image acquisition of immunolabelled cells. Multiple parameters of labelled nuclei were analysed in 1.82 s per image using the built-in count nuclei assay from MetaMorph™ (Molecular Devices) image analysis software. Data were presented in two forms: summary graphs or heterogeneity profiles using frequency distributions within GraphPad Prism (SmartDrawNet). Results showed that VPA increased histone H3 acetylation in a concentration- and time-dependent manner in SK-N-SH cells. The same analysis was shown to accurately quantify histone acetylation changes in human tissue sections also. Trichostatin A, a known HDACi was used to validate VPA action. Western blotting was used to validate the specificity of the antibodies. Overall these data demonstrate that this novel method for quantifying average treatment effects and the heterogeneity within any given population of cells, is fast, reproducible and can be applied to many different cellular contexts (immunocyto- and immunohisto-chemistry).


Subject(s)
Histones/analysis , Histones/metabolism , Immunohistochemistry/methods , Acetylation/drug effects , Analysis of Variance , Blotting, Western , Brain/metabolism , Cell Count , Cell Line, Tumor , Cell Nucleus/metabolism , Dose-Response Relationship, Drug , Histone Deacetylase Inhibitors/pharmacology , Humans , Time Factors , Valproic Acid/pharmacology
18.
Br J Pharmacol ; 159(2): 285-303, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20015091

ABSTRACT

Epigenetics is a rapidly growing field and holds great promise for a range of human diseases, including brain disorders such as Rett syndrome, anxiety and depressive disorders, schizophrenia, Alzheimer disease and Huntington disease. This review is concerned with the pharmacology of epigenetics to treat disorders of the epigenome whether induced developmentally or manifested/acquired later in life. In particular, we will focus on brain disorders and their treatment by drugs that modify the epigenome. While the use of DNA methyl transferase inhibitors and histone deacetylase inhibitors in in vitro and in vivo models have demonstrated improvements in disease-related deficits, clinical trials in humans have been less promising. We will address recent advances in our understanding of the complexity of the epigenome with its many molecular players, and discuss evidence for a compromised epigenome in the context of an ageing or diseased brain. We will also draw on examples of species differences that may exist between humans and model systems, emphasizing the need for more robust pre-clinical testing. Finally, we will discuss fundamental issues to be considered in study design when targeting the epigenome.


Subject(s)
Brain Diseases/genetics , Epigenesis, Genetic/drug effects , Mental Disorders/genetics , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Animals , Anthracyclines/pharmacology , Brain Diseases/drug therapy , DNA Methylation/drug effects , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Huntington Disease/drug therapy , Huntington Disease/genetics , Mental Disorders/drug therapy , MicroRNAs/physiology , Mutation , Schizophrenia/drug therapy , Schizophrenia/genetics
19.
J Biol Chem ; 281(39): 29085-95, 2006 Sep 29.
Article in English | MEDLINE | ID: mdl-16864584

ABSTRACT

Neuro2a cells endogenously express cannabinoid type 1 (CB1) receptors. CB1 stimulation with HU210 activated ERK and induced the transcription factor Krox-24. A functional MEK-ERK pathway is an important requirement for CB1-mediated Krox-24 induction as blockade of MEK signaling by UO126 reduces both basal and CB1-mediated activation of Krox-24. CB1 receptor stimulation did not activate either JNK or p38 MAPK pathways or the pro-proliferation phosphatidylinositol 3-kinase (PI3K)-Akt pathway. However, serum removal or blockade of PI3K signaling by LY294002 transiently stimulated basal Krox-24 expression and increased CB1-mediated induction of Krox-24. This was consistent with a transient increase in pMEK, pERK, and pCREB levels following PI3K blockade. These data demonstrate that CB1-mediated activation of the Krox-24 transcription factor is negatively regulated through the PI3K-Akt pathway and reveals several points of signaling cross-talk between these two important kinase pathways.


Subject(s)
Early Growth Response Protein 1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Cannabinoid/metabolism , Animals , Cell Line , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , MAP Kinase Signaling System , Mice , Models, Biological , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL