Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Neuropathol Appl Neurobiol ; 46(7): 641-653, 2020 12.
Article in English | MEDLINE | ID: mdl-32744362

ABSTRACT

In late 2014, the first epigenome-wide association studies of DNA modifications in Alzheimer's disease brain samples were published. Over the last 5 years, further studies have been reported in the field and have highlighted consistent and robust alterations in DNA modifications in AD cortex. However, there are some caveats associated with the majority of studies undertaken to date; for example, they are predominantly restricted to profiling a limited number of loci, are principally focused on DNA methylation, are performed on bulk tissue at the end stage of disease and are restricted to nominating associations rather than demonstrating causal relationships. Consequently, the downstream interpretation of these studies is limited. Owing to recent advances in state-of-the-art cell profiling techniques, long-read genomic technologies and genetic engineering methodologies, identifying cell-type-specific causal epigenetic changes is becoming feasible. This review seeks to provide an overview of the last 5 years of epigenomic studies of DNA modifications in Alzheimer's disease brain samples and propose new avenues for future research.


Subject(s)
Alzheimer Disease/genetics , Brain/metabolism , DNA/metabolism , Epigenome/genetics , Brain/pathology , DNA/genetics , DNA Methylation/physiology , Epigenesis, Genetic/genetics , Humans
2.
Mol Psychiatry ; 23(5): 1145-1156, 2018 05.
Article in English | MEDLINE | ID: mdl-28630453

ABSTRACT

In order to determine the impact of the epigenetic response to traumatic stress on post-traumatic stress disorder (PTSD), this study examined longitudinal changes of genome-wide blood DNA methylation profiles in relation to the development of PTSD symptoms in two prospective military cohorts (one discovery and one replication data set). In the first cohort consisting of male Dutch military servicemen (n=93), the emergence of PTSD symptoms over a deployment period to a combat zone was significantly associated with alterations in DNA methylation levels at 17 genomic positions and 12 genomic regions. Evidence for mediation of the relation between combat trauma and PTSD symptoms by longitudinal changes in DNA methylation was observed at several positions and regions. Bioinformatic analyses of the reported associations identified significant enrichment in several pathways relevant for symptoms of PTSD. Targeted analyses of the significant findings from the discovery sample in an independent prospective cohort of male US marines (n=98) replicated the observed relation between decreases in DNA methylation levels and PTSD symptoms at genomic regions in ZFP57, RNF39 and HIST1H2APS2. Together, our study pinpoints three novel genomic regions where longitudinal decreases in DNA methylation across the period of exposure to combat trauma marks susceptibility for PTSD.


Subject(s)
Epigenesis, Genetic , Stress Disorders, Post-Traumatic/genetics , Adult , Cohort Studies , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genetic Predisposition to Disease , Genetic Testing/methods , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Longitudinal Studies , Male , Military Personnel/psychology , Prospective Studies , Repressor Proteins , Stress Disorders, Post-Traumatic/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
Cell Mol Life Sci ; 74(3): 509-523, 2017 02.
Article in English | MEDLINE | ID: mdl-27628303

ABSTRACT

Even though the etiology of Alzheimer's disease (AD) remains unknown, it is suggested that an interplay among genetic, epigenetic and environmental factors is involved. An increasing body of evidence pinpoints that dysregulation in the epigenetic machinery plays a role in AD. Recent developments in genomic technologies have allowed for high throughput interrogation of the epigenome, and epigenome-wide association studies have already identified unique epigenetic signatures for AD in the cortex. Considerable evidence suggests that early dysregulation in the brainstem, more specifically in the raphe nuclei and the locus coeruleus, accounts for the most incipient, non-cognitive symptomatology, indicating a potential causal relationship with the pathogenesis of AD. Here we review the advancements in epigenomic technologies and their application to the AD research field, particularly with relevance to the brainstem. In this respect, we propose the assessment of epigenetic signatures in the brainstem as the cornerstone of interrogating causality in AD. Understanding how epigenetic dysregulation in the brainstem contributes to AD susceptibility could be of pivotal importance for understanding the etiology of the disease and for the development of novel diagnostic and therapeutic strategies.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/pathology , Brain Stem/pathology , DNA Methylation , Epigenesis, Genetic , Animals , Brain Stem/metabolism , Dorsal Raphe Nucleus/metabolism , Dorsal Raphe Nucleus/pathology , Humans
4.
Mol Psychiatry ; 19(1): 88-98, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23164821

ABSTRACT

Although the mechanism of Aß action in the pathogenesis of Alzheimer's disease (AD) has remained elusive, it is known to increase the expression of the antagonist of canonical wnt signalling, Dickkopf-1 (Dkk1), whereas the silencing of Dkk1 blocks Aß neurotoxicity. We asked if clusterin, known to be regulated by wnt, is part of an Aß/Dkk1 neurotoxic pathway. Knockdown of clusterin in primary neurons reduced Aß toxicity and DKK1 upregulation and, conversely, Aß increased intracellular clusterin and decreased clusterin protein secretion, resulting in the p53-dependent induction of DKK1. To further elucidate how the clusterin-dependent induction of Dkk1 by Aß mediates neurotoxicity, we measured the effects of Aß and Dkk1 protein on whole-genome expression in primary neurons, finding a common pathway suggestive of activation of wnt-planar cell polarity (PCP)-c-Jun N-terminal kinase (JNK) signalling leading to the induction of genes including EGR1 (early growth response-1), NAB2 (Ngfi-A-binding protein-2) and KLF10 (Krüppel-like factor-10) that, when individually silenced, protected against Aß neurotoxicity and/or tau phosphorylation. Neuronal overexpression of Dkk1 in transgenic mice mimicked this Aß-induced pathway and resulted in age-dependent increases in tau phosphorylation in hippocampus and cognitive impairment. Furthermore, we show that this Dkk1/wnt-PCP-JNK pathway is active in an Aß-based mouse model of AD and in AD brain, but not in a tau-based mouse model or in frontotemporal dementia brain. Thus, we have identified a pathway whereby Aß induces a clusterin/p53/Dkk1/wnt-PCP-JNK pathway, which drives the upregulation of several genes that mediate the development of AD-like neuropathologies, thereby providing new mechanistic insights into the action of Aß in neurodegenerative diseases.


Subject(s)
Amyloid beta-Peptides/toxicity , Clusterin/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System/drug effects , Neurons/drug effects , Wnt Proteins/metabolism , Aged , Alzheimer Disease/pathology , Animals , Cells, Cultured , Clusterin/genetics , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/drug effects , Humans , Intercellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
5.
Mol Psychiatry ; 16(11): 1130-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21116278

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder with considerable evidence suggesting an initiation of disease in the entorhinal cortex and hippocampus and spreading thereafter to the rest of the brain. In this study, we combine genetics and imaging data obtained from the Alzheimer's Disease Neuroimaging Initiative and the AddNeuroMed study. To identify genetic susceptibility loci for AD, we conducted a genome-wide study of atrophy in regions associated with neurodegeneration in this condition. We identified one single-nucleotide polymorphism (SNP) with a disease-specific effect associated with entorhinal cortical volume in an intron of the ZNF292 gene (rs1925690; P-value=2.6 × 10(-8); corrected P-value for equivalent number of independent quantitative traits=7.7 × 10(-8)) and an intergenic SNP, flanking the ARPP-21 gene, with an overall effect on entorhinal cortical thickness (rs11129640; P-value=5.6 × 10(-8); corrected P-value=1.7 × 10(-7)). Gene-wide scoring also highlighted PICALM as the most significant gene associated with entorhinal cortical thickness (P-value=6.7 × 10(-6)).


Subject(s)
Alzheimer Disease/genetics , Brain/pathology , Genome-Wide Association Study , Magnetic Resonance Imaging , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Alzheimer Disease/pathology , Apolipoprotein E4/genetics , Atrophy , Carrier Proteins/genetics , Disease Progression , Entorhinal Cortex/pathology , Female , Genetic Predisposition to Disease , Hippocampus/pathology , Humans , Introns , Linkage Disequilibrium , Male , Monomeric Clathrin Assembly Proteins/genetics , Nerve Tissue Proteins/genetics , Organ Size , Phosphoproteins/genetics , Risk Factors
6.
Neuroscience ; 165(3): 662-74, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-19879926

ABSTRACT

We previously detailed how intrahippocampal inoculation of C57BL/6J mice with murine modified scrapie (ME7) leads to chronic neurodegeneration (Cunningham C, Deacon R, Wells H, Boche D, Waters S, Diniz CP, Scott H, Rawlins JN, Perry VH (2003) Eur J Neurosci 17:2147-2155.). Our characterization of the ME7-model is based on inoculation of this murine modified scrapie agent into C57BL/6J mice from Harlan laboratories. This agent in the C57BL/6J host generates a disease that spans a 24-week time course. The hippocampal pathology shows progressive misfolded prion (PrP(Sc)) deposition, astrogliosis and leads to behavioural dysfunction underpinned by the early synaptic loss that precedes neuronal death. The Harlan C57BL/6J, although widely used as a wild type mouse, are a sub-strain harbouring a spontaneous deletion of alpha-synuclein with the full description C57BL/6JOlaHsd. Recently alpha-synuclein has been shown to ameliorate the synaptic loss in a mouse model lacking the synaptic chaperone CSP-alpha. This opens a potential confound of the ME7-model, particularly with respect to the signature synaptic loss that underpin the physiological and behavioural dysfunction. To investigate if this strain-selective loss of a candidate disease modifier impacts on signature ME7 pathology, we compared cohorts of C57BL/6JOlaHsd (alpha-synuclein negative) with the founder strain from Charles Rivers (C57BL/6JCrl, alpha-synuclein positive). There were subtle changes in behaviour when comparing control animals from the two sub-strains indicating potentially significant consequences for studies assuming neurobiogical identity of both strains. However, there was no evidence that the absence of alpha-synuclein modifies disease. Indeed, accumulation of PrP(Sc), synaptic loss and the behavioural dysfunction associated with the ME7-agent was the same in both genetic backgrounds. Our data suggest that alpha-synuclein deficiency does not contribute to the compartment specific processes that give rise to prion disease mediated synaptotoxicity and neurodegeneration.


Subject(s)
Disease Progression , Scrapie/physiopathology , alpha-Synuclein/deficiency , Animals , Behavior, Animal/physiology , Cohort Studies , Disease Models, Animal , Female , Hippocampus/pathology , Hippocampus/physiopathology , Mice , Mice, Inbred C57BL , PrPSc Proteins/metabolism , Random Allocation , Scrapie/pathology , Species Specificity , Synapses/pathology , Time Factors , alpha-Synuclein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL