Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Neurobiol Aging ; 141: 160-170, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38964013

ABSTRACT

Women have a higher incidence of Alzheimer's disease (AD), even after adjusting for increased longevity. Thus, there is an urgent need to identify genes that underpin sex-associated risk of AD. PIN1 is a key regulator of the tau phosphorylation signaling pathway; however, potential differences in PIN1 expression, in males and females, are still unknown. We analyzed brain transcriptomic datasets focusing on sex differences in PIN1 mRNA levels in an aging and AD cohort, which revealed reduced PIN1 levels primarily within females. We validated this observation in an independent dataset (ROS/MAP), which also revealed that PIN1 is negatively correlated with multiregional neurofibrillary tangle density and global cognitive function in females only. Additional analysis revealed a decrease in PIN1 in subjects with mild cognitive impairment (MCI) compared with aged individuals, again driven predominantly by female subjects. Histochemical analysis of PIN1 in AD and control male and female neocortex revealed an overall decrease in axonal PIN1 protein levels in females. These findings emphasize the importance of considering sex differences in AD research.

2.
bioRxiv ; 2023 Aug 16.
Article in English | MEDLINE | ID: mdl-37645898

ABSTRACT

Women have a higher incidence of Alzheimer's disease (AD), even after adjusting for increased longevity. Thus, there is an urgent need to identify the molecular networks that underpin the sex-associated risk of AD. Recent efforts have identified PIN1 as a key regulator of tau phosphorylation signaling pathway. Pin1 is the only gene, to date, that when deleted can cause both tau and Aß-related pathologies in an age-dependent manner. We analyzed multiple brain transcriptomic datasets focusing on sex differences in PIN1 mRNA levels, in an aging and AD cohort, which revealed reduced PIN1 levels driven by females. Then, we validated this observation in an independent dataset (ROS/MAP) which also revealed that PIN1 is negatively correlated with multiregional neurofibrillary tangle density and global cognitive function, in females only. Additional analysis revealed a decrease in PIN1 in subjects with mild cognitive impairment (MCI) compared with aged individuals, again, driven predominantly by female subjects. Our results show that while both male and female AD patients show decreased PIN1 expression, changes occur before the onset of clinical symptoms of AD in females and correlate to early events associated with AD risk (e.g., synaptic dysfunction). These changes are specific to neurons, and may be a potential prognostic marker to assess AD risk in the aging population and even more so in AD females with increased risk of AD.

3.
Int J Mol Sci ; 23(24)2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36555213

ABSTRACT

A reoccurring issue in neuroepigenomic studies, especially in the context of neurodegenerative disease, is the use of (heterogeneous) bulk tissue, which generates noise during epigenetic profiling. A workable solution to this issue is to quantify epigenetic patterns in individually isolated neuronal cells using laser capture microdissection (LCM). For this purpose, we established a novel approach for targeted DNA methylation profiling of individual genes that relies on a combination of LCM and limiting dilution bisulfite pyrosequencing (LDBSP). Using this approach, we determined cytosine-phosphate-guanine (CpG) methylation rates of single alleles derived from 50 neurons that were isolated from unfixed post-mortem brain tissue. In the present manuscript, we describe the general workflow and, as a showcase, demonstrate how targeted methylation analysis of various genes, in this case, RHBDF2, OXT, TNXB, DNAJB13, PGLYRP1, C3, and LMX1B, can be performed simultaneously. By doing so, we describe an adapted data analysis pipeline for LDBSP, allowing one to include and correct CpG methylation rates derived from multi-allele reactions. In addition, we show that the efficiency of LDBSP on DNA derived from LCM neurons is similar to the efficiency obtained in previously published studies using this technique on other cell types. Overall, the method described here provides the user with a more accurate estimation of the DNA methylation status of each target gene in the analyzed cell pools, thereby adding further validity to this approach.


Subject(s)
Neurodegenerative Diseases , Humans , Sequence Analysis, DNA/methods , DNA Methylation , Brain , High-Throughput Nucleotide Sequencing , Lasers , Molecular Chaperones , Apoptosis Regulatory Proteins
4.
Neurobiol Aging ; 95: 15-25, 2020 11.
Article in English | MEDLINE | ID: mdl-32745806

ABSTRACT

Whether a cell lives or dies is controlled by an array of intercepting and dynamic molecular pathways. Although there is evidence of neuronal loss in Alzheimer's disease (AD) and multiple programmed cell death (PCD) pathways have been implicated in this process, there has been no comprehensive evaluation of the dominant pathway responsible for cell death in AD. Likewise, the relative dominance of survival and PCD pathways in AD remains unclear. Here, we present the results of hypothesis-driven bioinformatic analysis of PCD and survival pathway activation in paired methylation and expression data from the middle temporal gyrus (MTG) as well as expression from laser-captured cells from the MTG and hippocampus. The results not only indicate activation of cell death pathways in AD-of which apoptosis is responsible for the largest fraction of upregulated genes-but also of cell survival pathways. These results are indicative of a complex balance between survival and death pathways in AD that future studies should work to delineate at a single cell level.


Subject(s)
Alzheimer Disease/pathology , Apoptosis , Cell Survival , Neurons/pathology , Alzheimer Disease/genetics , Computational Biology , DNA Methylation , Datasets as Topic , Epigenome , Hippocampus/cytology , Humans , Temporal Lobe/cytology , Temporal Lobe/metabolism , Transcriptome
5.
J Alzheimers Dis ; 70(3): 691-713, 2019.
Article in English | MEDLINE | ID: mdl-31256118

ABSTRACT

We used Illumina Human HT-12 v4 arrays to compare RNA expression of middle temporal gyrus (MTG; BA21) in Alzheimer's disease (AD = 97) and non-demented controls (ND = 98). A total of 938 transcripts were highly differentially expressed (adj p < 0.01; log2 FC ≥ |0.500|, with 411 overexpressed and 527 underexpressed in AD. Our results correlated with expression profiling in neurons from AD and ND obtained by laser capture microscopy in MTG from an independent dataset (log2 FC correlation: r = 0.504; p = 2.2e-16). Additionally, selected effects were validated by qPCR. ANOVA analysis yielded no difference between genders in response to AD, but some gender specific genes were detected (e.g., IL8 and AGRN in males, and HSPH1 and GRM1 in females). Several transcripts were associated with Braak staging (e.g., AEBP1 and DNALI1), antemortem MMSE (e.g., AEBP1 and GFAP), and tangle density (e.g., RNU1G2, and DNALI1). At the pathway level, we detected enrichment of synaptic vesicle processes and GABAergic transmission genes. Finally, applying the Weighted Correlation Network Analysis, we identified four expression modules enriched for neuronal and synaptic genes, mitochondria-associated membrane, chemical stimulus and olfactory receptor and non-coding RNA metabolism genes. Our results represent an extensive description of MTG mRNA profiling in a large sample of AD and ND. These data provide a list of genes associated with AD, and correlated to neurofibrillary tangles density. In addition, these data emphasize the importance of mitochondrial membranes and transcripts related to olfactory receptors in AD.


Subject(s)
Alzheimer Disease , Mitochondrial Membranes/physiology , Neurofibrillary Tangles , Neurons/physiology , Temporal Lobe/metabolism , Aged , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Autopsy , Female , Gene Expression Profiling/methods , Genetic Association Studies/methods , Humans , Male , Neurofibrillary Tangles/metabolism , Neurofibrillary Tangles/pathology , RNA, Messenger/metabolism , Transcriptome
6.
Brain Res ; 1719: 217-224, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31176712

ABSTRACT

We explored RNA expression changes in the middle temporal gyrus (MTG) of Alzheimer's Disease patients (AD) by RNA sequencing the whole transcriptome of 8 AD and 8 Non-Demented (ND) controls. We used three additional expression datasets from related brain regions to validate the findings. The results highlighted the upregulation of AEBP1 and downregulation of NRN1 in AD, as well as their association with Braak staging and neurofibrillary tangles density. Furthermore, more than 400 protein-coding RNAs enriched for "Clathrin-mediated endocytosis" were validated in independent datasets from the same brain region. Finally, using in silico prediction analysis we found a signature of 52 non-protein coding RNAs that perturb key pathways involved in GABAergic transmission and peptide chain elongation. The association of AEBP1 in our data confirmed other published work examining gene expression in the hippocampus of AD patients. NRN1 is involved in neurite outgrowth, and in previous studies it has been shown to reverse synaptic defects and cognitive function impairment in Tg2576 mice. Finally, our results on non-protein coding RNAs suggest a role of these transcripts in altering synaptic and amyloid-ß associated pathways.


Subject(s)
Alzheimer Disease/metabolism , Carboxypeptidases/genetics , Neuropeptides/genetics , Repressor Proteins/genetics , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Brain/metabolism , Carboxypeptidases/metabolism , Cognitive Dysfunction/metabolism , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression/genetics , Hippocampus/metabolism , Humans , Male , Neurofibrillary Tangles/genetics , Neurofibrillary Tangles/metabolism , Neuropeptides/metabolism , RNA/metabolism , Repressor Proteins/metabolism , Temporal Lobe/metabolism , tau Proteins/metabolism
7.
Neurobiol Aging ; 68: 134-141, 2018 08.
Article in English | MEDLINE | ID: mdl-29803514

ABSTRACT

Histone deacetylase (HDAC) inhibitors have been widely reported to have considerable therapeutic potential in a host of neurodegenerative diseases. However, HDAC inhibitor selectivity and specificity in specific cell classes have been a source of much debate. To address the role of HDAC2 in specific cell classes, and in disease, we examined glial protein and mRNA levels in the substantia nigra (SN) of Parkinson's disease (PD) and normal controls (NCs) by immunohistochemistry and laser captured microdissection followed by quantitative real time polymerase chain reaction. Differential expression analysis in immunohistochemically defined laser capture microglia revealed significant upregulation of HDAC2 in the PD SN compared to NC subjects. Complementary in vivo evidence reveals significant upregulation of HDAC2 protein levels in PD SN microglia compared to NC subjects. Correspondingly, human immortalized telencephalic/mesencephalic microglial cells reveal significant upregulation of HDAC2 in the presence of the potent microglial activator lipopolysaccharide. These data provide evidence that selective inhibition of HDAC2 in PD SN microglia could be a promising approach to treat microglial-initiated nigral dopaminergic neuronal cell loss in PD.


Subject(s)
Histone Deacetylase 2/antagonists & inhibitors , Histone Deacetylase 2/metabolism , Microglia/enzymology , Parkinson Disease/etiology , Parkinson Disease/genetics , Substantia Nigra/cytology , Substantia Nigra/enzymology , Aged , Aged, 80 and over , Cells, Cultured , Dopaminergic Neurons/pathology , Female , Histone Deacetylase 2/physiology , Humans , Immunohistochemistry , Laser Capture Microdissection , Male , Molecular Targeted Therapy , Parkinson Disease/drug therapy , Parkinson Disease/pathology , Polymerase Chain Reaction , Up-Regulation
8.
Alzheimers Dement ; 14(6): 775-786, 2018 06.
Article in English | MEDLINE | ID: mdl-29396107

ABSTRACT

INTRODUCTION: Our laboratories have demonstrated that accumulation of oligomeric amyloid ß (OAß) in neurons is an essential step leading to OAß-mediated mitochondrial dysfunction. METHODS: Alzheimer's disease (AD) and matching control hippocampal neurons, astrocytes, and microglia were isolated by laser-captured microdissection from the same subjects, followed by whole-transcriptome sequencing. Complementary in vitro work was performed in OAß-treated differentiated SH-SY5Y, followed by the use of a novel CoQ10 analogue for protection. This compound is believed to be effective both in suppressing reactive oxygen species and also functioning in mitochondrial electron transport. RESULTS: We report decreases in the same mitochondrial-encoded mRNAs in Alzheimer's disease laser-captured CA1 neurons and in OAß-treated SH-SY5Y cells, but not in laser-captured microglia and astrocytes. Pretreatment with a novel CoQ10 analogue, protects neuronal mitochondria from OAß-induced mitochondrial changes. DISCUSSION: Similarity of expression changes in neurons from Alzheimer's disease brain and neuronal cells treated with OAß, and the effect of a CoQ10 analogue on the latter, suggests a pretreatment option to prevent OAß toxicity, long before the damage is apparent.


Subject(s)
Amyloid beta-Peptides/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , RNA, Mitochondrial/metabolism , Aged , Alzheimer Disease/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Cell Line, Tumor , Female , Hippocampus/metabolism , Humans , In Vitro Techniques , Laser Capture Microdissection , Male , Microglia/drug effects , Microglia/metabolism , Microscopy, Electron, Transmission , Neurons/drug effects , RNA, Messenger/genetics , RNA, Mitochondrial/genetics , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
10.
Neurobiol Aging ; 63: 12-21, 2018 03.
Article in English | MEDLINE | ID: mdl-29207277

ABSTRACT

Expression array data from dozens of laboratories, including our own, show significant changes in expression of many genes in Alzheimer's disease (AD) patients compared with normal controls. These data typically rely on brain homogenates, and information about transcripts specific to microglia and other central nervous system (CNS) cell types, which far outnumber microglia-specific transcripts, is lost. We therefore used single-cell laser capture methods to assess the full range of microglia-specific expression changes that occur in different brain regions (substantia nigra and hippocampus CA1) and disease states (AD, Parkinson's disease, and normal controls). Two novel pathways, neuronal repair and viral processing were identified. Based on KEGG analysis (Kyoto Encyclopedia of Genes and Genomes, a collection of biological pathways), one of the most significant viruses was hepatitis B virus (HBV) (false discovery rate < 0.00000001). Immunohistochemical analysis using HBV-core antibody in HBV-positive control, amnestic mild cognitive impairment, and HBV-positive AD cases show increased HBV immunoreactivity as disease pathology increases. These results are the first, to our knowledge, to show regional differences in human microglia. In addition, these data reveal new functions for microglia and suggest a novel risk factor for AD.


Subject(s)
Alzheimer Disease/virology , Brain/virology , Hepatitis B virus , Laser Capture Microdissection , Microglia/virology , Parkinson Disease/virology , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Brain/pathology , Female , Humans , Male , Microglia/pathology , Parkinson Disease/pathology , Risk Factors
11.
PLoS One ; 12(7): e0177814, 2017.
Article in English | MEDLINE | ID: mdl-28700589

ABSTRACT

Recent epigenetic association studies have identified a new gene, ANK1, in the pathogenesis of Alzheimer's disease (AD). Although strong associations were observed, brain homogenates were used to generate the data, introducing complications because of the range of cell types analyzed. In order to address the issue of cellular heterogeneity in homogenate samples we isolated microglial, astrocytes and neurons by laser capture microdissection from CA1 of hippocampus in the same individuals with a clinical and pathological diagnosis of AD and matched control cases. Using this unique RNAseq data set, we show that in the hippocampus, ANK1 is significantly (p<0.0001) up-regulated 4-fold in AD microglia, but not in neurons or astrocytes from the same individuals. These data provide evidence that microglia are the source of ANK1 differential expression previously identified in homogenate samples in AD.


Subject(s)
Alzheimer Disease/metabolism , Ankyrins/genetics , Microglia/metabolism , Up-Regulation , Aged , Aged, 80 and over , Alzheimer Disease/pathology , Ankyrins/metabolism , Case-Control Studies , Female , Hippocampus/cytology , Hippocampus/metabolism , Humans , Male , Neurons/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Neurobiol Aging ; 58: 225-237, 2017 10.
Article in English | MEDLINE | ID: mdl-28716532

ABSTRACT

The need for a reliable, simple, and inexpensive blood test for Alzheimer's disease (AD) suitable for use in a primary care setting is widely recognized. This has led to a large number of publications describing blood tests for AD, which have, for the most part, not been replicable. We have chosen to examine transcripts expressed by the cellular, leukocyte compartment of blood. We have used hypothesis-based cDNA arrays and quantitative PCR to quantify the expression of selected sets of genes followed by multivariate analyses in multiple independent samples. Rather than a single study with no replicates, we chose an experimental design in which there were multiple replicates using different platforms and different sample populations. We have divided 177 blood samples and 27 brain samples into multiple replicates to demonstrate the ability to distinguish early clinical AD (Clinical Dementia Rating scale 0.5), Parkinson's disease (PD), and cognitively unimpaired APOE4 homozygotes, as well as to determine persons at risk for future cognitive impairment with significant accuracy. We assess our methods in a training/test set and also show that the variables we use distinguish AD, PD, and control brain. Importantly, we describe the variability of the weights assigned to individual transcripts in multivariate analyses in repeated studies and suggest that the variability we describe may be the cause of inability to repeat many earlier studies. Our data constitute a proof of principle that multivariate analysis of the transcriptome related to cell stress and inflammation of peripheral blood leukocytes has significant potential as a minimally invasive and inexpensive diagnostic tool for diagnosis and early detection of risk for AD.


Subject(s)
Alzheimer Disease/diagnosis , Cognitive Dysfunction/diagnosis , Hematologic Tests/methods , Leukocytes , Parkinson Disease/diagnosis , Transcriptome , Aged , Aged, 80 and over , Biomarkers/blood , Diagnosis, Differential , Early Diagnosis , Female , Humans , Male , Middle Aged , Multivariate Analysis , Prodromal Symptoms , Risk , Sensitivity and Specificity
13.
Alzheimers Dement ; 13(5): 510-519, 2017 May.
Article in English | MEDLINE | ID: mdl-27793643

ABSTRACT

INTRODUCTION: We have comprehensively described the expression profiles of mitochondrial DNA and nuclear DNA genes that encode subunits of the respiratory oxidative phosphorylation (OXPHOS) complexes (I-V) in the hippocampus from young controls, age matched, mild cognitively impaired (MCI), and Alzheimer's disease (AD) subjects. METHODS: Hippocampal tissues from 44 non-AD controls (NC), 10 amnestic MCI, and 18 AD cases were analyzed on Affymetrix Hg-U133 plus 2.0 arrays. RESULTS: The microarray data revealed significant down regulation in OXPHOS genes in AD, particularly those encoded in the nucleus. In contrast, there was up regulation of the same gene(s) in MCI subjects compared to AD and ND cases. No significant differences were observed in mtDNA genes identified in the array between AD, ND, and MCI subjects except one mt-ND6. DISCUSSION: Our findings suggest that restoration of the expression of nuclear-encoded OXPHOS genes in aging could be a viable strategy for blunting AD progression.


Subject(s)
Aging/genetics , Alzheimer Disease/genetics , Cognition Disorders/genetics , Mitochondria/genetics , Oxidative Phosphorylation , Adult , Aged, 80 and over , Autopsy , Female , Hippocampus , Humans , Male , Oligonucleotide Array Sequence Analysis
14.
Neuroepigenetics ; 6: 1-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27158594

ABSTRACT

We describe a novel method for assessing the "open" or "closed" state of chromatin at selected locations within the genome. This method combines the use of Benzonase, which can digest DNA in the presence of actin, with qPCR to define digested regions. We demonstrate the application of this method in brain homogenates and laser captured cells. We also demonstrate application to selected sites within more than one gene and multiple sites within one gene. We demonstrate the validity of the method by treating cells with valproate, known to render chromatin more permissive, and by comparison with classical digestion with DNase I in an in vitro preparation. Although we demonstrate the use of this method in brain tissue we also recognize its applicability to other tissue types.

15.
Neurobiol Aging ; 36(12): 3121-3129, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26553823

ABSTRACT

We have previously reported in Alzheimer's disease (AD) the mislocalization of epigenetic molecules between the cell nucleus and the cytoplasm. We have extended our finding to include the aberrant localization of histone 3 trimethylation on lysine 4 (H3k4me3), an epigenetic mark associated with actively transcribing genes as well as those poised for transcription. These findings raise the question of where the ectopic localization of H3k4me3 fits within the cascade of cell biological events in the progression of AD. We, therefore, examined the expression and intracellular location of H3k4me3 as a function of Braak stage and also in relation to a series of tau markers that are indicative of disease state. Both lines of evidence showed that ectopic localization of H3k4me3 is early in the course of disease. Because of the known role of H3k4me3 in the expression of synaptic genes, our data suggest an epigenetic role in synaptic deficits early in the course of AD.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Epigenesis, Genetic/genetics , Histones/genetics , Histones/metabolism , Aged , Aged, 80 and over , Female , Gene Expression , Humans , Lysine/metabolism , Male , Methylation , Middle Aged , Transcription, Genetic/genetics , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...