Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cell ; 181(2): 382-395.e21, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32246942

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease characterized by attack on oligodendrocytes within the central nervous system (CNS). Despite widespread use of immunomodulatory therapies, patients may still face progressive disability because of failure of myelin regeneration and loss of neurons, suggesting additional cellular pathologies. Here, we describe a general approach for identifying specific cell types in which a disease allele exerts a pathogenic effect. Applying this approach to MS risk loci, we pinpoint likely pathogenic cell types for 70%. In addition to T cell loci, we unexpectedly identified myeloid- and CNS-specific risk loci, including two sites that dysregulate transcriptional pause release in oligodendrocytes. Functional studies demonstrated inhibition of transcriptional elongation is a dominant pathway blocking oligodendrocyte maturation. Furthermore, pause release factors are frequently dysregulated in MS brain tissue. These data implicate cell-intrinsic aberrations outside of the immune system and suggest new avenues for therapeutic development. VIDEO ABSTRACT.


Subject(s)
Cell Communication/genetics , Disease/genetics , Oligodendroglia/metabolism , Animals , Brain/metabolism , Central Nervous System/metabolism , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Humans , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/physiopathology , Myelin Sheath/metabolism , Neurons/metabolism , Oligodendroglia/physiology , Risk Factors
2.
Proc Natl Acad Sci U S A ; 117(45): 28422-28432, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33109720

ABSTRACT

The human cerebral cortex contains many cell types that likely underwent independent functional changes during evolution. However, cell-type-specific regulatory landscapes in the cortex remain largely unexplored. Here we report epigenomic and transcriptomic analyses of the two main cortical neuronal subtypes, glutamatergic projection neurons and GABAergic interneurons, in human, chimpanzee, and rhesus macaque. Using genome-wide profiling of the H3K27ac histone modification, we identify neuron-subtype-specific regulatory elements that previously went undetected in bulk brain tissue samples. Human-specific regulatory changes are uncovered in multiple genes, including those associated with language, autism spectrum disorder, and drug addiction. We observe preferential evolutionary divergence in neuron subtype-specific regulatory elements and show that a substantial fraction of pan-neuronal regulatory elements undergoes subtype-specific evolutionary changes. This study sheds light on the interplay between regulatory evolution and cell-type-dependent gene-expression programs, and provides a resource for further exploration of human brain evolution and function.


Subject(s)
Cerebral Cortex/metabolism , Evolution, Molecular , Neurons/metabolism , Animals , Autism Spectrum Disorder/genetics , Brain/metabolism , Epigenesis, Genetic , Epigenomics , Gene Expression , Histone Code , Humans , Interneurons/metabolism , Macaca mulatta/genetics , Pan troglodytes/genetics , Primates/genetics , Regulatory Elements, Transcriptional , Regulatory Sequences, Nucleic Acid , Transcriptome
3.
Mol Psychiatry ; 24(5): 653-673, 2019 05.
Article in English | MEDLINE | ID: mdl-29955163

ABSTRACT

Delineating the pathophysiology of psychiatric disorders has been extremely challenging but technological advances in recent decades have facilitated a deeper interrogation of molecular processes in the human brain. Initial candidate gene expression studies of the postmortem brain have evolved into genome wide profiling of the transcriptome and the epigenome, a critical regulator of gene expression. Here, we review the potential and challenges of direct molecular characterization of the postmortem human brain, and provide a brief overview of recent transcriptional and epigenetic studies with respect to neuropsychiatric disorders. Such information can now be leveraged and integrated with the growing number of genome-wide association databases to provide a functional context of trait-associated genetic variants linked to psychiatric illnesses and related phenotypes. While it is clear that the field is still developing and challenges remain to be surmounted, these recent advances nevertheless hold tremendous promise for delineating the neurobiological underpinnings of mental diseases and accelerating the development of novel medication strategies.


Subject(s)
Brain/physiopathology , Mental Disorders/genetics , Mental Disorders/physiopathology , Autopsy/methods , Epigenesis, Genetic/genetics , Gene Expression Regulation/genetics , Genetic Predisposition to Disease/genetics , Genome/genetics , Genome-Wide Association Study , Genotype , Humans , Phenotype , Transcriptome/genetics
4.
Nucleic Acids Res ; 44(6): 2593-612, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26612861

ABSTRACT

The brain is built from a large number of cell types which have been historically classified using location, morphology and molecular markers. Recent research suggests an important role of epigenetics in shaping and maintaining cell identity in the brain. To elucidate the role of DNA methylation in neuronal differentiation, we developed a new protocol for separation of nuclei from the two major populations of human prefrontal cortex neurons--GABAergic interneurons and glutamatergic (GLU) projection neurons. Major differences between the neuronal subtypes were revealed in CpG, non-CpG and hydroxymethylation (hCpG). A dramatically greater number of undermethylated CpG sites in GLU versus GABA neurons were identified. These differences did not directly translate into differences in gene expression and did not stem from the differences in hCpG methylation, as more hCpG methylation was detected in GLU versus GABA neurons. Notably, a comparable number of undermethylated non-CpG sites were identified in GLU and GABA neurons, and non-CpG methylation was a better predictor of subtype-specific gene expression compared to CpG methylation. Regions that are differentially methylated in GABA and GLU neurons were significantly enriched for schizophrenia risk loci. Collectively, our findings suggest that functional differences between neuronal subtypes are linked to their epigenetic specification.


Subject(s)
DNA Methylation , Epigenesis, Genetic , GABAergic Neurons/metabolism , Genetic Loci , Neurons/metabolism , Prefrontal Cortex/metabolism , Adult , Autopsy , Brain Mapping , CpG Islands , GABAergic Neurons/cytology , Glutamic Acid/metabolism , Humans , Male , Microtomy , Middle Aged , Neurons/cytology , Organ Specificity , Prefrontal Cortex/anatomy & histology , Risk Factors , Schizophrenia/genetics , Schizophrenia/metabolism , Schizophrenia/pathology
5.
Hum Mol Genet ; 23(18): 4801-13, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24781207

ABSTRACT

Editing of the pre-mRNA for the serotonin receptor 2C (5-HT2CR) by site-specific adenosine deamination (A-to-I pre-mRNA editing) substantially increases the functional plasticity of this key neurotransmitter receptor and is thought to contribute to homeostatic mechanisms in neurons. 5-HT2CR mRNA editing generates up to 24 different receptor isoforms. The extent of editing correlates with 5-HT2CR functional activity: more highly edited isoforms exhibit the least function. Altered 5-HT2CR editing has been reported in postmortem brains of suicide victims. We report a comparative analysis of the connections among 5-HT2CR editing, genome-wide gene expression and DNA methylation in suicide victims, individuals with major depressive disorder and non-psychiatric controls. The results confirm previous findings of an overrepresentation of highly edited mRNA variants (which encode hypoactive 5-HT2CR receptors) in the brains of suicide victims. A large set of genes for which the expression level is associated with editing was detected. This signature set of editing-associated genes is significantly enriched for genes that are involved in synaptic transmission, genes that are preferentially expressed in neurons, and genes whose expression is correlated with the level of DNA methylation. Notably, we report that the link between 5-HT2CR editing and gene expression is disrupted in suicide victims. The results suggest that the postulated homeostatic function of 5-HT2CR editing is dysregulated in individuals who committed suicide.


Subject(s)
Depressive Disorder, Major/genetics , Gene Regulatory Networks , Prefrontal Cortex/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Suicide , Autopsy , Case-Control Studies , DNA Methylation , Gene Expression Profiling , Humans , Neurons/metabolism , RNA Editing , Receptor, Serotonin, 5-HT2C/metabolism
6.
EMBO J ; 31(17): 3635-46, 2012 Aug 29.
Article in English | MEDLINE | ID: mdl-22850675

ABSTRACT

Stomatin proteins oligomerize at membranes and have been implicated in ion channel regulation and membrane trafficking. To obtain mechanistic insights into their function, we determined three crystal structures of the conserved stomatin domain of mouse stomatin that assembles into a banana-shaped dimer. We show that dimerization is crucial for the repression of acid-sensing ion channel 3 (ASIC3) activity. A hydrophobic pocket at the inside of the concave surface is open in the presence of an internal peptide ligand and closes in the absence of this ligand, and we demonstrate a function of this pocket in the inhibition of ASIC3 activity. In one crystal form, stomatin assembles via two conserved surfaces into a cylindrical oligomer, and these oligomerization surfaces are also essential for the inhibition of ASIC3-mediated currents. The assembly mode of stomatin uncovered in this study might serve as a model to understand oligomerization processes of related membrane-remodelling proteins, such as flotillin and prohibitin.


Subject(s)
Acid Sensing Ion Channels/metabolism , Blood Proteins/metabolism , Membrane Proteins/metabolism , Acid Sensing Ion Channels/chemistry , Animals , Blood Proteins/chemistry , Blood Proteins/genetics , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Dimerization , Fibroblasts , HEK293 Cells , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Transgenic , Protein Structure, Tertiary , Rats
7.
Nucleic Acids Res ; 42(1): 109-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24057217

ABSTRACT

We applied Illumina Human Methylation450K array to perform a genomic-scale single-site resolution DNA methylation analysis in neuronal and nonneuronal (primarily glial) nuclei separated from the orbitofrontal cortex of postmortem human brain. The findings were validated using enhanced reduced representation bisulfite sequencing. We identified thousands of sites differentially methylated (DM) between neuronal and nonneuronal cells. The DM sites were depleted within CpG-island-containing promoters but enriched in predicted enhancers. Classification of the DM sites into those undermethylated in neurons (neuronal type) and those undermethylated in nonneuronal cells (glial type), combined with findings of others that methylation within control elements typically negatively correlates with gene expression, yielded large sets of predicted neuron-specific and non-neuron-specific genes. These sets of predicted genes were in excellent agreement with the available direct measurements of gene expression in human and mouse. We also found a distinct set of DNA methylation patterns that were unique for neuronal cells. In particular, neuronal-type differential methylation was overrepresented in CpG island shores, enriched within gene bodies but not in intergenic regions, and preferentially harbored binding motifs for a distinct set of transcription factors, including neuron-specific activity-dependent factors. Finally, non-CpG methylation was substantially more prevalent in neurons than in nonneuronal cells.


Subject(s)
Brain/metabolism , DNA Methylation , Enhancer Elements, Genetic , Neuroglia/metabolism , Neurons/metabolism , Adult , Animals , Binding Sites , Cell Nucleus/genetics , CpG Islands , Evolution, Molecular , Gene Expression , Genome, Human , Humans , Male , Mice , Nucleotide Motifs , Transcription Factors/metabolism , Transcription Initiation Site , Young Adult
8.
J Physiol ; 592(4): 557-69, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24247984

ABSTRACT

There are five mammalian stomatin-domain genes, all of which encode peripheral membrane proteins that can modulate ion channel function. Here we examined the ability of stomatin-like protein 1 (STOML1) to modulate the proton-sensitive members of the acid-sensing ion channel (ASIC) family. STOML1 profoundly inhibits ASIC1a, but has no effect on the splice variant ASIC1b. The inactivation time constant of ASIC3 is also accelerated by STOML1. We examined STOML1 null mutant mice with a ß-galactosidase-neomycin cassette gene-trap reporter driven from the STOML1 gene locus, which indicated that STOML1 is expressed in at least 50% of dorsal root ganglion (DRG) neurones. Patch clamp recordings from mouse DRG neurones identified a trend for larger proton-gated currents in neurones lacking STOML1, which was due to a contribution of effects upon both transient and sustained currents, at different pH, a finding consistent with an endogenous inhibitory function for STOML1.


Subject(s)
Acid Sensing Ion Channels/metabolism , Nerve Tissue Proteins/metabolism , Action Potentials , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Ganglia, Spinal/cytology , Hydrogen-Ion Concentration , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/physiology , Protein Subunits/metabolism
9.
Mol Pharmacol ; 83(6): 1166-75, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23508685

ABSTRACT

Serotonin 5-HT(2A) and metabotropic glutamate 2 (mGlu2) are G protein-coupled receptors suspected in the pathophysiology of psychiatric disorders, such as schizophrenia, depression, and suicide. Previous findings demonstrate that mGlu2 mRNA expression is down-regulated in brain cortical regions of 5-HT2A knockout (KO) mice. However, the molecular mechanism responsible for this alteration remains unknown. We show here repressive epigenetic changes at the promoter region of the mGlu2 gene in frontal cortex of 5-HT(2A)-KO mice. Disruption of 5-HT(2A) receptor-dependent signaling in mice was associated with decreased acetylation of histone H3 (H3ac) and H4 (H4ac) and increased tri-methylation of histone H3 at lysine 27 (H3K27me3) at the mGlu2 promoter, epigenetic changes that correlate with transcriptional repression. Neither methylation of histone H3 at lysine 4 (H3K4me1/2/3) nor tri-methylation of histone H3 at lysine 9 (H3K9me3) was affected. We found that Egr1, a transcription factor in which promoter activity was positively regulated by the 5-HT(2A) receptor agonist 4-bromo-3,6-dimethoxybenzocyclobuten-1-yl)methylamine hydrobromide, binds less to the mGlu2 promoter in frontal cortex of 5-HT(2A)-KO, compared with wild-type mice. Furthermore, expression of mGlu2 was increased by viral-mediated gene transfer of FLAG-tagged Egr1 in mouse frontal cortex. Together, these observations suggest that 5-HT(2A) receptor-dependent signaling epigenetically affects mGlu2 transcription in mouse frontal cortex.


Subject(s)
Epigenesis, Genetic , Frontal Lobe/metabolism , Promoter Regions, Genetic , Receptor, Serotonin, 5-HT2A/genetics , Receptors, Metabotropic Glutamate/genetics , Animals , DNA Methylation , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Histones/metabolism , Mice , Mice, Knockout , Protein Binding , Protein Processing, Post-Translational , Receptors, Metabotropic Glutamate/metabolism
10.
Nature ; 445(7124): 206-9, 2007 Jan 11.
Article in English | MEDLINE | ID: mdl-17167420

ABSTRACT

Touch and mechanical pain are first detected at our largest sensory surface, the skin. The cell bodies of sensory neurons that detect such stimuli are located in the dorsal root ganglia, and subtypes of these neurons are specialized to detect specific modalities of mechanical stimuli. Molecules have been identified that are necessary for mechanosensation in invertebrates but so far not in mammals. In Caenorhabditis elegans, mec-2 is one of several genes identified in a screen for touch insensitivity and encodes an integral membrane protein with a stomatin homology domain. Here we show that about 35% of skin mechanoreceptors do not respond to mechanical stimuli in mice with a mutation in stomatin-like protein 3 (SLP3, also called Stoml3), a mammalian mec-2 homologue that is expressed in sensory neurons. In addition, mechanosensitive ion channels found in many sensory neurons do not function without SLP3. Tactile-driven behaviours are also impaired in SLP3 mutant mice, including touch-evoked pain caused by neuropathic injury. SLP3 is therefore indispensable for the function of a subset of cutaneous mechanoreceptors, and our data support the idea that this protein is an essential subunit of a mammalian mechanotransducer.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Touch/physiology , Acid Sensing Ion Channels , Afferent Pathways , Animals , Electric Conductivity , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Male , Mechanoreceptors/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mutation/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Protein Structure, Tertiary , Rats , Sodium Channels/metabolism
11.
Nat Commun ; 14(1): 5714, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37714849

ABSTRACT

A repeat expansion in the C9orf72 (C9) gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here we investigate single nucleus transcriptomics (snRNA-seq) and epigenomics (snATAC-seq) in postmortem motor and frontal cortices from C9-ALS, C9-FTD, and control donors. C9-ALS donors present pervasive alterations of gene expression with concordant changes in chromatin accessibility and histone modifications. The greatest alterations occur in upper and deep layer excitatory neurons, as well as in astrocytes. In neurons, the changes imply an increase in proteostasis, metabolism, and protein expression pathways, alongside a decrease in neuronal function. In astrocytes, the alterations suggest activation and structural remodeling. Conversely, C9-FTD donors have fewer high-quality neuronal nuclei in the frontal cortex and numerous gene expression changes in glial cells. These findings highlight a context-dependent molecular disruption in C9-ALS and C9-FTD, indicating unique effects across cell types, brain regions, and diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Humans , Frontotemporal Dementia/genetics , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , Transcriptome/genetics , Epigenome , Mutation
12.
Nat Neurosci ; 25(4): 474-483, 2022 04.
Article in English | MEDLINE | ID: mdl-35332326

ABSTRACT

Chromosomal organization, scaling from the 147-base pair (bp) nucleosome to megabase-ranging domains encompassing multiple transcriptional units, including heritability loci for psychiatric traits, remains largely unexplored in the human brain. In this study, we constructed promoter- and enhancer-enriched nucleosomal histone modification landscapes for adult prefrontal cortex from H3-lysine 27 acetylation and H3-lysine 4 trimethylation profiles, generated from 388 controls and 351 individuals diagnosed with schizophrenia (SCZ) or bipolar disorder (BD) (n = 739). We mapped thousands of cis-regulatory domains (CRDs), revealing fine-grained, 104-106-bp chromosomal organization, firmly integrated into Hi-C topologically associating domain stratification by open/repressive chromosomal environments and nuclear topography. Large clusters of hyper-acetylated CRDs were enriched for SCZ heritability, with prominent representation of regulatory sequences governing fetal development and glutamatergic neuron signaling. Therefore, SCZ and BD brains show coordinated dysregulation of risk-associated regulatory sequences assembled into kilobase- to megabase-scaling chromosomal domains.


Subject(s)
Bipolar Disorder , Schizophrenia , Adult , Bipolar Disorder/genetics , Brain , Chromatin , Humans , Lysine/genetics , Schizophrenia/genetics
13.
Nat Commun ; 12(1): 3968, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172755

ABSTRACT

Cellular heterogeneity in the human brain obscures the identification of robust cellular regulatory networks, which is necessary to understand the function of non-coding elements and the impact of non-coding genetic variation. Here we integrate genome-wide chromosome conformation data from purified neurons and glia with transcriptomic and enhancer profiles, to characterize the gene regulatory landscape of two major cell classes in the human brain. We then leverage cell-type-specific regulatory landscapes to gain insight into the cellular etiology of several brain disorders. We find that Alzheimer's disease (AD)-associated epigenetic dysregulation is linked to neurons and oligodendrocytes, whereas genetic risk factors for AD highlighted microglia, suggesting that different cell types may contribute to disease risk, via different mechanisms. Moreover, integration of glutamatergic and GABAergic regulatory maps with genetic risk factors for schizophrenia (SCZ) and bipolar disorder (BD) identifies shared (parvalbumin-expressing interneurons) and distinct cellular etiologies (upper layer neurons for BD, and deeper layer projection neurons for SCZ). Collectively, these findings shed new light on cell-type-specific gene regulatory networks in brain disorders.


Subject(s)
Alzheimer Disease/genetics , Bipolar Disorder/genetics , Chromatin/ultrastructure , Schizophrenia/genetics , Acetylation , Alzheimer Disease/pathology , Bipolar Disorder/pathology , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation Sequencing , Enhancer Elements, Genetic , Epigenesis, Genetic , GABAergic Neurons/metabolism , Gene Expression Regulation , Genome-Wide Association Study , Histones/metabolism , Humans , Lysine/metabolism , Neuroglia/pathology , Neuroglia/ultrastructure , Neurons/pathology , Neurons/ultrastructure , Promoter Regions, Genetic , Schizophrenia/pathology
14.
Nat Commun ; 11(1): 5581, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33149216

ABSTRACT

The chromatin landscape of human brain cells encompasses key information to understanding brain function. Here we use ATAC-seq to profile the chromatin structure in four distinct populations of cells (glutamatergic neurons, GABAergic neurons, oligodendrocytes, and microglia/astrocytes) from three different brain regions (anterior cingulate cortex, dorsolateral prefrontal cortex, and primary visual cortex) in human postmortem brain samples. We find that chromatin accessibility varies greatly by cell type and, more moderately, by brain region, with glutamatergic neurons showing the largest regional variability. Transcription factor footprinting implicates cell-specific transcriptional regulators and infers cell-specific regulation of protein-coding genes, long intergenic noncoding RNAs and microRNAs. In vivo transgenic mouse experiments validate the cell type specificity of several of these human-derived regulatory sequences. We find that open chromatin regions in glutamatergic neurons are enriched for neuropsychiatric risk variants, particularly those associated with schizophrenia. Integration of cell-specific chromatin data with a bulk tissue study of schizophrenia brains increases statistical power and confirms that glutamatergic neurons are most affected. These findings illustrate the utility of studying the cell-type-specific epigenome in complex tissues like the human brain, and the potential of such approaches to better understand the genetic basis of human brain function.


Subject(s)
Astrocytes/metabolism , Chromatin/metabolism , GABAergic Neurons/metabolism , Microglia/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Schizophrenia/metabolism , Animals , Chromatin/genetics , Epigenesis, Genetic , Gene Expression Regulation/genetics , Gyrus Cinguli/cytology , Gyrus Cinguli/metabolism , Humans , Mice , Mice, Transgenic , MicroRNAs/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Promoter Regions, Genetic , RNA, Long Noncoding/metabolism , Risk Factors , Schizophrenia/genetics , Transcription Factors/metabolism , Visual Cortex/cytology , Visual Cortex/metabolism
15.
Sci Adv ; 4(9): eaau6190, 2018 09.
Article in English | MEDLINE | ID: mdl-30263963

ABSTRACT

Brain function depends on interaction of diverse cell types whose gene expression and identity are defined, in part, by epigenetic mechanisms. Neuronal DNA contains two major epigenetic modifications, methylcytosine (mC) and hydroxymethylcytosine (hmC), yet their cell type-specific landscapes and relationship with gene expression are poorly understood. We report high-resolution (h)mC analyses, together with transcriptome and histone modification profiling, in three major cell types in human prefrontal cortex: glutamatergic excitatory neurons, medial ganglionic eminence-derived γ-aminobutyric acid (GABA)ergic inhibitory neurons, and oligodendrocytes. We detected a unique association between hmC and gene expression in inhibitory neurons that differed significantly from the pattern in excitatory neurons and oligodendrocytes. We also found that risk loci associated with neuropsychiatric diseases were enriched near regions of reduced hmC in excitatory neurons and reduced mC in inhibitory neurons. Our findings indicate differential roles for mC and hmC in regulation of gene expression in different brain cell types, with implications for the etiology of human brain diseases.


Subject(s)
Brain Diseases/genetics , Brain/metabolism , DNA Methylation , Epigenesis, Genetic , Neural Inhibition/physiology , Neurons/metabolism , Adult , Brain/pathology , Brain Diseases/physiopathology , Cadaver , Cell Nucleus/genetics , Humans , Male , Neurons/pathology , Polymorphism, Single Nucleotide
16.
Sci Rep ; 8(1): 8868, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29892006

ABSTRACT

Elucidating brain cell type specific gene expression patterns is critical towards a better understanding of how cell-cell communications may influence brain functions and dysfunctions. We set out to compare and contrast five human and murine cell type-specific transcriptome-wide RNA expression data sets that were generated within the past several years. We defined three measures of brain cell type-relative expression including specificity, enrichment, and absolute expression and identified corresponding consensus brain cell "signatures," which were well conserved across data sets. We validated that the relative expression of top cell type markers are associated with proxies for cell type proportions in bulk RNA expression data from postmortem human brain samples. We further validated novel marker genes using an orthogonal ATAC-seq dataset. We performed multiscale coexpression network analysis of the single cell data sets and identified robust cell-specific gene modules. To facilitate the use of the cell type-specific genes for cell type proportion estimation and deconvolution from bulk brain gene expression data, we developed an R package, BRETIGEA. In summary, we identified a set of novel brain cell consensus signatures and robust networks from the integration of multiple datasets and therefore transcend limitations related to technical issues characteristic of each individual study.


Subject(s)
Databases, Nucleic Acid , Endothelial Cells/metabolism , Neuroglia/metabolism , Neurons/metabolism , Temporal Lobe , Transcriptome , Animals , Datasets as Topic , Endothelial Cells/cytology , Female , Gene Expression Profiling , Gene Regulatory Networks , Genetic Markers , Humans , Male , Mice , Mice, Inbred C57BL , Neuroglia/cytology , Neurons/cytology , Single-Cell Analysis , Temporal Lobe/cytology , Temporal Lobe/metabolism
17.
Clin Cancer Res ; 24(6): 1355-1363, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29351917

ABSTRACT

Purpose: The classification of medulloblastoma into WNT, SHH, group 3, and group 4 subgroups has become of critical importance for patient risk stratification and subgroup-tailored clinical trials. Here, we aimed to develop a simplified, clinically applicable classification approach that can be implemented in the majority of centers treating patients with medulloblastoma.Experimental Design: We analyzed 1,577 samples comprising previously published DNA methylation microarray data (913 medulloblastomas, 457 non-medulloblastoma tumors, 85 normal tissues), and 122 frozen and formalin-fixed paraffin-embedded medulloblastoma samples. Biomarkers were identified applying stringent selection filters and Linear Discriminant Analysis (LDA) method, and validated using DNA methylation microarray data, bisulfite pyrosequencing, and direct-bisulfite sequencing.Results: Using a LDA-based approach, we developed and validated a prediction method (EpiWNT-SHH classifier) based on six epigenetic biomarkers that allowed for rapid classification of medulloblastoma into the clinically relevant subgroups WNT, SHH, and non-WNT/non-SHH with excellent concordance (>99%) with current gold-standard methods, DNA methylation microarray, and gene signature profiling analysis. The EpiWNT-SHH classifier showed high prediction capacity using both frozen and formalin-fixed material, as well as diverse DNA methylation detection methods. Similarly, we developed a classifier specific for group 3 and group 4 tumors, based on five biomarkers (EpiG3-G4) with good discriminatory capacity, allowing for correct assignment of more than 92% of tumors. EpiWNT-SHH and EpiG3-G4 methylation profiles remained stable across tumor primary, metastasis, and relapse samples.Conclusions: The EpiWNT-SHH and EpiG3-G4 classifiers represent a new simplified approach for accurate, rapid, and cost-effective molecular classification of single medulloblastoma DNA samples, using clinically applicable DNA methylation detection methods. Clin Cancer Res; 24(6); 1355-63. ©2018 AACR.


Subject(s)
Biomarkers, Tumor , Cerebellar Neoplasms/diagnosis , Cerebellar Neoplasms/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Medulloblastoma/diagnosis , Medulloblastoma/genetics , Biopsy , CpG Islands , DNA Methylation , Epigenesis, Genetic , Epigenomics/methods , Female , Gene Expression Profiling , Humans , Male , Reproducibility of Results
18.
Front Mol Neurosci ; 10: 70, 2017.
Article in English | MEDLINE | ID: mdl-28360836

ABSTRACT

Characterizing the distinct functions of the T-type ion channel subunits Cav3.1, 3.2 or 3.3 has proven difficult due to their highly conserved amino-acid sequences and the lack of pharmacological blockers specific for each subunit. To precisely determine the expression pattern of the Cav3.2 channel in the nervous system we generated two knock-in mouse strains that express EGFP or Cre recombinase under the control of the Cav3.2 gene promoter. We show that in the brains of these animals, the Cav3.2 channel is predominantly expressed in the dentate gyrus of the hippocampus. In the peripheral nervous system, the activation of the promoter starts at E9.5 in neural crest cells that will give rise to dorsal root ganglia (DRG) neurons, but not sympathetic neurons. As development progresses the number of DRG cells expressing the Cav3.2 channel reaches around 7% of the DRG at E16.5, and remains constant until E18.5. Characterization of sensory neuron subpopulations at E18.5 showed that EGFP+ cells are a heterogeneous population consisting mainly of TrkB+ and TrkC+ cells, while only a small percentage of DRG cells were TrkA+. Genetic tracing of the sensory nerve end-organ innervation of the skin showed that the activity of the Cav3.2 channel promoter in sensory progenitors marks many mechanoreceptor and nociceptor endings, but spares slowly adapting mechanoreceptors with endings associated with Merkel cells. Our genetic analysis reveals for the first time that progenitors that express the Cav3.2 T-type calcium channel, defines a sensory specific lineage that populates a large proportion of the DRG. Using our Cav3.2-Cre mice together with AAV viruses containing a conditional fluorescent reporter (tdTomato) we could also show that Cre expression is largely restricted to two functionally distinct sensory neuron types in the adult ganglia. Cav3.2 positive neurons innervating the skin were found to only form lanceolate endings on hair follicles and are probably identical to D-hair receptors. A second population of nociceptive sensory neurons expressing the Cav3.2 gene was found to be positive for the calcitonin-gene related peptide but these neurons are deep tissue nociceptors that do not innervate the skin.

19.
Genes (Basel) ; 8(6)2017 May 30.
Article in English | MEDLINE | ID: mdl-28556790

ABSTRACT

We employed Illumina 450 K Infinium microarrays to profile DNA methylation (DNAm) in neuronal nuclei separated by fluorescence-activated sorting from the postmortem orbitofrontal cortex (OFC) of heroin users who died from heroin overdose (N = 37), suicide completers (N = 22) with no evidence of heroin use and from control subjects who did not abuse illicit drugs and died of non-suicide causes (N = 28). We identified 1298 differentially methylated CpG sites (DMSs) between heroin users and controls, and 454 DMSs between suicide completers and controls (p < 0.001). DMSs and corresponding genes (DMGs) in heroin users showed significant differences in the preferential context of hyper and hypo DM. HyperDMSs were enriched in gene bodies and exons but depleted in promoters, whereas hypoDMSs were enriched in promoters and enhancers. In addition, hyperDMGs showed preference for genes expressed specifically by glutamatergic as opposed to GABAergic neurons and enrichment for axonogenesis- and synaptic-related gene ontology categories, whereas hypoDMGs were enriched for transcription factor activity- and gene expression regulation-related terms. Finally, we found that the DNAm-based "epigenetic age" of neurons from heroin users was younger than that in controls. Suicide-related results were more difficult to interpret. Collectively, these findings suggest that the observed DNAm differences could represent functionally significant marks of heroin-associated plasticity in the OFC.

20.
Nat Neurosci ; 20(9): 1247-1259, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28783139

ABSTRACT

Antipsychotic drugs remain the standard for schizophrenia treatment. Despite their effectiveness in treating hallucinations and delusions, prolonged exposure to antipsychotic medications leads to cognitive deficits in both schizophrenia patients and animal models. The molecular mechanisms underlying these negative effects on cognition remain to be elucidated. Here we demonstrate that chronic antipsychotic drug exposure increases nuclear translocation of NF-κB in both mouse and human frontal cortex, a trafficking event triggered via 5-HT2A-receptor-dependent downregulation of the NF-κB repressor IκBα. This upregulation of NF-κB activity led to its increased binding at the Hdac2 promoter, thereby augmenting Hdac2 transcription. Deletion of HDAC2 in forebrain pyramidal neurons prevented the negative effects of antipsychotic treatment on synaptic remodeling and cognition. Conversely, virally mediated activation of NF-κB signaling decreased cortical synaptic plasticity via HDAC2. Together, these observations may aid in developing therapeutic strategies to improve the outcome of schizophrenia treatment.


Subject(s)
Antipsychotic Agents/adverse effects , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Histone Deacetylase 2/metabolism , NF-kappa B/metabolism , Synapses/metabolism , Animals , Antipsychotic Agents/toxicity , Cognition Disorders/genetics , Frontal Lobe/drug effects , Frontal Lobe/metabolism , HEK293 Cells , Histone Deacetylase 2/deficiency , Histone Deacetylase 2/genetics , Humans , Male , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , Synapses/drug effects , Transcriptional Activation/drug effects , Transcriptional Activation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL