Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
J Neurosci ; 43(19): 3582-3597, 2023 05 10.
Article in English | MEDLINE | ID: mdl-37037607

ABSTRACT

Regional cellular heterogeneity is a fundamental feature of the human neocortex; however, details of this heterogeneity are still undefined. We used single-nucleus RNA-sequencing to examine cell-specific transcriptional features in the dorsolateral PFC (DLPFC) and the subgenual anterior cingulate cortex (sgACC), regions implicated in major psychiatric disorders. Droplet-based nuclei-capture and library preparation were performed on replicate samples from 8 male donors without history of psychiatric or neurologic disorder. Unsupervised clustering identified major neural cell classes. Subsequent iterative clustering of neurons further revealed 20 excitatory and 22 inhibitory subclasses. Inhibitory cells were consistently more abundant in the sgACC and excitatory neuron subclusters exhibited considerable variability across brain regions. Excitatory cell subclasses also exhibited greater within-class transcriptional differences between the two regions. We used these molecular definitions to determine which cell classes might be enriched in loci carrying a genetic signal in genome-wide association studies or for differentially expressed genes in mental illness. We found that the heritable signals of psychiatric disorders were enriched in neurons and that, while the gene expression changes detected in bulk-RNA-sequencing studies were dominated by glial cells, some alterations could be identified in specific classes of excitatory and inhibitory neurons. Intriguingly, only two excitatory cell classes exhibited concomitant region-specific enrichment for both genome-wide association study loci and transcriptional dysregulation. In sum, by detailing the molecular and cellular diversity of the DLPFC and sgACC, we were able to generate hypotheses on regional and cell-specific dysfunctions that may contribute to the development of mental illness.SIGNIFICANCE STATEMENT Dysfunction of the subgenual anterior cingulate cortex has been implicated in mood disorders, particularly major depressive disorder, and the dorsolateral PFC, a subsection of the PFC involved in executive functioning, has been implicated in schizophrenia. Understanding the cellular composition of these regions is critical to elucidating the neurobiology underlying psychiatric and neurologic disorders. We studied cell type diversity of the subgenual anterior cingulate cortex and dorsolateral PFC of humans with no neuropsychiatric illness using a clustering analysis of single-nuclei RNA-sequencing data. Defining the transcriptomic profile of cellular subpopulations in these cortical regions is a first step to demystifying the cellular and molecular pathways involved in psychiatric disorders.


Subject(s)
Depressive Disorder, Major , Dorsolateral Prefrontal Cortex , Humans , Male , Depressive Disorder, Major/metabolism , Gyrus Cinguli/metabolism , Prefrontal Cortex/physiology , Genome-Wide Association Study , Solitary Nucleus/metabolism
2.
Transl Psychiatry ; 13(1): 93, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36932057

ABSTRACT

Recent postmortem transcriptomic studies of schizophrenia (SCZ) have shown hundreds of differentially expressed genes. However, the extent to which these gene expression changes reflect antipsychotic drug (APD) exposure remains uncertain. We compared differential gene expression in the prefrontal cortex of SCZ patients who tested positive for APDs at the time of death with SCZ patients who did not. APD exposure was associated with numerous changes in the brain transcriptome, especially among SCZ patients on atypical APDs. Brain transcriptome data from macaques chronically treated with APDs showed that APDs affect the expression of many functionally relevant genes, some of which show expression changes in the same directions as those observed in SCZ. Co-expression modules enriched for synaptic function showed convergent patterns between SCZ and some of the APD effects, while those associated with inflammation and glucose metabolism exhibited predominantly divergent patterns between SCZ and APD effects. In contrast, major cell-type shifts inferred in SCZ were primarily unaffected by APD use. These results show that APDs may confound SCZ-associated gene expression changes in postmortem brain tissue. Disentangling these effects will help identify causal genes and improve our neurobiological understanding of SCZ.


Subject(s)
Antipsychotic Agents , Schizophrenia , Humans , Schizophrenia/drug therapy , Schizophrenia/genetics , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Brain/metabolism , Prefrontal Cortex/metabolism , Transcriptome
3.
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
4.
Neuropsychopharmacology ; 46(7): 1364-1372, 2021 06.
Article in English | MEDLINE | ID: mdl-33558674

ABSTRACT

Despite strong evidence of heritability and growing discovery of genetic markers for major mental illness, little is known about how gene expression in the brain differs across psychiatric diagnoses, or how known genetic risk factors shape these differences. Here we investigate expressed genes and gene transcripts in postmortem subgenual anterior cingulate cortex (sgACC), a key component of limbic circuits linked to mental illness. RNA obtained postmortem from 200 donors diagnosed with bipolar disorder, schizophrenia, major depression, or no psychiatric disorder was deeply sequenced to quantify expression of over 85,000 gene transcripts, many of which were rare. Case-control comparisons detected modest expression differences that were correlated across disorders. Case-case comparisons revealed greater expression differences, with some transcripts showing opposing patterns of expression between diagnostic groups, relative to controls. The ~250 rare transcripts that were differentially-expressed in one or more disorder groups were enriched for genes involved in synapse formation, cell junctions, and heterotrimeric G-protein complexes. Common genetic variants were associated with transcript expression (eQTL) or relative abundance of alternatively spliced transcripts (sQTL). Common genetic variants previously associated with disease risk were especially enriched for sQTLs, which together accounted for disproportionate fractions of diagnosis-specific heritability. Genetic risk factors that shape the brain transcriptome may contribute to diagnostic differences between broad classes of mental illness.


Subject(s)
Bipolar Disorder , Depressive Disorder, Major , Bipolar Disorder/genetics , Depressive Disorder, Major/genetics , Gyrus Cinguli , Humans , RNA , Transcriptome
5.
Nat Commun ; 11(1): 2990, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32533064

ABSTRACT

Structural variants (SVs) contribute to many disorders, yet, functionally annotating them remains a major challenge. Here, we integrate SVs with RNA-sequencing from human post-mortem brains to quantify their dosage and regulatory effects. We show that genic and regulatory SVs exist at significantly lower frequencies than intergenic SVs. Functional impact of copy number variants (CNVs) stems from both the proportion of genic and regulatory content altered and loss-of-function intolerance of the gene. We train a linear model to predict expression effects of rare CNVs and use it to annotate regulatory disruption of CNVs from 14,891 independent genome-sequenced individuals. Pathogenic deletions implicated in neurodevelopmental disorders show significantly more extreme regulatory disruption scores and if rank ordered would be prioritized higher than using frequency or length alone. This work shows the deleteriousness of regulatory SVs, particularly those altering CTCF sites and provides a simple approach for functionally annotating the regulatory consequences of CNVs.


Subject(s)
Brain/metabolism , DNA Copy Number Variations , Gene Expression Regulation , Genetic Variation , Genome, Human/genetics , Autopsy/methods , Brain/pathology , Female , Gene Expression Profiling/methods , Humans , Male , Neurodevelopmental Disorders/genetics , Sequence Analysis, RNA/methods
6.
Genome Med ; 12(1): 19, 2020 02 19.
Article in English | MEDLINE | ID: mdl-32075678

ABSTRACT

BACKGROUND: Midbrain dopaminergic neurons (MDN) represent 0.0005% of the brain's neuronal population and mediate cognition, food intake, and metabolism. MDN are also posited to underlay the neurobiological dysfunction of schizophrenia (SCZ), a severe neuropsychiatric disorder that is characterized by psychosis as well as multifactorial medical co-morbidities, including metabolic disease, contributing to markedly increased morbidity and mortality. Paradoxically, however, the genetic risk sequences of psychosis and traits associated with metabolic disease, such as body mass, show very limited overlap. METHODS: We investigated the genomic interaction of SCZ with medical conditions and traits, including body mass index (BMI), by exploring the MDN's "spatial genome," including chromosomal contact landscapes as a critical layer of cell type-specific epigenomic regulation. Low-input Hi-C protocols were applied to 5-10 × 103 dopaminergic and other cell-specific nuclei collected by fluorescence-activated nuclei sorting from the adult human midbrain. RESULTS: The Hi-C-reconstructed MDN spatial genome revealed 11 "Euclidean hot spots" of clustered chromatin domains harboring risk sequences for SCZ and elevated BMI. Inter- and intra-chromosomal contacts interconnecting SCZ and BMI risk sequences showed massive enrichment for brain-specific expression quantitative trait loci (eQTL), with gene ontologies, regulatory motifs and proteomic interactions related to adipogenesis and lipid regulation, dopaminergic neurogenesis and neuronal connectivity, and reward- and addiction-related pathways. CONCLUSIONS: We uncovered shared nuclear topographies of cognitive and metabolic risk variants. More broadly, our PsychENCODE sponsored Hi-C study offers a novel genomic approach for the study of psychiatric and medical co-morbidities constrained by limited overlap of their respective genetic risk architectures on the linear genome.


Subject(s)
Dopaminergic Neurons/metabolism , Polymorphism, Genetic , Quantitative Trait Loci , Schizophrenia/genetics , Adipogenesis , Animals , Body Mass Index , Chromosomes/genetics , Cognition , Humans , Lipid Metabolism , Mesencephalon/cytology , Mesencephalon/metabolism , Mice , Mice, Inbred C57BL , Neurogenesis , Schizophrenia/metabolism , Schizophrenia/pathology
7.
J Neurosci ; 39(42): 8285-8290, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31619498

ABSTRACT

Active communication between researchers and society is necessary for the scientific community's involvement in developing science-based policies. This need is recognized by governmental and funding agencies that compel scientists to increase their public engagement and disseminate research findings in an accessible fashion. Storytelling techniques can help convey science by engaging people's imagination and emotions. Yet, many researchers are uncertain about how to approach scientific storytelling, or feel they lack the tools to undertake it. Here we explore some of the techniques intrinsic to crafting scientific narratives, as well as the reasons why scientific storytelling may be an optimal way of communicating research to nonspecialists. We also point out current communication gaps between science and society, particularly in the context of neurodiverse audiences and those that include neurological and psychiatric patients. Present shortcomings may turn into areas of synergy with the potential to link neuroscience education, research, and advocacy.


Subject(s)
Communication , Information Dissemination , Journalism, Medical , Neurosciences , Humans
8.
Sci Data ; 6(1): 180, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31551426

ABSTRACT

Schizophrenia and bipolar disorder are serious mental illnesses that affect more than 2% of adults. While large-scale genetics studies have identified genomic regions associated with disease risk, less is known about the molecular mechanisms by which risk alleles with small effects lead to schizophrenia and bipolar disorder. In order to fill this gap between genetics and disease phenotype, we have undertaken a multi-cohort genomics study of postmortem brains from controls, individuals with schizophrenia and bipolar disorder. Here we present a public resource of functional genomic data from the dorsolateral prefrontal cortex (DLPFC; Brodmann areas 9 and 46) of 986 individuals from 4 separate brain banks, including 353 diagnosed with schizophrenia and 120 with bipolar disorder. The genomic data include RNA-seq and SNP genotypes on 980 individuals, and ATAC-seq on 269 individuals, of which 264 are a subset of individuals with RNA-seq. We have performed extensive preprocessing and quality control on these data so that the research community can take advantage of this public resource available on the Synapse platform at http://CommonMind.org .


Subject(s)
Bipolar Disorder , Schizophrenia , Bipolar Disorder/genetics , Bipolar Disorder/pathology , Cohort Studies , Epigenomics , Humans , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Schizophrenia/genetics , Schizophrenia/pathology , Transcriptome
9.
Lancet ; 392(10145): 378-379, 2018 08 04.
Article in English | MEDLINE | ID: mdl-30102165
10.
Nat Neurosci ; 21(8): 1126-1136, 2018 08.
Article in English | MEDLINE | ID: mdl-30038276

ABSTRACT

Risk variants for schizophrenia affect more than 100 genomic loci, yet cell- and tissue-specific roles underlying disease liability remain poorly characterized. We have generated for two cortical areas implicated in psychosis, the dorsolateral prefrontal cortex and anterior cingulate cortex, 157 reference maps from neuronal, neuron-depleted and bulk tissue chromatin for two histone marks associated with active promoters and enhancers, H3-trimethyl-Lys4 (H3K4me3) and H3-acetyl-Lys27 (H3K27ac). Differences between neuronal and neuron-depleted chromatin states were the major axis of variation in histone modification profiles, followed by substantial variability across subjects and cortical areas. Thousands of significant histone quantitative trait loci were identified in neuronal and neuron-depleted samples. Risk variants for schizophrenia, depressive symptoms and neuroticism were significantly over-represented in neuronal H3K4me3 and H3K27ac landscapes. Our Resource, sponsored by PsychENCODE and CommonMind, highlights the critical role of cell-type-specific signatures at regulatory and disease-associated noncoding sequences in the human frontal lobe.


Subject(s)
Epigenesis, Genetic/genetics , Frontal Lobe/metabolism , Frontal Lobe/pathology , Histones/genetics , Schizophrenia/genetics , Schizophrenia/metabolism , Alzheimer Disease/genetics , Brain Mapping , Chromatin/genetics , Depression/genetics , Depression/pathology , Educational Status , Genetic Predisposition to Disease/genetics , Genetic Variation , Genome-Wide Association Study , Gyrus Cinguli/pathology , Humans , Neurotic Disorders/genetics , Neurotic Disorders/pathology , Prefrontal Cortex/pathology , Risk
11.
Eur J Neurosci ; 48(3): 1884-1895, 2018 08.
Article in English | MEDLINE | ID: mdl-30033547

ABSTRACT

Dopamine transporters (DAT) are implicated in the pathogenesis and treatment of attention-deficit hyperactivity disorder (ADHD) and are upregulated by chronic treatment with methylphenidate, commonly prescribed for ADHD. Methylation of the DAT1 gene in brain and blood has been associated with DAT expression in rodents' brains. Here we tested the association between methylation of the DAT1 promoter derived from blood and DAT availability in the striatum of unmedicated ADHD adult participants and in that of healthy age-matched controls (HC) using Positron Emission Tomography (PET) and [11 C]cocaine. Results showed no between-group differences in DAT1 promoter methylation or striatal DAT availability. However, the degree of methylation in the promoter region of DAT1 correlated negatively with DAT availability in caudate in ADHD participants only. DAT availability in VS correlated with inattention scores in ADHD participants. We verified in a postmortem cohort with ADHD diagnosis and without, that DAT1 promoter methylation in peripheral blood correlated positively with DAT1 promoter methylation extracted from substantia nigra (SN) in both groups. In the cohort without ADHD diagnosis, DAT1 gene expression in SN further correlated positively with DAT protein expression in caudate; however, the sample size of the cohort with ADHD was insufficient to investigate DAT1 and DAT expression levels. Overall, these findings suggest that peripheral DAT1 promoter methylation may be predictive of striatal DAT availability in adults with ADHD. Due to the small sample size, more work is needed to validate whether DAT1 methylation in blood predicts DAT1 methylation in SN in ADHD and controls.


Subject(s)
Attention Deficit Disorder with Hyperactivity/blood , Attention Deficit Disorder with Hyperactivity/genetics , Caudate Nucleus/metabolism , DNA Methylation , Dopamine Plasma Membrane Transport Proteins/blood , Dopamine Plasma Membrane Transport Proteins/genetics , Adult , Female , Humans , Male , Promoter Regions, Genetic , Substantia Nigra/metabolism
12.
Transl Psychiatry ; 8(1): 72, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29581423

ABSTRACT

Genome-wide association studies (GWAS) of complex, heritable, behavioral phenotypes have yielded an incomplete accounting of the genetic influences. The identified loci explain only a portion of the observed heritability, and few of the loci have been shown to be functional. It is clear that current GWAS techniques overlook key components of phenotypically relevant genetic variation, either because of sample size, as is frequently asserted, or because of methodology. Here we use arginine vasopressin receptor 1a (AVPR1a) as an in-depth model of a methodologic limitation of GWAS: the functional genetic variation (in the form of short tandem repeats) of this key gene involved in affiliative behavior cannot be captured by current GWAS methodologies. Importantly, we find evidence of differential allele expression, twofold or more, in at least a third of human brain samples heterozygous for a reporter SNP in the AVPR1a transcript. We also show that this functional effect and a downstream phenotype, externalizing behavior, are predicted by AVPR1a STRs but not SNPs.


Subject(s)
Attention Deficit and Disruptive Behavior Disorders/genetics , Brain/metabolism , Gene Expression , Microsatellite Repeats , Receptors, Vasopressin/genetics , Cohort Studies , Female , Finland , Gene Frequency , Genome-Wide Association Study , Genotype , Haplotypes , Humans , Male , Polymorphism, Single Nucleotide , White People/genetics
13.
Schizophr Bull ; 43(4): 891-899, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28126896

ABSTRACT

Background: Postsynaptic density-95 (PSD-95) protein expression is dysregulated in schizophrenia in a variety of brain regions. We have designed experiments to examine PSD-95 mRNA splice variant expression in the dorsolateral prefrontal cortex from subjects with schizophrenia. Methods: We performed quantitative PCR and western blot analysis to measure PSD-95 expression in schizophrenia vs control subjects, rodent haloperidol treatment studies, rodent postmortem interval studies, and GluN1 knockdown (KD) mice vs controls. Results: We found decreased mRNA expression of beta (t = 4.506, df = 383, P < .0001) and truncated (t = 3.378, df = 383, P = .0008) isoforms of PSD-95, whereas alpha was unchanged. Additionally, we found decreased PSD-95 protein expression in schizophrenia (t = 2.746, df = 71, P = .0076). We found no correlation between PSD-95 protein and alpha, beta, or truncated mRNA isoforms in schizophrenia. PSD-95 beta transcript was increased (t = 3.346, df = 14, P < .05) in the GluN1 KD mouse model of schizophrenia. There was an increase in PSD-95 alpha mRNA expression (t = 2.905, df = 16, P < .05) in rats following long-term haloperidol administration. Conclusions: Our findings describe a unique pathophysiology of specific PSD-95 isoform dysregulation in schizophrenia, chronic neuroleptic treatment, and a genetic lesion mouse model of drastically reduced N-methyl-d-aspartate receptor (NMDAR) complex expression. These data indicate that regulation of PSD-95 is multifaceted, may be isoform specific, and biologically relevant for synaptic signaling function. Specifically, NMDAR-mediated synaptic remodeling, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor trafficking and interaction may be impaired in schizophrenia by decreased PSD-95 beta and truncated expression (respectively). Further, increased PSD-95 beta transcript in the GluN1 KD mouse model poses a potential compensatory rescue of NMDAR-mediated function via increased postsynaptic throughput of the severely reduced GluN1 signal. Together, these data propose that disruption of excitatory signaling complexes through genetic (GluN1 KD), pharmacologic (antipsychotics), or disease (schizophrenia) mechanisms specifically dysregulates PSD-95 expression.


Subject(s)
Antipsychotic Agents/pharmacology , Disks Large Homolog 4 Protein/metabolism , Haloperidol/pharmacology , Prefrontal Cortex/metabolism , Protein Isoforms/metabolism , RNA Splice Sites , Schizophrenia/metabolism , Adult , Aged , Animals , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Nerve Tissue Proteins , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate
14.
ACS Chem Neurosci ; 8(3): 662-668, 2017 03 15.
Article in English | MEDLINE | ID: mdl-27959513

ABSTRACT

Postmortem brain studies support dysregulated expression of the histone deacetylase enzymes, HDAC1 and HDAC2, as a central feature in diseases including schizophrenia, bipolar disorder, and depression. Our objective was to investigate HDAC expression in a large postmortem sample set representing healthy and disease brains. We used >700 well-characterized samples from patients diagnosed with schizophrenia (n = 175), major depressive disorder (n = 135), and bipolar disorder (n = 61) to measure HDAC1 and HDAC2 transcript levels by quantitative real-time PCR in dorsolateral prefrontal cortex (DLPFC) and caudate compared to control samples. HDAC expression was calculated relative to the geometric mean of ß-2-microglobulin, ß-glucuronidase, and ß-actin. In adult-age DLPFC, HDAC2 was decreased by 34% in schizophrenia samples compared to controls (p < 10-4). HDAC2 was significantly upregulated in major depressive disorder samples by 17% versus controls (p = 0.002). Neither smoking history nor therapeutic drugs impacted HDAC2 levels and no HDAC1 patient-control differences were observed. In caudate, HDAC levels were unchanged between patient and control groups. In control DLPFC, age fetal week 14 to 97 years (n = 326), both HDAC1 and HDAC2 levels sharply declined around birth and stabilized thereafter. Using by far the largest postmortem sample set on this topic, our major finding (decreased HDAC2 transcript) showed notable specificity in disease (schizophrenia but not major depressive disorder), HDAC subtype (HDAC2 but not HDAC1) and brain region (DLPFC but not caudate). These differences shape understanding of regional components of neural circuitry in the diseased brain and set a benchmark to quantify HDAC density and distribution using in vivo neuroimaging tools.


Subject(s)
Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Prefrontal Cortex/metabolism , Schizophrenia/pathology , Up-Regulation/physiology , Adolescent , Adult , Aged , Aged, 80 and over , Analysis of Variance , Bipolar Disorder/metabolism , Bipolar Disorder/pathology , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/pathology , Diagnosis , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Humans , Middle Aged , Psychotic Disorders/metabolism , Psychotic Disorders/pathology , RNA, Messenger/metabolism , Schizophrenia/metabolism , Young Adult
15.
Biol Psychiatry ; 81(2): 162-170, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27113501

ABSTRACT

BACKGROUND: The nervous system may include more than 100 residue-specific posttranslational modifications of histones forming the nucleosome core that are often regulated in cell-type-specific manner. On a genome-wide scale, some of the histone posttranslational modification landscapes show significant overlap with the genetic risk architecture for several psychiatric disorders, fueling PsychENCODE and other large-scale efforts to comprehensively map neuronal and nonneuronal epigenomes in hundreds of specimens. However, practical guidelines for efficient generation of histone chromatin immunoprecipitation followed by deep sequencing (ChIP-seq) datasets from postmortem brains are needed. METHODS: Protocols and quality controls are given for the following: 1) extraction, purification, and NeuN neuronal marker immunotagging of nuclei from adult human cerebral cortex; 2) fluorescence-activated nuclei sorting; 3) preparation of chromatin by micrococcal nuclease digest; 4) ChIP for open chromatin-associated histone methylation and acetylation; and 5) generation and sequencing of ChIP-seq libraries. RESULTS: We present a ChIP-seq pipeline for epigenome mapping in the neuronal and nonneuronal nuclei from the postmortem brain. This includes a stepwise system of quality controls and user-friendly data presentation platforms. CONCLUSIONS: Our practical guidelines will be useful for projects aimed at histone posttranslational modification mapping in chromatin extracted from hundreds of postmortem brain samples in cell-type-specific manner.


Subject(s)
Cerebral Cortex/metabolism , Epigenesis, Genetic , Epigenomics/methods , High-Throughput Nucleotide Sequencing/methods , Histones/metabolism , Nucleosomes/metabolism , Acetylation , Antigens, Nuclear/metabolism , Chromatin Immunoprecipitation , Humans , Methylation , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Protein Processing, Post-Translational
16.
Nat Neurosci ; 19(11): 1442-1453, 2016 11.
Article in English | MEDLINE | ID: mdl-27668389

ABSTRACT

Over 100 genetic loci harbor schizophrenia-associated variants, yet how these variants confer liability is uncertain. The CommonMind Consortium sequenced RNA from dorsolateral prefrontal cortex of people with schizophrenia (N = 258) and control subjects (N = 279), creating a resource of gene expression and its genetic regulation. Using this resource, ∼20% of schizophrenia loci have variants that could contribute to altered gene expression and liability. In five loci, only a single gene was involved: FURIN, TSNARE1, CNTN4, CLCN3 or SNAP91. Altering expression of FURIN, TSNARE1 or CNTN4 changed neurodevelopment in zebrafish; knockdown of FURIN in human neural progenitor cells yielded abnormal migration. Of 693 genes showing significant case-versus-control differential expression, their fold changes were ≤ 1.33, and an independent cohort yielded similar results. Gene co-expression implicates a network relevant for schizophrenia. Our findings show that schizophrenia is polygenic and highlight the utility of this resource for mechanistic interpretations of genetic liability for brain diseases.


Subject(s)
Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Multifactorial Inheritance/genetics , Schizophrenia/genetics , Brain/metabolism , Female , Genome-Wide Association Study , Humans , Male , Polymorphism, Single Nucleotide , Risk
18.
PLoS One ; 11(2): e0148558, 2016.
Article in English | MEDLINE | ID: mdl-26848839

ABSTRACT

Genetic variation and early adverse environmental events work together to increase risk for schizophrenia. γ-aminobutyric acid (GABA), the major inhibitory neurotransmitter in adult mammalian brain, plays a major role in normal brain development, and has been strongly implicated in the pathobiology of schizophrenia. GABA synthesis is controlled by two glutamic acid decarboxylase (GAD) genes, GAD1 and GAD2, both of which produce a number of alternative transcripts. Genetic variants in the GAD1 gene are associated with increased risk for schizophrenia, and reduced expression of its major transcript in the human dorsolateral prefrontal cortex (DLPFC). No consistent changes in GAD2 expression have been found in brains from patients with schizophrenia. In this work, with the use of RNA sequencing and PCR technologies, we confirmed and tracked the expression of an alternative truncated transcript of GAD2 (ENST00000428517) in human control DLPFC homogenates across lifespan besides the well-known full length transcript of GAD2. In addition, using quantitative RT-PCR, expression of GAD2 full length and truncated transcripts were measured in the DLPFC of patients with schizophrenia, bipolar disorder and major depression. The expression of GAD2 full length transcript is decreased in the DLPFC of schizophrenia and bipolar disorder patients, while GAD2 truncated transcript is increased in bipolar disorder patients but decreased in schizophrenia patients. Moreover, the patients with schizophrenia with completed suicide or positive nicotine exposure showed significantly higher expression of GAD2 full length transcript. Alternative transcripts of GAD2 may be important in the growth and development of GABA-synthesizing neurons as well as abnormal GABA signaling in the DLPFC of patients with schizophrenia and affective disorders.


Subject(s)
Alternative Splicing , Glutamate Decarboxylase/metabolism , Mood Disorders/genetics , Prefrontal Cortex/metabolism , Schizophrenia/genetics , Adolescent , Adult , Child , Child, Preschool , Female , Gene-Environment Interaction , Genetic Predisposition to Disease , Glutamate Decarboxylase/chemistry , Humans , Infant , Infant, Newborn , Male , Mood Disorders/metabolism , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Regression Analysis , Schizophrenia/metabolism
19.
Cell Rep ; 13(6): 1073-1080, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26526993

ABSTRACT

Brain-derived neurotrophic factor (BDNF) plays a key role in energy balance. In population studies, SNPs of the BDNF locus have been linked to obesity, but the mechanism by which these variants cause weight gain is unknown. Here, we examined human hypothalamic BDNF expression in association with 44 BDNF SNPs. We observed that the minor C allele of rs12291063 is associated with lower human ventromedial hypothalamic BDNF expression (p < 0.001) and greater adiposity in both adult and pediatric cohorts (p values < 0.05). We further demonstrated that the major T allele for rs12291063 possesses a binding capacity for the transcriptional regulator, heterogeneous nuclear ribonucleoprotein D0B, knockdown of which disrupts transactivation by the T allele. Binding and transactivation functions are both disrupted by substituting C for T. These findings provide a rationale for BDNF augmentation as a targeted treatment for obesity in individuals who have the rs12291063 CC genotype.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Obesity/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Brain-Derived Neurotrophic Factor/metabolism , Case-Control Studies , Child , Female , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein D0 , Heterogeneous-Nuclear Ribonucleoprotein D/metabolism , Humans , Hypothalamus/metabolism , Introns , Male , Middle Aged , Protein Binding
20.
Am J Psychiatry ; 172(11): 1122-30, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26206074

ABSTRACT

OBJECTIVE: CHRNA7, coding α-7 nicotinic acetylcholine receptor (α7 nAChR), is involved in cognition through interneuron modulation of dopamine and glutamate signaling. CHRNA7 and its partially duplicated chimeric gene CHRFAM7A have been implicated in schizophrenia through linkage and association studies. METHOD: Expression of CHRNA7 and CHRFAM7A mRNA was measured in the postmortem prefrontal cortex in more than 700 subjects, including patients with schizophrenia, bipolar disorder, major depression, and normal comparison subjects. The effects of antipsychotics and nicotine, as well as associations of CHRNA7 SNPs with gene expression, were explored. Fluorescent in-situ hybridization was used to examine coexpression of both transcripts in the human cortex. RESULTS: CHRFAM7A expression and CHRFAM7A/CHRNA7 ratios were higher in fetal compared with postnatal life, whereas CHRNA7 expression was relatively stable. CHRFAM7A expression was significantly elevated in all diagnostic groups, while CHRNA7 expression was reduced in the schizophrenia group and increased in the major depression group compared with the comparison group. CHRFAM7A/CHRNA7 ratios were significantly increased in the schizophrenia and bipolar disorder groups compared with the comparison group. There was no effect of nicotine or antipsychotics and no association of SNPs in CHRNA7 with expression. CHRNA7 and CHRFAM7A mRNAs were expressed in the same neuronal nuclei of the human neocortex. CONCLUSIONS: These data show preferential fetal CHRFAM7A expression in the human prefrontal cortex and suggest abnormalities in the CHRFAM7A/CHRNA7 ratios in schizophrenia and bipolar disorder, due mainly to overexpression of CHRFAM7A. Given that these transcripts are coexpressed in a subset of human cortical neurons and can interact to alter function of nAChRs, these results support the concept of aberrant function of nAChRs in mental illness.


Subject(s)
Bipolar Disorder/genetics , Depressive Disorder, Major/genetics , Fetus/metabolism , Prefrontal Cortex/metabolism , RNA, Messenger/metabolism , Schizophrenia/genetics , alpha7 Nicotinic Acetylcholine Receptor/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Bipolar Disorder/metabolism , Case-Control Studies , Depressive Disorder, Major/metabolism , Female , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Schizophrenia/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...