Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
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
3.
Nature ; 478(7370): 519-23, 2011 Oct 26.
Article in English | MEDLINE | ID: mdl-22031444

ABSTRACT

Previous investigations have combined transcriptional and genetic analyses in human cell lines, but few have applied these techniques to human neural tissue. To gain a global molecular perspective on the role of the human genome in cortical development, function and ageing, we explore the temporal dynamics and genetic control of transcription in human prefrontal cortex in an extensive series of post-mortem brains from fetal development through ageing. We discover a wave of gene expression changes occurring during fetal development which are reversed in early postnatal life. One half-century later in life, this pattern of reversals is mirrored in ageing and in neurodegeneration. Although we identify thousands of robust associations of individual genetic polymorphisms with gene expression, we also demonstrate that there is no association between the total extent of genetic differences between subjects and the global similarity of their transcriptional profiles. Hence, the human genome produces a consistent molecular architecture in the prefrontal cortex, despite millions of genetic differences across individuals and races. To enable further discovery, this entire data set is freely available (from Gene Expression Omnibus: accession GSE30272; and dbGaP: accession phs000417.v1.p1) and can also be interrogated via a biologist-friendly stand-alone application (http://www.libd.org/braincloud).


Subject(s)
Aging/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Transcriptome/genetics , Autopsy , Fetus/metabolism , Genome, Human/genetics , Humans , Polymorphism, Single Nucleotide/genetics , Prefrontal Cortex/embryology , Racial Groups/genetics , Time Factors
4.
J Neurosci ; 34(14): 4929-40, 2014 Apr 02.
Article in English | MEDLINE | ID: mdl-24695712

ABSTRACT

Early in development, GABA, an inhibitory neurotransmitter in adults, is excitatory. NKCC1 (SLC12A2) encodes one of two cation chloride cotransporters mediating the conversion of GABA from excitatory to inhibitory. Using 3' and 5' RACE and PCR, we verified previously characterized alternative transcripts of NKCC1a (1-27) and NKCC1b (1-27(Δ21)), identified new NKCC1 transcripts, and explored their expression patterns during human prefrontal cortical development. A novel ultra-short transcript (1-2a) was expressed preferentially in the fetus. Expression of NKCC1b and 1-2a were decreased in schizophrenia compared with controls (NKCC1b: 0.8-fold decrease, p = 0.013; 1-2a: 0.8-fold decrease, p = 0.006). Furthermore, the expression of NKCC1b was associated with NKCC1 polymorphism rs3087889. The minor allele at rs3087889, associated with reduced NKCC1b expression (homozygous for major allele: N = 37; homozygous for minor allele: N = 15; 1.5-fold decrease; p < 0.01), was also associated with a modest increase in schizophrenia risk in a case-control sample (controls: N = 435; cases: N = 397, OR = 1.5). This same allele was then found associated with cognitive (n = 369) and fMRI (n = 313) intermediate phenotypes associated with schizophrenia-working memory (Cohen's d = 0.35), global cognition or g (d = 0.18), and prefrontal inefficiency (d = 0.36) as measured by BOLD fMRI during a working memory task. Together, these preclinical and clinical results suggest that variation in NKCC1 may increase risk for schizophrenia via alterations of mRNA expression at the molecular level and impairment of optimal prefrontal function at the macro or systems level.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Prefrontal Cortex/metabolism , Schizophrenia/pathology , Solute Carrier Family 12, Member 2/metabolism , Adolescent , Adult , Aged , Child , Child, Preschool , Cognition Disorders/diagnosis , Cognition Disorders/etiology , Cohort Studies , DNA, Recombinant , Female , Fetus , Genotype , HEK293 Cells , Humans , Infant , Infant, Newborn , Male , Middle Aged , Mutation/genetics , Oxygen/blood , Postmortem Changes , Prefrontal Cortex/blood supply , Prefrontal Cortex/embryology , Prefrontal Cortex/growth & development , Psychiatric Status Rating Scales , Solute Carrier Family 12, Member 2/genetics , Young Adult
5.
Am J Hum Genet ; 90(2): 260-72, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22305529

ABSTRACT

The human prefrontal cortex (PFC), a mastermind of the brain, is one of the last brain regions to mature. To investigate the role of epigenetics in the development of PFC, we examined DNA methylation in ∼14,500 genes at ∼27,000 CpG loci focused on 5' promoter regions in 108 subjects range in age from fetal to elderly. DNA methylation in the PFC shows unique temporal patterns across life. The fastest changes occur during the prenatal period, slow down markedly after birth and continue to slow further with aging. At the genome level, the transition from fetal to postnatal life is typified by a reversal of direction, from demethylation prenatally to increased methylation postnatally. DNA methylation is strongly associated with genotypic variants and correlates with expression of a subset of genes, including genes involved in brain development and in de novo DNA methylation. Our results indicate that promoter DNA methylation in the human PFC is a highly dynamic process modified by genetic variance and regulating gene transcription. Additional discovery is made possible with a stand-alone application, BrainCloudMethyl.


Subject(s)
Aging/genetics , DNA Methylation , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Child , Child, Preschool , CpG Islands/genetics , Epigenesis, Genetic , Female , Fetus/metabolism , Genetic Variation , Humans , Infant , Infant, Newborn , Male , Middle Aged , Prefrontal Cortex/embryology , Promoter Regions, Genetic , Quantitative Trait Loci , Sex Factors , Young Adult
6.
Proc Natl Acad Sci U S A ; 109(30): 12165-70, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22689948

ABSTRACT

Neuregulin 1 (NRG1) and ErbB4, critical neurodevelopmental genes, are implicated in schizophrenia, but the mediating mechanisms are unknown. Here we identify a genetically regulated, pharmacologically targetable, risk pathway associated with schizophrenia and with ErbB4 genetic variation involving increased expression of a PI3K-linked ErbB4 receptor (CYT-1) and the phosphoinositide 3-kinase subunit, p110δ (PIK3CD). In human lymphoblasts, NRG1-mediated phosphatidyl-inositol,3,4,5 triphosphate [PI(3,4,5)P3] signaling is predicted by schizophrenia-associated ErbB4 genotype and PIK3CD levels and is impaired in patients with schizophrenia. In human brain, the same ErbB4 genotype again predicts increased PIK3CD expression. Pharmacological inhibition of p110δ using the small molecule inhibitor, IC87114, blocks the effects of amphetamine in a mouse pharmacological model of psychosis and reverses schizophrenia-related phenotypes in a rat neonatal ventral hippocampal lesion model. Consistent with these antipsychotic-like properties, IC87114 increases AKT phosphorylation in brains of treated mice, implicating a mechanism of action. Finally, in two family-based genetic studies, PIK3CD shows evidence of association with schizophrenia. Our data provide insight into a mechanism of ErbB4 association with schizophrenia; reveal a previously unidentified biological and disease link between NRG1-ErbB4, p110δ, and AKT; and suggest that p110δ is a previously undescribed therapeutic target for the treatment of psychiatric disorders.


Subject(s)
Adenine/analogs & derivatives , ErbB Receptors/metabolism , Neuregulin-1/metabolism , Phosphoinositide-3 Kinase Inhibitors , Quinazolines/pharmacology , Schizophrenia/genetics , Schizophrenia/metabolism , Signal Transduction/physiology , Adenine/chemistry , Adenine/pharmacology , Amphetamine/antagonists & inhibitors , Analysis of Variance , Animals , Antipsychotic Agents/pharmacology , B-Lymphocytes , Blotting, Western , Cell Line, Transformed , Class I Phosphatidylinositol 3-Kinases , ErbB Receptors/genetics , Flow Cytometry , Genetic Association Studies , Humans , Mice , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Quinazolines/chemistry , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, ErbB-4 , Reverse Transcriptase Polymerase Chain Reaction , Schizophrenia/drug therapy
7.
Lancet ; 392(10145): 378-379, 2018 08 04.
Article in English | MEDLINE | ID: mdl-30102165
8.
J Neurosci ; 32(15): 5216-22, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22496567

ABSTRACT

The neuron-specific K(+)-Cl(-) cotransporter SLC12A5, also known as KCC2, helps mediate the electrophysiological effects of GABA. The pattern of KCC2 expression during early brain development suggests that its upregulation drives the postsynaptic switch of GABA from excitation to inhibition. We previously found decreased expression of full-length KCC2 in the postmortem hippocampus of patients with schizophrenia, but not in the dorsolateral prefrontal cortex (DLPFC). Using PCR and rapid amplification of cDNA ends, we discovered several previously unrecognized alternative KCC2 transcripts in both human adult and fetal brain in addition to the previously identified full-length (NM_020708.3) and truncated (AK098371) transcripts. We measured the expression levels of four relatively abundant truncated splice variants, including three novel transcripts (ΔEXON6, EXON2B, and EXON6B) and one previously described transcript (AK098371), in a large human cohort of nonpsychiatric controls across the lifespan, and in patients with schizophrenia and affective disorders. In SH-SY5Y cell lines, these transcripts were translated into proteins and expressed at their predicted sizes. Expression of the EXON6B transcript is increased in the DLPFC of patients with schizophrenia (p = 0.03) but decreased in patients with major depression (p = 0.04). The expression of AK098371 is associated with a GAD1 single nucleotide polymorphism (rs3749034) that previously has been associated with GAD67 expression and risk for schizophrenia. Our data confirm the developmental regulation of KCC2 expression, and provide evidence that KCC2 transcripts are differentially expressed in schizophrenia and affective disorders. Alternate transcripts from KCC2 may participate in the abnormal GABA signaling in the DLPFC associated with schizophrenia.


Subject(s)
Mood Disorders/genetics , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Schizophrenia/genetics , Symporters/genetics , Adult , Aged, 80 and over , Bipolar Disorder/genetics , Bipolar Disorder/metabolism , Blotting, Western , Cell Line , DNA, Complementary/biosynthesis , DNA, Complementary/isolation & purification , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Exons/genetics , Female , Gene Amplification , Genetic Variation/genetics , Genetic Variation/physiology , Genotype , Glutamate Decarboxylase/genetics , Humans , Male , Middle Aged , Mood Disorders/metabolism , Polymerase Chain Reaction , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , RNA/biosynthesis , RNA/isolation & purification , Schizophrenia/metabolism , Symporters/metabolism , Transfection , Young Adult , K Cl- Cotransporters
9.
Proc Natl Acad Sci U S A ; 107(35): 15619-24, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20713722

ABSTRACT

Structural and polymorphic variations in Neuregulin 3 (NRG3), 10q22-23 are associated with a broad spectrum of neurodevelopmental disorders including developmental delay, cognitive impairment, autism, and schizophrenia. NRG3 is a member of the neuregulin family of EGF proteins and a ligand for the ErbB4 receptor tyrosine kinase that plays pleotropic roles in neurodevelopment. Several genes in the NRG-ErbB signaling pathway including NRG1 and ErbB4 have been implicated in genetic predisposition to schizophrenia. Previous fine mapping of the 10q22-23 locus in schizophrenia identified genome-wide significant association between delusion severity and polymorphisms in intron 1 of NRG3 (rs10883866, rs10748842, and rs6584400). The biological mechanisms remain unknown. We identified significant association of these SNPs with increased risk for schizophrenia in 350 families with an affected offspring and confirmed association to patient delusion and positive symptom severity. Molecular cloning and cDNA sequencing in human brain revealed that NRG3 undergoes complex splicing, giving rise to multiple structurally distinct isoforms. RNA expression profiling of these isoforms in the prefrontal cortex of 400 individuals revealed that NRG3 expression is developmentally regulated and pathologically increased in schizophrenia. Moreover, we show that rs10748842 lies within a DNA ultraconserved element and homedomain and strongly predicts brain expression of NRG3 isoforms that contain a unique developmentally regulated 5' exon (P = 1.097E(-12) to 1.445E(-15)). Our observations strengthen the evidence that NRG3 is a schizophrenia susceptibility gene, provide quantitative insight into NRG3 transcription traits in the human brain, and reveal a probable mechanistic basis for disease association.


Subject(s)
Brain/metabolism , Neuregulins/genetics , Polymorphism, Single Nucleotide , Schizophrenia/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Blotting, Western , Brain/pathology , Case-Control Studies , Child , Child, Preschool , Family Health , Female , Gene Frequency , Genetic Predisposition to Disease , Genotype , Humans , Infant , Infant, Newborn , Male , Middle Aged , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Risk Factors , Schizophrenia/diagnosis , Schizophrenia/metabolism , Young Adult
10.
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
11.
J Neurosci ; 31(30): 11088-95, 2011 Jul 27.
Article in English | MEDLINE | ID: mdl-21795557

ABSTRACT

GABA signaling molecules are critical for both human brain development and the pathophysiology of schizophrenia. We examined the expression of transcripts derived from three genes related to GABA signaling [GAD1 (GAD67 and GAD25), SLC12A2 (NKCC1), and SLC12A5 (KCC2)] in the prefrontal cortex (PFC) and hippocampal formation of a large cohort of nonpsychiatric control human brains (n = 240) across the lifespan (from fetal week 14 to 80 years) and in patients with schizophrenia (n = 30-31), using quantitative RT-PCR. We also examined whether a schizophrenia risk-associated promoter SNP in GAD1 (rs3749034) is related to expression of these transcripts. Our studies revealed that development and maturation of both the PFC and hippocampal formation are characterized by progressive switches in expression from GAD25 to GAD67 and from NKCC1 to KCC2. Previous studies have demonstrated that the former leads to GABA synthesis, and the latter leads to switching from excitatory to inhibitory neurotransmission. In the hippocampal formation, GAD25/GAD67 and NKCC1/KCC2 ratios are increased in patients with schizophrenia, reflecting a potentially immature GABA physiology. Remarkably, GAD25/GAD67 and NKCC1/KCC2 expression ratios are associated with rs3749034 genotype, with risk alleles again predicting a relatively less mature pattern. These findings suggest that abnormalities in GABA signaling critical to brain development contribute to genetic risk for schizophrenia.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Schizophrenia/pathology , Signal Transduction/physiology , gamma-Aminobutyric Acid/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Antipsychotic Agents/pharmacology , Cerebral Cortex/drug effects , Child , Child, Preschool , Cohort Studies , Female , Fetus , Gene Expression Regulation, Developmental/drug effects , Genotype , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Hippocampus/drug effects , Humans , Infant , Infant, Newborn , Male , Middle Aged , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 2 , Symporters/genetics , Symporters/metabolism , gamma-Aminobutyric Acid/genetics , K Cl- Cotransporters
12.
Proc Natl Acad Sci U S A ; 106(37): 15873-8, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19805229

ABSTRACT

Disrupted-In-Schizophrenia-1 (DISC1) is a promising susceptibility gene for major mental illness, but the mechanism of the clinical association is unknown. We searched for DISC1 transcripts in adult and fetal human brain and tested whether their expression is altered in patients with schizophrenia and is associated with genetic variation in DISC1. Many alternatively spliced transcripts were identified, including groups lacking exon 3 (Delta3), exons 7 and 8 (Delta7Delta8), an exon 3 insertion variant (extra short variant-1, Esv1), and intergenic splicing between TSNAX and DISC1. Isoforms Delta7Delta8, Esv1, and Delta3, which encode truncated DISC1 proteins, were expressed more abundantly during fetal development than during postnatal ages, and their expression was higher in the hippocampus of patients with schizophrenia. Schizophrenia risk-associated polymorphisms [non-synonymous SNPs rs821616 (Cys704Ser) and rs6675281 (Leu607Phe), and rs821597] were associated with the expression of Delta3 and Delta7Delta8. Moreover, the same allele at rs6675281, which predicted higher expression of these transcripts in the hippocampus, was associated with higher expression of DISC1Delta7Delta8 in lymphoblasts in an independent sample. Our results implicate a molecular mechanism of genetic risk associated with DISC1 involving specific alterations in gene processing.


Subject(s)
Alternative Splicing , Nerve Tissue Proteins/genetics , Polymorphism, Single Nucleotide , Schizophrenia/genetics , Adult , Base Sequence , Brain/embryology , Brain/metabolism , Case-Control Studies , DNA Primers/genetics , Exons , Female , Fetal Development/genetics , Fetus/metabolism , Hippocampus/metabolism , Humans , Male , Middle Aged , Nerve Tissue Proteins/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors , Schizophrenia/metabolism , Up-Regulation
13.
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
14.
J Neurosci ; 30(17): 5992-7, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20427658

ABSTRACT

We explored the effect of single-nucleotide polymorphisms (SNPs) in the fibroblast growth factor 20 gene (FGF20) associated with risk for Parkinson's disease on brain structure and function in a large sample of healthy young-adult human subjects and also in elderly subjects to look at the interaction between genetic variations and age (N = 237; 116 men; 18-87 years). We analyzed high-resolution anatomical magnetic resonance images using voxel-based morphometry, a quantitative neuroanatomical technique. We also measured FGF20 mRNA expression in postmortem human brain tissue to determine the molecular correlates of these SNPs (N = 108; 72 men; 18-74 years). We found that the T allele carriers of rs12720208 in the 3'-untranslated region had relatively larger hippocampal volume (p = 0.0059) and diminished verbal episodic memory (p = 0.048) and showed steeper decreases of hippocampal volume with normal aging (p = 0.026). In postmortem brain, T allele carriers had greater expression of hippocampal FGF20 mRNA (p = 0.037), consistent with a previously characterized microRNA mechanism. The C allele matches a predicted miR-433 microRNA binding domain, whereas the T allele disrupts it, resulting in higher FGF20 protein translation. The strong FGF20 genetic effects in hippocampus are presumably mediated by activation of the FGFR1 (FGF receptor 1), which is expressed in mammalian brain most abundantly in the hippocampus. These associations, from mRNA expression to brain morphology to cognition and an interaction with aging, confirm a role of FGF20 in human brain structure and function during development and aging.


Subject(s)
Aging/metabolism , Aging/pathology , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hippocampus/metabolism , Hippocampus/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Cognition/physiology , Female , Genotype , Humans , Male , MicroRNAs/metabolism , Middle Aged , Organ Size , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Young Adult
15.
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
16.
J Clin Invest ; 117(3): 672-82, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17290303

ABSTRACT

Dopamine- and cAMP-regulated phosphoprotein of molecular weight 32 kDa (DARPP-32), encoded by PPP1R1B, is a pivotal integrator of information in dopaminoceptive neurons, regulating the response to neuroleptics, psychotomimetics, and drugs of abuse, and affecting striatal function and plasticity. Despite extensive preclinical work, there are almost no data on DARPP-32 function in humans. Here, we identify, through resequencing in 298 chromosomes, a frequent PPP1R1B haplotype predicting mRNA expression of PPP1R1B isoforms in postmortem human brain. This haplotype was associated with enhanced performance on several cognitive tests that depend on frontostriatal function. Multimodal imaging of healthy subjects revealed an impact of the haplotype on neostriatal volume, activation, and the functional connectivity of the prefrontal cortex. The haplotype was associated with the risk for schizophrenia in 1 family-based association analysis. Our convergent results identify a prefrontal-neostriatal system affected by variation in PPP1R1B and suggest that DARPP-32 plays a pivotal role in cognitive function and possibly in the pathogenesis of schizophrenia.


Subject(s)
Cognition/physiology , Dopamine and cAMP-Regulated Phosphoprotein 32/physiology , Neostriatum/physiology , Prefrontal Cortex/physiology , Schizophrenia/genetics , Chromosomes, Human/genetics , Dopamine and cAMP-Regulated Phosphoprotein 32/genetics , Haplotypes , Humans , Neostriatum/anatomy & histology , Polymorphism, Single Nucleotide , Prefrontal Cortex/anatomy & histology , Risk , Sequence Analysis, DNA
17.
Hum Genet ; 127(4): 441-52, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20084519

ABSTRACT

The etiology of schizophrenia likely involves genetic interactions. DISC1, a promising candidate susceptibility gene, encodes a protein which interacts with many other proteins, including CIT, NDEL1, NDE1, FEZ1 and PAFAH1B1, some of which also have been associated with psychosis. We tested for epistasis between these genes in a schizophrenia case-control study using machine learning algorithms (MLAs: random forest, generalized boosted regression andMonteCarlo logic regression). Convergence of MLAs revealed a subset of seven SNPs that were subjected to 2-SNP interaction modeling using likelihood ratio tests for nested unconditional logistic regression models. Of the 7C2 = 21 interactions, four were significant at the α = 0.05 level: DISC1 rs1411771-CIT rs10744743 OR = 3.07 (1.37, 6.98) p = 0.007; CIT rs3847960-CIT rs203332 OR = 2.90 (1.45, 5.79) p = 0.003; CIT rs3847960-CIT rs440299 OR = 2.16 (1.04, 4.46) p = 0.038; one survived Bonferroni correction (NDEL1 rs4791707-CIT rs10744743 OR = 4.44 (2.22, 8.88) p = 0.00013). Three of four interactions were validated via functional magnetic resonance imaging (fMRI) in an independent sample of healthy controls; risk associated alleles at both SNPs predicted prefrontal cortical inefficiency during the N-back task, a schizophrenia-linked intermediate biological phenotype: rs3847960-rs440299; rs1411771-rs10744743, rs4791707-rs10744743 (SPM5 p < 0.05, corrected), although we were unable to statistically replicate the interactions in other clinical samples. Interestingly, the CIT SNPs are proximal to exons that encode theDISC1 interaction domain. In addition, the 3' UTR DISC1 rs1411771 is predicted to be an exonic splicing enhancer and the NDEL1 SNP is ~3,000 bp from the exon encoding the region of NDEL1 that interacts with the DISC1 protein, giving a plausible biological basis for epistasis signals validated by fMRI.


Subject(s)
Carrier Proteins/genetics , Epistasis, Genetic , Intracellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Schizophrenia/genetics , Adolescent , Adult , Aged , Algorithms , Alleles , Artificial Intelligence , Case-Control Studies , Female , Humans , Likelihood Functions , Logistic Models , Magnetic Resonance Imaging , Male , Middle Aged , Models, Genetic , Monte Carlo Method , Polymorphism, Single Nucleotide , Risk Factors , Schizophrenia/pathology , Young Adult
18.
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
19.
J Neurosci ; 28(35): 8709-23, 2008 Aug 27.
Article in English | MEDLINE | ID: mdl-18753372

ABSTRACT

The COMT (catechol-O-methyltransferase) gene has been linked to a spectrum of human phenotypes, including cognition, anxiety, pain sensitivity and psychosis. Doubts about its clinical impact exist, however, because of the complexity of human COMT polymorphism and clinical variability. We generated transgenic mice overexpressing a human COMT-Val polymorphism (Val-tg), and compared them with mice containing a null COMT mutation. Increased COMT enzyme activity in Val-tg mice resulted in disrupted attentional set-shifting abilities, and impaired working and recognition memory, but blunted stress responses and pain sensitivity. Conversely, COMT disruption improved working memory, but increased stress responses and pain sensitivity. Amphetamine ameliorated recognition memory deficits in COMT-Val-tg mice but disrupted it in wild types, illustrating COMT modulation of the inverted-U relationship between cognition and dopamine. COMT-Val-tg mice showed increased prefrontal cortex (PFC) calcium/calmodulin-dependent protein kinase II (CaMKII) levels, whereas COMT deficiency decreased PFC CaMKII but increased PFC CaMKKbeta and CaMKIV levels, suggesting the involvement of PFC CaMK pathways in COMT-regulated cognitive function and adaptive stress responses. Our data indicate a critical role for the COMT gene in an apparent evolutionary trade-off between cognitive and affective functions.


Subject(s)
Catechol O-Methyltransferase/genetics , Cognition/physiology , Exploratory Behavior/physiology , Mutation/genetics , Stress, Psychological/genetics , Acoustic Stimulation/methods , Amphetamine/pharmacology , Analysis of Variance , Animals , Attention/physiology , Behavior, Animal , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Dopamine/metabolism , Dopamine Agents/pharmacology , Humans , Hyperthermia, Induced/methods , Inhibition, Psychological , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Pain Measurement/methods , Pain Threshold/physiology , Reaction Time/genetics , Reflex, Startle/genetics , Stress, Psychological/enzymology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL