23 results on '"Akbarian S"'
Search Results
2. Genetic vulnerability to DUSP22 promoter hypermethylation is involved in the relation between in utero famine exposure and schizophrenia
- Author
-
Onderzoeksgroep 2, Brain, Onderzoeksgroep 11, Affectieve & Psychotische Med., Onderzoeksgroep 1, Pathologie, TN groep Hol, Onderzoeksgroep 5, Psychiatrie_Medisch, Boks, M P, Houtepen, L C, Xu, Z, He, Y, Ursini, G, Maihofer, A X, Rajarajan, P, Yu, Q, Xu, H, Wu, Y, Wang, S, Shi, J P, Hulshoff Pol, H E, Strengman, E, Rutten, B P F, Jaffe, A E, Kleinman, J E, Baker, D G, Hol, E M, Akbarian, S, Nievergelt, C M, De Witte, L D, Vinkers, C H, Weinberger, D R, Yu, J, Kahn, R S, Onderzoeksgroep 2, Brain, Onderzoeksgroep 11, Affectieve & Psychotische Med., Onderzoeksgroep 1, Pathologie, TN groep Hol, Onderzoeksgroep 5, Psychiatrie_Medisch, Boks, M P, Houtepen, L C, Xu, Z, He, Y, Ursini, G, Maihofer, A X, Rajarajan, P, Yu, Q, Xu, H, Wu, Y, Wang, S, Shi, J P, Hulshoff Pol, H E, Strengman, E, Rutten, B P F, Jaffe, A E, Kleinman, J E, Baker, D G, Hol, E M, Akbarian, S, Nievergelt, C M, De Witte, L D, Vinkers, C H, Weinberger, D R, Yu, J, and Kahn, R S
- Published
- 2018
3. Author Correction: Transitioning sleeping position detection in late pregnancy using computer vision from controlled to real-world settings: an observational study.
- Author
-
Kember AJ, Zia H, Elangainesan P, Hsieh ME, Adijeh R, Li I, Ritchie L, Akbarian S, Taati B, Hobson SR, and Dolatabadi E
- Published
- 2024
- Full Text
- View/download PDF
4. Transitioning sleeping position detection in late pregnancy using computer vision from controlled to real-world settings: an observational study.
- Author
-
Kember AJ, Zia H, Elangainesan P, Hsieh ME, Adijeh R, Li I, Ritchie L, Akbarian S, Taati B, Hobson SR, and Dolatabadi E
- Subjects
- Humans, Female, Pregnancy, Adult, Prospective Studies, Posture physiology, Video Recording, Neural Networks, Computer, Sleep physiology
- Abstract
Sleeping on the back after 28 weeks of pregnancy has recently been associated with giving birth to a small-for-gestational-age infant and late stillbirth, but whether a causal relationship exists is currently unknown and difficult to study prospectively. This study was conducted to build a computer vision model that can automatically detect sleeping position in pregnancy under real-world conditions. Real-world overnight video recordings were collected from an ongoing, Canada-wide, prospective, four-night, home sleep apnea study and controlled-setting video recordings were used from a previous study. Images were extracted from the videos and body positions were annotated. Five-fold cross validation was used to train, validate, and test a model using state-of-the-art deep convolutional neural networks. The dataset contained 39 pregnant participants, 13 bed partners, 12,930 images, and 47,001 annotations. The model was trained to detect pillows, twelve sleeping positions, and a sitting position in both the pregnant person and their bed partner simultaneously. The model significantly outperformed a previous similar model for the three most commonly occurring natural sleeping positions in pregnant and non-pregnant adults, with an 82-to-89% average probability of correctly detecting them and a 15-to-19% chance of failing to detect them when any one of them is present., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. tRNA epitranscriptomic alterations associated with opioid-induced reward-seeking and long-term opioid withdrawal in male mice.
- Author
-
Blaze J, Browne CJ, Futamura R, Javidfar B, Zachariou V, Nestler EJ, and Akbarian S
- Subjects
- Animals, Male, Mice, Epigenesis, Genetic drug effects, Analgesics, Opioid pharmacology, Analgesics, Opioid administration & dosage, Mice, Knockout, Drug-Seeking Behavior drug effects, Drug-Seeking Behavior physiology, Reward, Morphine pharmacology, Morphine administration & dosage, Morphine adverse effects, Mice, Inbred C57BL, Substance Withdrawal Syndrome genetics, Prefrontal Cortex metabolism, Prefrontal Cortex drug effects, RNA, Transfer genetics
- Abstract
DNA cytosine methylation has been documented as a potential epigenetic mechanism of transcriptional regulation underlying opioid use disorder. However, methylation of RNA cytosine residues, which would drive another level of biological influence as an epitranscriptomic mechanism of gene and protein regulation has not been studied in the context of addiction. Here, we probed whether chronic morphine exposure could alter tRNA cytosine methylation (m
5 C) and resulting expression levels in the medial prefrontal cortex (mPFC), a brain region crucial for reward processing and executive function that exhibits opioid-induced molecular restructuring. We identified dynamic changes in glycine tRNA (tRNAGly GCC ) cytosine methylation, corresponding to altered expression levels of this tRNA at multiple timepoints following 15 days of daily morphine. Additionally, a robust increase in methylation, coupled with decreased expression, was present after 30 days of withdrawal, suggesting that repeated opioid administration produces changes to the tRNA regulome long after discontinuation. Furthermore, forebrain-wide knockout of neuronal Nsun2, a tRNA methyltransferase, was associated with disruption of opioid conditioned place preference, and this effect was recapitulated by regional mPFC Nsun2 knockout. Taken together, these studies provide a foundational link between the regulation of tRNA cytosine methylation and opioid reward and highlight the tRNA machinery as a potential therapeutic target in addiction., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
6. Genomic data resources of the Brain Somatic Mosaicism Network for neuropsychiatric diseases.
- Author
-
Garrison MA, Jang Y, Bae T, Cherskov A, Emery SB, Fasching L, Jones A, Moldovan JB, Molitor C, Pochareddy S, Peters MA, Shin JH, Wang Y, Yang X, Akbarian S, Chess A, Gage FH, Gleeson JG, Kidd JM, McConnell M, Mills RE, Moran JV, Park PJ, Sestan N, Urban AE, Vaccarino FM, Walsh CA, Weinberger DR, Wheelan SJ, and Abyzov A
- Subjects
- Humans, Autism Spectrum Disorder genetics, Brain, Genomics, Mosaicism, Genome, Human, Mental Disorders genetics
- Abstract
Somatic mosaicism is defined as an occurrence of two or more populations of cells having genomic sequences differing at given loci in an individual who is derived from a single zygote. It is a characteristic of multicellular organisms that plays a crucial role in normal development and disease. To study the nature and extent of somatic mosaicism in autism spectrum disorder, bipolar disorder, focal cortical dysplasia, schizophrenia, and Tourette syndrome, a multi-institutional consortium called the Brain Somatic Mosaicism Network (BSMN) was formed through the National Institute of Mental Health (NIMH). In addition to genomic data of affected and neurotypical brains, the BSMN also developed and validated a best practices somatic single nucleotide variant calling workflow through the analysis of reference brain tissue. These resources, which include >400 terabytes of data from 1087 subjects, are now available to the research community via the NIMH Data Archive (NDA) and are described here., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
7. Unsupervised recognition of components from the interaction of BSA with Fe cluster in different conditions utilizing 2D fluorescence spectroscopy.
- Author
-
Kompany-Zareh M, Akbarian S, and Najafpour MM
- Subjects
- Phenylalanine, Spectrometry, Fluorescence methods, Tyrosine, Serum Albumin, Bovine chemistry, Tryptophan
- Abstract
The excitation-emission fluorescence spectroscopy combined with three-way analysis was applied for discriminating the pure BSA and BSA/Fe
3 O(OAc)6 ClO4 (Fe) using unsupervised classification methods. Herein, the interaction of bovine serum albumin (BSA) and Fe clusters as an artificial enzyme is studied by extracting the intrinsic excitation-emission (EEM) fluorescence of BSA. The conformation of BSA changes with pH, temperature, and Fe concentration. Three-way fluorescence data were recorded for BSA and BSA/Fe during different days. The obtained results showed that the Fe clusters cause changes in the structure of BSA conformation as a function of pH, temperature, and Fe concentration. Also, the denaturation pathway of the BSA molecule is significantly different in the presence of Fe clusters. Both techniques of PARAFAC and PCA were used in the excitation-emission fluorescence matrices (EEM) of solutions at three different pH (5.0, 7.0, and 9.0) and temperatures (15.0, 25.0, and 35.0 °C) values. Also, we reported the results of the change in concentrations of Fe (4.0, 6.0, and 8.0 mg) using these methods. These three amino acids (tyrosine, tryptophan, and phenylalanine) indicate all datasets and their similarities and differences. The spectral differences were more remarkable in different pH values compared to different temperatures. Also, we could distinguish between the groups of protein samples properly in different concentrations of Fe using low-cost EEM spectral images and PARAFAC., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
8. Chromatin domain alterations linked to 3D genome organization in a large cohort of schizophrenia and bipolar disorder brains.
- Author
-
Girdhar K, Hoffman GE, Bendl J, Rahman S, Dong P, Liao W, Hauberg ME, Sloofman L, Brown L, Devillers O, Kassim BS, Wiseman JR, Park R, Zharovsky E, Jacobov R, Flatow E, Kozlenkov A, Gilgenast T, Johnson JS, Couto L, Peters MA, Phillips-Cremins JE, Hahn CG, Gur RE, Tamminga CA, Lewis DA, Haroutunian V, Dracheva S, Lipska BK, Marenco S, Kundakovic M, Fullard JF, Jiang Y, Roussos P, and Akbarian S
- Subjects
- Adult, Brain, Chromatin, Humans, Lysine genetics, Bipolar Disorder genetics, Schizophrenia genetics
- 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, 10
4 -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., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
9. A prefrontal-paraventricular thalamus circuit requires juvenile social experience to regulate adult sociability in mice.
- Author
-
Yamamuro K, Bicks LK, Leventhal MB, Kato D, Im S, Flanigan ME, Garkun Y, Norman KJ, Caro K, Sadahiro M, Kullander K, Akbarian S, Russo SJ, and Morishita H
- Subjects
- Animals, Behavior, Animal, Interneurons physiology, Male, Neural Pathways physiology, Optogenetics, Social Isolation, Midline Thalamic Nuclei physiology, Neurons physiology, Prefrontal Cortex physiology, Social Behavior
- Abstract
Juvenile social isolation reduces sociability in adulthood, but the underlying neural circuit mechanisms are poorly understood. We found that, in male mice, 2 weeks of social isolation immediately following weaning leads to a failure to activate medial prefrontal cortex neurons projecting to the posterior paraventricular thalamus (mPFC→pPVT) during social exposure in adulthood. Chemogenetic or optogenetic suppression of mPFC→pPVT activity in adulthood was sufficient to induce sociability deficits without affecting anxiety-related behaviors or preference toward rewarding food. Juvenile isolation led to both reduced excitability of mPFC→pPVT neurons and increased inhibitory input drive from low-threshold-spiking somatostatin interneurons in adulthood, suggesting a circuit mechanism underlying sociability deficits. Chemogenetic or optogenetic stimulation of mPFC→pPVT neurons in adulthood could rescue the sociability deficits caused by juvenile isolation. Our study identifies a pair of specific medial prefrontal cortex excitatory and inhibitory neuron populations required for sociability that are profoundly affected by juvenile social experience.
- Published
- 2020
- Full Text
- View/download PDF
10. A computational tool (H-MAGMA) for improved prediction of brain-disorder risk genes by incorporating brain chromatin interaction profiles.
- Author
-
Sey NYA, Hu B, Mah W, Fauni H, McAfee JC, Rajarajan P, Brennand KJ, Akbarian S, and Won H
- Subjects
- Brain Diseases metabolism, Genomics, Humans, Risk Factors, Brain metabolism, Brain Diseases genetics, Chromatin metabolism, Genetic Predisposition to Disease, Polymorphism, Single Nucleotide
- Abstract
Most risk variants for brain disorders identified by genome-wide association studies reside in the noncoding genome, which makes deciphering biological mechanisms difficult. A commonly used tool, multimarker analysis of genomic annotation (MAGMA), addresses this issue by aggregating single nucleotide polymorphism associations to nearest genes. Here we developed a platform, Hi-C-coupled MAGMA (H-MAGMA), that advances MAGMA by incorporating chromatin interaction profiles from human brain tissue across two developmental epochs and two brain cell types. By analyzing gene regulatory relationships in the disease-relevant tissue, H-MAGMA identified neurobiologically relevant target genes. We applied H-MAGMA to five psychiatric disorders and four neurodegenerative disorders to interrogate biological pathways, developmental windows and cell types implicated for each disorder. Psychiatric-disorder risk genes tended to be expressed during mid-gestation and in excitatory neurons, whereas neurodegenerative-disorder risk genes showed increasing expression over time and more diverse cell-type specificities. H-MAGMA adds to existing analytic frameworks to help identify the neurobiological principles of brain disorders.
- Published
- 2020
- Full Text
- View/download PDF
11. CommonMind Consortium provides transcriptomic and epigenomic data for Schizophrenia and Bipolar Disorder.
- Author
-
Hoffman GE, Bendl J, Voloudakis G, Montgomery KS, Sloofman L, Wang YC, Shah HR, Hauberg ME, Johnson JS, Girdhar K, Song L, Fullard JF, Kramer R, Hahn CG, Gur R, Marenco S, Lipska BK, Lewis DA, Haroutunian V, Hemby S, Sullivan P, Akbarian S, Chess A, Buxbaum JD, Crawford GE, Domenici E, Devlin B, Sieberts SK, Peters MA, and Roussos P
- Subjects
- Cohort Studies, Epigenomics, Humans, Prefrontal Cortex metabolism, Prefrontal Cortex pathology, Transcriptome, Bipolar Disorder genetics, Bipolar Disorder pathology, Schizophrenia genetics, Schizophrenia pathology
- 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 .
- Published
- 2019
- Full Text
- View/download PDF
12. Genetic vulnerability to DUSP22 promoter hypermethylation is involved in the relation between in utero famine exposure and schizophrenia.
- Author
-
Boks MP, Houtepen LC, Xu Z, He Y, Ursini G, Maihofer AX, Rajarajan P, Yu Q, Xu H, Wu Y, Wang S, Shi JP, Hulshoff Pol HE, Strengman E, Rutten BPF, Jaffe AE, Kleinman JE, Baker DG, Hol EM, Akbarian S, Nievergelt CM, De Witte LD, Vinkers CH, Weinberger DR, Yu J, and Kahn RS
- Abstract
Epigenetic changes may account for the doubled risk to develop schizophrenia in individuals exposed to famine in utero. We therefore investigated DNA methylation in a unique sample of patients and healthy individuals conceived during the great famine in China. Subsequently, we examined two case-control samples without famine exposure in whole blood and brain tissue. To shed light on the causality of the relation between famine exposure and DNA methylation, we exposed human fibroblasts to nutritional deprivation. In the famine-exposed schizophrenia patients, we found significant hypermethylation of the dual specificity phosphatase 22 (DUSP22) gene promoter (Chr6:291687-293285) (N = 153, p = 0.01). In this sample, DUSP22 methylation was also significantly higher in patients independent of famine exposure (p = 0.025), suggesting that hypermethylation of DUSP22 is also more generally involved in schizophrenia risk. Similarly, DUSP22 methylation was also higher in two separate case-control samples not exposed to famine using DNA from whole blood (N = 64, p = 0.03) and postmortem brains (N = 214, p = 0.007). DUSP22 methylation showed strong genetic regulation across chromosomes by a region on chromosome 16 which was consistent with new 3D genome interaction data. The presence of a direct link between famine and DUSP22 transcription was supported by data from cultured human fibroblasts that showed increased methylation (p = 0.048) and expression (p = 0.019) in response to nutritional deprivation (N = 10). These results highlight an epigenetic locus that is genetically regulated across chromosomes and that is involved in the response to early-life exposure to famine and that is relevant for a major psychiatric disorder.
- Published
- 2018
- Full Text
- View/download PDF
13. Cell-specific histone modification maps in the human frontal lobe link schizophrenia risk to the neuronal epigenome.
- Author
-
Girdhar K, Hoffman GE, Jiang Y, Brown L, Kundakovic M, Hauberg ME, Francoeur NJ, Wang YC, Shah H, Kavanagh DH, Zharovsky E, Jacobov R, Wiseman JR, Park R, Johnson JS, Kassim BS, Sloofman L, Mattei E, Weng Z, Sieberts SK, Peters MA, Harris BT, Lipska BK, Sklar P, Roussos P, and Akbarian S
- Subjects
- 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, Epigenesis, Genetic genetics, Frontal Lobe metabolism, Frontal Lobe pathology, Histones genetics, Schizophrenia genetics, Schizophrenia metabolism
- 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.
- Published
- 2018
- Full Text
- View/download PDF
14. Allele-specific expression in a family quartet with autism reveals mono-to-biallelic switch and novel transcriptional processes of autism susceptibility genes.
- Author
-
Lin CY, Chang KW, Lin CY, Wu JY, Coon H, Huang PH, Ho HN, Akbarian S, Gau SS, and Huang HS
- Subjects
- Adaptor Proteins, Signal Transducing, Carrier Proteins genetics, Carrier Proteins metabolism, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Female, Humans, Low Density Lipoprotein Receptor-Related Protein-2, Male, MicroRNAs genetics, Pedigree, Prefrontal Cortex metabolism, Transcription Factors genetics, Transcription Factors metabolism, Alleles, Autism Spectrum Disorder genetics, RNA Editing
- Abstract
Autism spectrum disorder (ASD) is a highly prevalent neurodevelopmental disorder, and the exact causal mechanism is unknown. Dysregulated allele-specific expression (ASE) has been identified in persons with ASD; however, a comprehensive analysis of ASE has not been conducted in a family quartet with ASD. To fill this gap, we analyzed ASE using genomic DNA from parent and offspring and RNA from offspring's postmortem prefrontal cortex (PFC); one of the two offspring had been diagnosed with ASD. DNA- and RNA-sequencing revealed distinct ASE patterns from the PFC of both offspring. However, only the PFC of the offspring with ASD exhibited a mono-to-biallelic switch for LRP2BP and ZNF407. We also identified a novel site of RNA-editing in KMT2C in addition to new monoallelically-expressed genes and miRNAs. Our results demonstrate the prevalence of ASE in human PFC and ASE abnormalities in the PFC of a person with ASD. Taken together, these findings may provide mechanistic insights into the pathogenesis of ASD.
- Published
- 2018
- Full Text
- View/download PDF
15. Antipsychotic-induced Hdac2 transcription via NF-κB leads to synaptic and cognitive side effects.
- Author
-
Ibi D, de la Fuente Revenga M, Kezunovic N, Muguruza C, Saunders JM, Gaitonde SA, Moreno JL, Ijaz MK, Santosh V, Kozlenkov A, Holloway T, Seto J, García-Bea A, Kurita M, Mosley GE, Jiang Y, Christoffel DJ, Callado LF, Russo SJ, Dracheva S, López-Giménez JF, Ge Y, Escalante CR, Meana JJ, Akbarian S, Huntley GW, and González-Maeso J
- Subjects
- 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, Antipsychotic Agents adverse effects, Cognition Disorders chemically induced, Cognition Disorders metabolism, Histone Deacetylase 2 metabolism, NF-kappa B metabolism, Synapses metabolism
- 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-HT
2A -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.- Published
- 2017
- Full Text
- View/download PDF
16. Neuronal Deletion of Kmt2a/Mll1 Histone Methyltransferase in Ventral Striatum is Associated with Defective Spike-Timing-Dependent Striatal Synaptic Plasticity, Altered Response to Dopaminergic Drugs, and Increased Anxiety.
- Author
-
Shen EY, Jiang Y, Javidfar B, Kassim B, Loh YE, Ma Q, Mitchell AC, Pothula V, Stewart AF, Ernst P, Yao WD, Martin G, Shen L, Jakovcevski M, and Akbarian S
- Subjects
- Action Potentials drug effects, Animals, Animals, Newborn, Circadian Rhythm drug effects, Circadian Rhythm genetics, Disease Models, Animal, Female, Histone-Lysine N-Methyltransferase genetics, Locomotion drug effects, Locomotion genetics, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Myeloid-Lymphoid Leukemia Protein genetics, Nerve Tissue Proteins metabolism, Neurons drug effects, Oligonucleotide Array Sequence Analysis, Signal Transduction drug effects, Signal Transduction genetics, Action Potentials genetics, Anxiety drug therapy, Anxiety genetics, Anxiety metabolism, Anxiety physiopathology, Dopamine Agents pharmacology, Histone-Lysine N-Methyltransferase deficiency, Myeloid-Lymphoid Leukemia Protein deficiency, Neuronal Plasticity genetics, Neurons physiology, Ventral Striatum cytology
- Abstract
Lysine (K) methyltransferase 2a (Kmt2a) and other regulators of H3 lysine 4 methylation, a histone modification enriched at promoters and enhancers, are widely expressed throughout the brain, but molecular and cellular phenotypes in subcortical areas remain poorly explored. We report that Kmt2a conditional deletion in postnatal forebrain is associated with excessive nocturnal activity and with absent or blunted responses to stimulant and dopaminergic agonist drugs, in conjunction with near-complete loss of spike-timing-dependent long-term potentiation in medium spiny neurons (MSNs). Selective ablation of Kmt2a, but not the ortholog Kmt2b, in adult ventral striatum/nucleus accumbens neurons markedly increased anxiety scores in multiple behavioral paradigms. Striatal transcriptome sequencing in adult mutants identified 262 Kmt2a-sensitive genes, mostly downregulated in Kmt2a-deficient mice. Transcriptional repression includes the 5-Htr2a serotonin receptor, strongly associated with anxiety- and depression-related disorders in human and animal models. Consistent with the role of Kmt2a in promoting gene expression, the transcriptional regulators Bahcc1, Isl1, and Sp9 were downregulated and affected by H3K4 promoter hypomethylation. Therefore, Kmt2a regulates synaptic plasticity in striatal neurons and provides an epigenetic drug target for anxiety and dopamine-mediated behaviors.
- Published
- 2016
- Full Text
- View/download PDF
17. Chemogenetic Inactivation of Dorsal Anterior Cingulate Cortex Neurons Disrupts Attentional Behavior in Mouse.
- Author
-
Koike H, Demars MP, Short JA, Nabel EM, Akbarian S, Baxter MG, and Morishita H
- Subjects
- Animals, Attention drug effects, Clozapine administration & dosage, Clozapine analogs & derivatives, GABA Antagonists administration & dosage, Genetic Vectors, Gyrus Cinguli drug effects, Male, Mice, Mice, Inbred C57BL, Neurons drug effects, Pentylenetetrazole administration & dosage, Reaction Time, Receptor, Muscarinic M4 genetics, Receptor, Muscarinic M4 metabolism, Attention physiology, Gyrus Cinguli physiology, Neurons physiology, Receptor, Muscarinic M4 physiology
- Abstract
Attention is disrupted commonly in psychiatric disorders, yet mechanistic insight remains limited. Deficits in this function are associated with dorsal anterior cingulate cortex (dACC) excitotoxic lesions and pharmacological disinhibition; however, a causal relationship has not been established at the cellular level. Moreover, this association has not yet been examined in a genetically tractable species such as mice. Here, we reveal that dACC neurons causally contribute to attention processing by combining a chemogenetic approach that reversibly suppresses neural activity with a translational, touchscreen-based attention task in mice. We virally expressed inhibitory hM4Di DREADD (designer receptor exclusively activated by a designer drug) in dACC neurons, and examined the effects of this inhibitory action with the attention-based five-choice serial reaction time task. DREADD inactivation of the dACC neurons during the task significantly increased omission and correct response latencies, indicating that the neuronal activities of dACC contribute to attention and processing speed. Selective inactivation of excitatory neurons in the dACC not only increased omission, but also decreased accuracy. The effect of inactivating dACC neurons was selective to attention as response control, motivation, and locomotion remain normal. This finding suggests that dACC excitatory neurons play a principal role in modulating attention to task-relevant stimuli. This study establishes a foundation to chemogenetically dissect specific cell-type and circuit mechanisms underlying attentional behaviors in a genetically tractable species.
- Published
- 2016
- Full Text
- View/download PDF
18. Back to the past in schizophrenia genomics.
- Author
-
Sharp AJ and Akbarian S
- Subjects
- Humans, DNA Methylation genetics, Epigenesis, Genetic genetics, Epigenomics, Fetal Development genetics, Schizophrenia genetics
- Published
- 2016
- Full Text
- View/download PDF
19. The PsychENCODE project.
- Author
-
Akbarian S, Liu C, Knowles JA, Vaccarino FM, Farnham PJ, Crawford GE, Jaffe AE, Pinto D, Dracheva S, Geschwind DH, Mill J, Nairn AC, Abyzov A, Pochareddy S, Prabhakar S, Weissman S, Sullivan PF, State MW, Weng Z, Peters MA, White KP, Gerstein MB, Amiri A, Armoskus C, Ashley-Koch AE, Bae T, Beckel-Mitchener A, Berman BP, Coetzee GA, Coppola G, Francoeur N, Fromer M, Gao R, Grennan K, Herstein J, Kavanagh DH, Ivanov NA, Jiang Y, Kitchen RR, Kozlenkov A, Kundakovic M, Li M, Li Z, Liu S, Mangravite LM, Mattei E, Markenscoff-Papadimitriou E, Navarro FC, North N, Omberg L, Panchision D, Parikshak N, Poschmann J, Price AJ, Purcaro M, Reddy TE, Roussos P, Schreiner S, Scuderi S, Sebra R, Shibata M, Shieh AW, Skarica M, Sun W, Swarup V, Thomas A, Tsuji J, van Bakel H, Wang D, Wang Y, Wang K, Werling DM, Willsey AJ, Witt H, Won H, Wong CC, Wray GA, Wu EY, Xu X, Yao L, Senthil G, Lehner T, Sklar P, and Sestan N
- Subjects
- Animals, Brain pathology, Chromosome Mapping methods, Humans, Mental Disorders diagnosis, Transcriptome genetics, Brain physiology, Chromosome Mapping trends, Epigenesis, Genetic genetics, Genetic Code genetics, Mental Disorders genetics
- Published
- 2015
- Full Text
- View/download PDF
20. Analytical tools and current challenges in the modern era of neuroepigenomics.
- Author
-
Maze I, Shen L, Zhang B, Garcia BA, Shao N, Mitchell A, Sun H, Akbarian S, Allis CD, and Nestler EJ
- Subjects
- Animals, Chromatin, Genome-Wide Association Study, Humans, Statistics as Topic, Central Nervous System metabolism, Epigenomics, Proteomics
- Abstract
Over the past decade, rapid advances in epigenomics research have extensively characterized critical roles for chromatin regulatory events during normal periods of eukaryotic cell development and plasticity, as well as part of aberrant processes implicated in human disease. Application of such approaches to studies of the CNS, however, is more recent. Here we provide a comprehensive overview of available tools for analyzing neuroepigenomics data, as well as a discussion of pending challenges specific to the field of neuroscience. Integration of numerous unbiased genome-wide and proteomic approaches will be necessary to fully understand the neuroepigenome and the extraordinarily complex nature of the human brain. This will be critical to the development of future diagnostic and therapeutic strategies aimed at alleviating the vast array of heterogeneous and genetically distinct disorders of the CNS.
- Published
- 2014
- Full Text
- View/download PDF
21. Epigenetics in the human brain.
- Author
-
Houston I, Peter CJ, Mitchell A, Straubhaar J, Rogaev E, and Akbarian S
- Subjects
- DNA Methylation physiology, Humans, Mental Disorders genetics, Mental Disorders metabolism, Brain Chemistry genetics, Epigenesis, Genetic physiology
- Abstract
Many cellular constituents in the human brain permanently exit from the cell cycle during pre- or early postnatal development, but little is known about epigenetic regulation of neuronal and glial epigenomes during maturation and aging, including changes in mood and psychosis spectrum disorders and other cognitive or emotional disease. Here, we summarize the current knowledge base as it pertains to genome organization in the human brain, including the regulation of DNA cytosine methylation and hydroxymethylation, and a subset of (altogether >100) residue-specific histone modifications associated with gene expression, and silencing and various other functional chromatin states. We propose that high-resolution mapping of epigenetic markings in postmortem brain tissue or neural cultures derived from induced pluripotent cells (iPS), in conjunction with transcriptome profiling and whole-genome sequencing, will increasingly be used to define the molecular pathology of specific cases diagnosed with depression, schizophrenia, autism, or other major psychiatric disease. We predict that these highly integrative explorations of genome organization and function will provide an important alternative to conventional approaches in human brain studies, which mainly are aimed at uncovering group effects by diagnosis but generally face limitations because of cohort size.
- Published
- 2013
- Full Text
- View/download PDF
22. Epigenetic mechanisms in psychiatry.
- Author
-
Akbarian S and Nestler EJ
- Subjects
- Humans, Psychiatry methods, Epigenesis, Genetic genetics, Mental Disorders genetics, Mental Disorders psychology, Psychiatry trends
- Published
- 2013
- Full Text
- View/download PDF
23. Drug-induced activation of dopamine D(1) receptor signaling and inhibition of class I/II histone deacetylase induce chromatin remodeling in reward circuitry and modulate cocaine-related behaviors.
- Author
-
Schroeder FA, Penta KL, Matevossian A, Jones SR, Konradi C, Tapper AR, and Akbarian S
- Subjects
- Acetylation drug effects, Animals, Brain metabolism, Brain physiopathology, Brain Chemistry drug effects, Brain Chemistry genetics, Brain-Derived Neurotrophic Factor genetics, Chromatin Assembly and Disassembly genetics, Cocaine-Related Disorders genetics, Cocaine-Related Disorders metabolism, Cocaine-Related Disorders physiopathology, Disease Models, Animal, Dopamine metabolism, Dopamine Agonists pharmacology, Dopamine Uptake Inhibitors pharmacology, Enzyme Inhibitors pharmacology, Histone Deacetylase 1, Histone Deacetylases metabolism, Histones drug effects, Histones metabolism, Male, Mice, Mice, Inbred C57BL, Receptors, Dopamine D1 metabolism, Signal Transduction drug effects, Signal Transduction physiology, Synaptic Transmission drug effects, Synaptic Transmission physiology, Tyrosine 3-Monooxygenase genetics, Brain drug effects, Chromatin Assembly and Disassembly drug effects, Cocaine pharmacology, Histone Deacetylase Inhibitors, Receptors, Dopamine D1 drug effects, Reward
- Abstract
Chromatin remodeling, including histone modification, is involved in stimulant-induced gene expression and addiction behavior. To further explore the role of dopamine D(1) receptor signaling, we measured cocaine-related locomotor activity and place preference in mice pretreated for up to 10 days with the D(1) agonist SKF82958 and/or the histone deacetylase inhibitor (HDACi), sodium butyrate. Cotreatment with D(1) agonist and HDACi significantly enhanced cocaine-induced locomotor activity and place preference, in comparison to single-drug regimens. However, butyrate-mediated reward effects were transient and only apparent within 2 days after the last HDACi treatment. These behavioral changes were associated with histone modification changes in striatum and ventral midbrain: (1) a generalized increase in H3 phosphoacetylation in striatal neurons was dependent on activation of D(1) receptors; (2) H3 deacetylation at promoter sequences of tyrosine hydroxylase (Th) and brain-derived neurotrophic factor (Bdnf) in ventral midbrain, together with upregulation of the corresponding gene transcripts after cotreatment with D(1) agonist and HDACi. Collectively, these findings imply that D(1) receptor-regulated histone (phospho)acetylation and gene expression in reward circuitry is differentially regulated in a region-specific manner. Given that the combination of D(1) agonist and HDACi enhances cocaine-related sensitization and reward, the therapeutic benefits of D(1) receptor antagonists and histone acetyl-transferase inhibitors (HATi) warrant further investigation in experimental models of stimulant abuse.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.