Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Neurosci ; 35(13): 5097-108, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25834037

ABSTRACT

Neuronal histone H3-lysine 4 methylation landscapes are defined by sharp peaks at gene promoters and other cis-regulatory sequences, but molecular and cellular phenotypes after neuron-specific deletion of H3K4 methyl-regulators remain largely unexplored. We report that neuronal ablation of the H3K4-specific methyltransferase, Kmt2a/Mixed-lineage leukemia 1 (Mll1), in mouse postnatal forebrain and adult prefrontal cortex (PFC) is associated with increased anxiety and robust cognitive deficits without locomotor dysfunction. In contrast, only mild behavioral phenotypes were observed after ablation of the Mll1 ortholog Kmt2b/Mll2 in PFC. Impaired working memory after Kmt2a/Mll1 ablation in PFC neurons was associated with loss of training-induced transient waves of Arc immediate early gene expression critical for synaptic plasticity. Medial prefrontal layer V pyramidal neurons, a major output relay of the cortex, demonstrated severely impaired synaptic facilitation and temporal summation, two forms of short-term plasticity essential for working memory. Chromatin immunoprecipitation followed by deep sequencing in Mll1-deficient cortical neurons revealed downregulated expression and loss of the transcriptional mark, trimethyl-H3K4, at <50 loci, including the homeodomain transcription factor Meis2. Small RNA-mediated Meis2 knockdown in PFC was associated with working memory defects similar to those elicited by Mll1 deletion. Therefore, mature prefrontal neurons critically depend on maintenance of Mll1-regulated H3K4 methylation at a subset of genes with an essential role in cognition and emotion.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Memory, Short-Term/physiology , Myeloid-Lymphoid Leukemia Protein/metabolism , Neuronal Plasticity/physiology , Prefrontal Cortex/physiology , Animals , Behavior, Animal/physiology , Cytoskeletal Proteins/metabolism , Gene Expression , Gene Knockdown Techniques , Homeodomain Proteins/drug effects , Homeodomain Proteins/genetics , Male , Methylation , Mice , Mice, Transgenic , Mutation , Nerve Tissue Proteins/metabolism , Prosencephalon/physiology , Pyramidal Cells/physiology
2.
PLoS Biol ; 10(11): e1001427, 2012.
Article in English | MEDLINE | ID: mdl-23185133

ABSTRACT

Cognitive abilities and disorders unique to humans are thought to result from adaptively driven changes in brain transcriptomes, but little is known about the role of cis-regulatory changes affecting transcription start sites (TSS). Here, we mapped in human, chimpanzee, and macaque prefrontal cortex the genome-wide distribution of histone H3 trimethylated at lysine 4 (H3K4me3), an epigenetic mark sharply regulated at TSS, and identified 471 sequences with human-specific enrichment or depletion. Among these were 33 loci selectively methylated in neuronal but not non-neuronal chromatin from children and adults, including TSS at DPP10 (2q14.1), CNTN4 and CHL1 (3p26.3), and other neuropsychiatric susceptibility genes. Regulatory sequences at DPP10 and additional loci carried a strong footprint of hominid adaptation, including elevated nucleotide substitution rates and regulatory motifs absent in other primates (including archaic hominins), with evidence for selective pressures during more recent evolution and adaptive fixations in modern populations. Chromosome conformation capture at two neurodevelopmental disease loci, 2q14.1 and 16p11.2, revealed higher order chromatin structures resulting in physical contact of multiple human-specific H3K4me3 peaks spaced 0.5-1 Mb apart, in conjunction with a novel cis-bound antisense RNA linked to Polycomb repressor proteins and downregulated DPP10 expression. Therefore, coordinated epigenetic regulation via newly derived TSS chromatin could play an important role in the emergence of human-specific gene expression networks in brain that contribute to cognitive functions and neurological disease susceptibility in modern day humans.


Subject(s)
DNA Methylation , Histones/metabolism , Neurons/metabolism , Prefrontal Cortex/cytology , Transcription Initiation Site , Adult , Animals , Base Sequence , Child , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromosome Mapping , Cognition , Contactins/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Epigenesis, Genetic , Evolution, Molecular , Gene Regulatory Networks , Genetic Loci , Histones/genetics , Humans , Lysine/metabolism , Macaca/genetics , Mental Disorders/genetics , Neurons/cytology , Pan troglodytes/genetics , Phylogeny , Polycomb-Group Proteins/metabolism , Prefrontal Cortex/metabolism , Regulatory Sequences, Nucleic Acid , Species Specificity , Transcription, Genetic
3.
Biotechnol Bioeng ; 111(2): 295-308, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23904288

ABSTRACT

In biotechnological screening and production, oxygen supply is a crucial parameter. Even though oxygen transfer is well documented for viscous cultivations in stirred tanks, little is known about the gas/liquid oxygen transfer in shake flask cultures that become increasingly viscous during cultivation. Especially the oxygen transfer into the liquid film, adhering on the shake flask wall, has not yet been described for such cultivations. In this study, the oxygen transfer of chemical and microbial model experiments was measured and the suitability of the widely applied film theory of Higbie was studied. With numerical simulations of Fick's law of diffusion, it was demonstrated that Higbie's film theory does not apply for cultivations which occur at viscosities up to 10 mPa s. For the first time, it was experimentally shown that the maximum oxygen transfer capacity OTRmax increases in shake flasks when viscosity is increased from 1 to 10 mPa s, leading to an improved oxygen supply for microorganisms. Additionally, the OTRmax does not significantly undermatch the OTRmax at waterlike viscosities, even at elevated viscosities of up to 80 mPa s. In this range, a shake flask is a somehow self-regulating system with respect to oxygen supply. This is in contrary to stirred tanks, where the oxygen supply is steadily reduced to only 5% at 80 mPa s. Since, the liquid film formation at shake flask walls inherently promotes the oxygen supply at moderate and at elevated viscosities, these results have significant implications for scale-up.


Subject(s)
Bacteria/growth & development , Bacteria/metabolism , Bioreactors , Culture Media/chemistry , Oxygen/metabolism , Viscosity
4.
Nat Genet ; 56(7): 1366-1370, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38937606

ABSTRACT

The factors driving or preventing pathological expansion of tandem repeats remain largely unknown. Here, we assessed the FGF14 (GAA)·(TTC) repeat locus in 2,530 individuals by long-read and Sanger sequencing and identified a common 5'-flanking variant in 70.34% of alleles analyzed (3,463/4,923) that represents the phylogenetically ancestral allele and is present on all major haplotypes. This common sequence variation is present nearly exclusively on nonpathogenic alleles with fewer than 30 GAA-pure triplets and is associated with enhanced stability of the repeat locus upon intergenerational transmission and increased Fiber-seq chromatin accessibility.


Subject(s)
Alleles , Fibroblast Growth Factors , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Humans , Haplotypes , Genetic Variation , Genetic Loci
5.
Hum Mol Genet ; 20(9): 1701-11, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21300694

ABSTRACT

Spinal muscular atrophy (SMA), an inherited disease of motor neuron dysfunction, results from insufficient levels of the survival motor neuron (SMN) protein. Movement of the SMN protein as granules within cultured axons suggests that the pathogenesis of SMA may involve defects in neuronal transport, yet the nature of axon transport vesicles remains enigmatic. Here we show that SMN directly binds to the α-subunit of the coat protein I (COPI) vesicle coat protein. The α-COP protein co-immunoprecipitates with SMN, small nuclear ribonucleoprotein-associated assembly factors and ß-actin mRNA. Although typically Golgi associated, in neuronal cells α-COP localizes to lamellipodia and growth cones and moves within the axon, with a subset of these granules traveling together with SMN. Depletion of α-COP resulted in mislocalization of SMN and actin at the leading edge at the lamellipodia. We propose that neurons utilize the Golgi-associated COPI vesicle to deliver cargoes necessary for motor neuron integrity and function.


Subject(s)
Axons/metabolism , Coat Protein Complex I/metabolism , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Transport Vesicles/metabolism , Animals , Cell Line , Cell Survival , Coat Protein Complex I/genetics , Disease Models, Animal , Humans , Mice , Motor Neurons/cytology , Muscular Atrophy, Spinal/genetics , Protein Binding , Protein Transport , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism , Transport Vesicles/genetics
6.
Nat Commun ; 12(1): 7243, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34903713

ABSTRACT

Regulatory mechanisms associated with repeat-rich sequences and chromosomal conformations in mature neurons remain unexplored. Here, we map cell-type specific chromatin domain organization in adult mouse cerebral cortex and report strong enrichment of Endogenous Retrovirus 2 (ERV2) repeat sequences in the neuron-specific heterochromatic B2NeuN+ megabase-scaling subcompartment. Single molecule long-read sequencing and comparative Hi-C chromosomal contact mapping in wild-derived SPRET/EiJ (Mus spretus) and laboratory inbred C57BL/6J (Mus musculus) reveal neuronal reconfigurations tracking recent ERV2 expansions in the murine germline, with significantly higher B2NeuN+ contact frequencies at sites with ongoing insertions in Mus musculus. Neuronal ablation of the retrotransposon silencer Kmt1e/Setdb1 triggers B2NeuN+ disintegration and rewiring with open chromatin domains enriched for cellular stress response genes, along with severe neuroinflammation and proviral assembly with infiltration of dendrites . We conclude that neuronal megabase-scale chromosomal architectures include an evolutionarily adaptive heterochromatic organization which, upon perturbation, results in transcriptional dysregulation and unleashes ERV2 proviruses with strong neuronal tropism.


Subject(s)
Chromosomes/metabolism , Neurons/metabolism , Retroelements/genetics , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Chromosomes/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endogenous Retroviruses/genetics , Evolution, Molecular , Gene Amplification , Gene Silencing , Genes, Intracisternal A-Particle/genetics , Genome, Viral/genetics , Gliosis/genetics , Gliosis/metabolism , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Mice , Microglia/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/virology , Proviruses/genetics , Virion/genetics , Virion/metabolism
7.
Genome Med ; 13(1): 118, 2021 07 19.
Article in English | MEDLINE | ID: mdl-34281603

ABSTRACT

BACKGROUND: Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, has been associated with neurological and neuropsychiatric illness in many individuals. We sought to further our understanding of the relationship between brain tropism, neuro-inflammation, and host immune response in acute COVID-19 cases. METHODS: Three brain regions (dorsolateral prefrontal cortex, medulla oblongata, and choroid plexus) from 5 patients with severe COVID-19 and 4 controls were examined. The presence of the virus was assessed by western blot against viral spike protein, as well as viral transcriptome analysis covering > 99% of SARS-CoV-2 genome and all potential serotypes. Droplet-based single-nucleus RNA sequencing (snRNA-seq) was performed in the same samples to examine the impact of COVID-19 on transcription in individual cells of the brain. RESULTS: Quantification of viral spike S1 protein and viral transcripts did not detect SARS-CoV-2 in the postmortem brain tissue. However, analysis of 68,557 single-nucleus transcriptomes from three distinct regions of the brain identified an increased proportion of stromal cells, monocytes, and macrophages in the choroid plexus of COVID-19 patients. Furthermore, differential gene expression, pseudo-temporal trajectory, and gene regulatory network analyses revealed transcriptional changes in the cortical microglia associated with a range of biological processes, including cellular activation, mobility, and phagocytosis. CONCLUSIONS: Despite the absence of detectable SARS-CoV-2 in the brain at the time of death, the findings suggest significant and persistent neuroinflammation in patients with acute COVID-19.


Subject(s)
Brain/metabolism , COVID-19/immunology , Gene Expression Profiling/methods , Immunity/genetics , Immunity/immunology , Transcriptome , Choroid Plexus/metabolism , Gene Expression , Gene Regulatory Networks , Humans , Inflammation , Microglia , Prefrontal Cortex/metabolism , SARS-CoV-2/genetics
8.
Stem Cell Reports ; 16(3): 505-518, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33636110

ABSTRACT

The host response to SARS-CoV-2, the etiologic agent of the COVID-19 pandemic, demonstrates significant interindividual variability. In addition to showing more disease in males, the elderly, and individuals with underlying comorbidities, SARS-CoV-2 can seemingly afflict healthy individuals with profound clinical complications. We hypothesize that, in addition to viral load and host antibody repertoire, host genetic variants influence vulnerability to infection. Here we apply human induced pluripotent stem cell (hiPSC)-based models and CRISPR engineering to explore the host genetics of SARS-CoV-2. We demonstrate that a single-nucleotide polymorphism (rs4702), common in the population and located in the 3' UTR of the protease FURIN, influences alveolar and neuron infection by SARS-CoV-2 in vitro. Thus, we provide a proof-of-principle finding that common genetic variation can have an impact on viral infection and thus contribute to clinical heterogeneity in COVID-19. Ongoing genetic studies will help to identify high-risk individuals, predict clinical complications, and facilitate the discovery of drugs.


Subject(s)
COVID-19/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide/genetics , 3' Untranslated Regions/genetics , Adolescent , Adult , Animals , COVID-19/virology , Cell Line , Chlorocebus aethiops , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Female , Furin/genetics , Host-Pathogen Interactions/genetics , Humans , Induced Pluripotent Stem Cells/virology , Male , Neurons/virology , Peptide Hydrolases/genetics , SARS-CoV-2/pathogenicity , Vero Cells
9.
bioRxiv ; 2020 Sep 21.
Article in English | MEDLINE | ID: mdl-32995783

ABSTRACT

The host response to SARS-CoV-2, the etiologic agent of the COVID-19 pandemic, demonstrates significant inter-individual variability. In addition to showing more disease in males, the elderly, and individuals with underlying comorbidities, SARS-CoV-2 can seemingly render healthy individuals with profound clinical complications. We hypothesize that, in addition to viral load and host antibody repertoire, host genetic variants also impact vulnerability to infection. Here we apply human induced pluripotent stem cell (hiPSC)-based models and CRISPR-engineering to explore the host genetics of SARS-CoV-2. We demonstrate that a single nucleotide polymorphism (rs4702), common in the population at large, and located in the 3'UTR of the protease FURIN, impacts alveolar and neuron infection by SARS-CoV-2 in vitro. Thus, we provide a proof-of-principle finding that common genetic variation can impact viral infection, and thus contribute to clinical heterogeneity in SARS-CoV-2. Ongoing genetic studies will help to better identify high-risk individuals, predict clinical complications, and facilitate the discovery of drugs that might treat disease.

10.
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
11.
Nat Commun ; 10(1): 4112, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511512

ABSTRACT

Many neuropsychiatric risk genes contribute to epigenetic regulation but little is known about specific chromatin-associated mechanisms governing the formation of neuronal connectivity. Here we show that transcallosal connectivity is critically dependent on C11orf46, a nuclear protein encoded in the chromosome 11p13 WAGR risk locus. C11orf46 haploinsufficiency was associated with hypoplasia of the corpus callosum. C11orf46 knockdown disrupted transcallosal projections and was rescued by wild type C11orf46 but not the C11orf46R236H mutant associated with intellectual disability. Multiple genes encoding key regulators of axonal development, including Sema6a, were hyperexpressed in C11orf46-knockdown neurons. RNA-guided epigenetic editing of Sema6a gene promoters via a dCas9-SunTag system with C11orf46 binding normalized SEMA6A expression and rescued transcallosal dysconnectivity via repressive chromatin remodeling by the SETDB1 repressor complex. Our study demonstrates that interhemispheric communication is sensitive to locus-specific remodeling of neuronal chromatin, revealing the therapeutic potential for shaping the brain's connectome via gene-targeted designer activators and repressor proteins.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Corpus Callosum/metabolism , Epigenesis, Genetic , Gene Editing , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Semaphorins/genetics , Animals , Axons/metabolism , Epigenome , Gene Expression Regulation , Genetic Predisposition to Disease , HEK293 Cells , Histone-Lysine N-Methyltransferase , Humans , Mice, Inbred C57BL , Nerve Net/metabolism , Neurites/metabolism , Phenotype , Protein Binding , Protein Methyltransferases/metabolism
12.
Nat Genet ; 49(8): 1239-1250, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28671686

ABSTRACT

We report locus-specific disintegration of megabase-scale chromosomal conformations in brain after neuronal ablation of Setdb1 (also known as Kmt1e; encodes a histone H3 lysine 9 methyltransferase), including a large topologically associated 1.2-Mb domain conserved in humans and mice that encompasses >70 genes at the clustered protocadherin locus (hereafter referred to as cPcdh). The cPcdh topologically associated domain (TADcPcdh) in neurons from mutant mice showed abnormal accumulation of the transcriptional regulator and three-dimensional (3D) genome organizer CTCF at cryptic binding sites, in conjunction with DNA cytosine hypomethylation, histone hyperacetylation and upregulated expression. Genes encoding stochastically expressed protocadherins were transcribed by increased numbers of cortical neurons, indicating relaxation of single-cell constraint. SETDB1-dependent loop formations bypassed 0.2-1 Mb of linear genome and radiated from the TADcPcdh fringes toward cis-regulatory sequences within the cPcdh locus, counterbalanced shorter-range facilitative promoter-enhancer contacts and carried loop-bound polymorphisms that were associated with genetic risk for schizophrenia. We show that the SETDB1 repressor complex, which involves multiple KRAB zinc finger proteins, shields neuronal genomes from excess CTCF binding and is critically required for structural maintenance of TADcPcdh.


Subject(s)
Chromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Neurons/metabolism , Animals , CCCTC-Binding Factor , Cadherins/genetics , Cell Line , DNA Methylation , Epigenesis, Genetic , Female , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Humans , Male , Mice , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Domains , Repressor Proteins/metabolism
13.
Neuropsychopharmacology ; 41(10): 2512-20, 2016 09.
Article in English | MEDLINE | ID: mdl-27074815

ABSTRACT

Genetic factors impact behavioral traits relevant to numerous psychiatric disorders and risk-taking behaviors, and different lines of evidence have indicated that discrete neurobiological systems contribute to such individual differences. In this study, we explored the relationship of genetic variants of the prodynorphin (PDYN) gene, which is enriched in the striatonigral/striatomesencephalic pathway, a key neuronal circuit implicated in positive 'Go' behavioral choice and action. Our multidisciplinary approach revealed that the single nucleotide polymorphism (SNP) rs2235749 (in high linkage disequilibrium with rs910080) modifies striatal PDYN expression via impaired binding of miR-365, a microRNA that targets the PDYN 3'-untranslated region (3'UTR), and is significantly associated to novelty- and reward-related behavioral traits in humans and translational animal models. Carriers of the rs2235749G allele exhibited increased levels of PDYN 3'UTR in vitro and had elevated mRNA expression in the medial nucleus accumbens shell (NAcSh) and caudate nucleus in postmortem human brains. There was an association of rs2235749 with novelty-seeking trait and a strong genotype-dose association with positive reinforcement behavior in control subjects, which differed in cannabis-dependent individuals. Using lentiviral miRZip-365 constructs selectively expressed in Pdyn-neurons of the NAcSh, we demonstrated that the Pdyn-miR365 interaction in the NAcSh directly influences novelty-seeking exploratory behavior and facilitates self-administration of natural reward. Overall, this translational study suggests that genetically determined miR-365-mediated epigenetic regulation of PDYN expression in mesolimbic striatonigral/striatomesencephalic circuits possibly contributes to novelty seeking and positive reinforcement traits.


Subject(s)
Corpus Striatum/cytology , Enkephalins/genetics , Exploratory Behavior/physiology , MicroRNAs/metabolism , Neurons/metabolism , Polymorphism, Single Nucleotide/genetics , Protein Precursors/genetics , Reinforcement, Psychology , 3' Untranslated Regions/genetics , Animals , Conditioning, Operant/physiology , Decision Making , Enkephalins/metabolism , Genotype , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Male , MicroRNAs/genetics , Protein Precursors/metabolism , RNA, Messenger/metabolism , Rats , Rats, Long-Evans , Substantia Nigra/cytology , Transduction, Genetic , Transfection
14.
Biol Psychiatry ; 80(10): 765-774, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27184921

ABSTRACT

BACKGROUND: Early childhood malnutrition affects 113 million children worldwide, impacting health and increasing vulnerability for cognitive and behavioral disorders later in life. Molecular signatures after childhood malnutrition, including the potential for intergenerational transmission, remain unexplored. METHODS: We surveyed blood DNA methylomes (~483,000 individual CpG sites) in 168 subjects across two generations, including 50 generation 1 individuals hospitalized during the first year of life for moderate to severe protein-energy malnutrition, then followed up to 48 years in the Barbados Nutrition Study. Attention deficits and cognitive performance were evaluated with the Connors Adult Attention Rating Scale and Wechsler Abbreviated Scale of Intelligence. Expression of nutrition-sensitive genes was explored by quantitative reverse transcriptase polymerase chain reaction in rat prefrontal cortex. RESULTS: We identified 134 nutrition-sensitive, differentially methylated genomic regions, with most (87%) specific for generation 1. Multiple neuropsychiatric risk genes, including COMT, IFNG, MIR200B, SYNGAP1, and VIPR2 showed associations of specific methyl-CpGs with attention and IQ. IFNG expression was decreased in prefrontal cortex of rats showing attention deficits after developmental malnutrition. CONCLUSIONS: Early childhood malnutrition entails long-lasting epigenetic signatures associated with liability for attention and cognition, and limited potential for intergenerational transmission.


Subject(s)
Attention Deficit Disorder with Hyperactivity/etiology , Behavior, Animal , Cognitive Dysfunction/etiology , DNA Methylation , Epigenesis, Genetic , Prefrontal Cortex/metabolism , Protein-Energy Malnutrition/complications , Adolescent , Adult , Animals , Attention Deficit Disorder with Hyperactivity/genetics , Barbados , Cognitive Dysfunction/genetics , DNA Methylation/genetics , Disease Models, Animal , Epigenesis, Genetic/genetics , Follow-Up Studies , Humans , Infant , Middle Aged , Nutrition Surveys , Protein-Energy Malnutrition/genetics , Rats , Young Adult
15.
Schizophr Res ; 167(1-3): 28-34, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25458568

ABSTRACT

Expression of GAD1 GABA synthesis enzyme is highly regulated by neuronal activity and reaches mature levels in the prefrontal cortex not before adolescence. A significant portion of cases diagnosed with schizophrenia show deficits in GAD1 RNA and protein levels in multiple areas of adult cerebral cortex, possibly reflecting molecular or cellular defects in subtypes of GABAergic interneurons essential for network synchronization and cognition. Here, we review 20years of progress towards a better understanding of disease-related regulation of GAD1 gene expression. For example, deficits in cortical GAD1 RNA in some cases of schizophrenia are associated with changes in the epigenetic architecture of the promoter, affecting DNA methylation patterns and nucleosomal histone modifications. These localized chromatin defects at the 5' end of GAD1 are superimposed by disordered locus-specific chromosomal conformations, including weakening of long-range promoter-enhancer loopings and physical disconnection of GAD1 core promoter sequences from cis-regulatory elements positioned 50 kilobases further upstream. Studies on the 3-dimensional architecture of the GAD1 locus in neurons, including developmentally regulated higher order chromatin compromised by the disease process, together with exploration of locus-specific epigenetic interventions in animal models, could pave the way for future treatments of psychosis and schizophrenia.


Subject(s)
Gene Expression Regulation/physiology , Glutamate Decarboxylase/metabolism , Prefrontal Cortex/metabolism , Schizophrenia/pathology , Animals , Glutamate Decarboxylase/genetics , Humans , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Prog Mol Biol Transl Sci ; 128: 199-228, 2014.
Article in English | MEDLINE | ID: mdl-25410546

ABSTRACT

Complex mechanisms shape the genome of brain cells into transcriptional units, clusters of condensed chromatin, and many other features that distinguish between various cell types and developmental stages sharing the same genetic material. Only a few years ago, the field's focus was almost entirely on a single mark, CpG methylation; the emerging complexity of neuronal and glial epigenomes now includes multiple types of DNA cytosine methylation, more than 100 residue-specific posttranslational histone modifications and histone variants, all of which superimposed by a dynamic and highly regulated three-dimensional organization of the chromosomal material inside the cell nucleus. Here, we provide an update on the most innovative approaches in neuroepigenetics and their potential contributions to approach cognitive functions and disorders unique to human. We propose that comprehensive, cell type-specific mappings of DNA and histone modifications, chromatin-associated RNAs, and chromosomal "loopings" and other determinants of three-dimensional genome organization will critically advance insight into the pathophysiology of the disease. For example, superimposing the epigenetic landscapes of neuronal and glial genomes onto genetic maps for complex disorders, ranging from Alzheimer's disease to schizophrenia, could provide important clues about neurological function for some of the risk-associated noncoding sequences in the human genome.


Subject(s)
Brain/physiology , Epigenesis, Genetic , Biomarkers/metabolism , Chromatin/metabolism , Genetic Association Studies , Humans
17.
Neuron ; 84(5): 997-1008, 2014 Dec 03.
Article in English | MEDLINE | ID: mdl-25467983

ABSTRACT

Three-dimensional chromosomal conformations regulate transcription by moving enhancers and regulatory elements into spatial proximity with target genes. Here we describe activity-regulated long-range loopings bypassing up to 0.5 Mb of linear genome to modulate NMDA glutamate receptor GRIN2B expression in human and mouse prefrontal cortex. Distal intronic and 3' intergenic loop formations competed with repressor elements to access promoter-proximal sequences, and facilitated expression via a "cargo" of AP-1 and NRF-1 transcription factors and TALE-based transcriptional activators. Neuronal deletion or overexpression of Kmt2a/Mll1 H3K4- and Kmt1e/Setdb1 H3K9-methyltransferase was associated with higher-order chromatin changes at distal regulatory Grin2b sequences and impairments in working memory. Genetic polymorphisms and isogenic deletions of loop-bound sequences conferred liability for cognitive performance and decreased GRIN2B expression. Dynamic regulation of chromosomal conformations emerges as a novel layer for transcriptional mechanisms impacting neuronal signaling and cognition.


Subject(s)
Chromatin/metabolism , Cognition/physiology , Gene Expression Regulation/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Aged , Aged, 80 and over , Animals , Animals, Newborn , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/ultrastructure , Chromatin/drug effects , Cognition/drug effects , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Neurons/metabolism , Neurons/ultrastructure , Polymorphism, Single Nucleotide/genetics , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/drug therapy , Schizophrenia/genetics , Schizophrenia/pathology , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
18.
Neuropsychopharmacology ; 38(1): 183-97, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22643929

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.


Subject(s)
Brain Chemistry/genetics , Epigenesis, Genetic/physiology , DNA Methylation/physiology , Humans , Mental Disorders/genetics , Mental Disorders/metabolism
19.
Trends Mol Med ; 17(7): 372-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21429800

ABSTRACT

Alterations in histone lysine methylation and other epigenetic regulators of gene expression contribute to changes in brain transcriptomes in mood and psychosis spectrum disorders, including depression and schizophrenia. Genetic association studies and animal models implicate multiple lysine methyltransferases and demethylases in the neurobiology of emotion and cognition. Here, we review the role of histone lysine methylation and transcriptional regulation in normal and diseased neurodevelopment and discuss various methyltransferases and demethylases as potential therapeutic targets in the treatment of neuropsychiatric disease.


Subject(s)
Histones/metabolism , Psychotic Disorders/pathology , Animals , Epigenomics , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Humans , Methylation , Protein Processing, Post-Translational
20.
Biotechnol Bioeng ; 98(5): 999-1007, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17497734

ABSTRACT

In this paper a novel and easily applied method to measure the mass transfer resistance of the sterile closures (e.g. cotton plug) of shaken bioreactors is introduced. This method requires no investment in special equipment (e.g. an oxygen sensor) and can be performed with the materials usually available in typical laboratories. The method is based on the model of Henzler et al. (1986), which mechanistically describes mass transfer through the sterile closure of a shaken bioreactor based on diffusion coupled with Stefan convection. The concentration dependency of the multi-component diffusion coefficients is taken into account. The water loss from two equivalent shaken bioreactors equipped with sterile closures during several days of shaking is measured. One flask contains distilled water, the other a saturated salt solution. From the water evaporation rate in each of the two flasks, the new model presented calculates the relative humidity in the environment, the average diffusion coefficient of oxygen in the sterile closure (D(O2)), and the diffusion coefficient of carbon dioxide (D(CO2)) . The diffusion coefficient of carbon dioxide (D(CO2)) only depends on the density and material properties of the sterile closure and not on the gas concentrations and is, therefore, an ideal parameter for the characterization of the mass transfer resistance. This new method is validated experimentally by comparing the diffusion coefficient of oxygen (D(O2)) to a measurement by the classic dynamic method; and by comparing the calculated relative humidity in the environment to a humidity sensor measurement.


Subject(s)
Bioreactors/microbiology , Gases/chemistry , Models, Theoretical , Water/chemistry , Bacteriological Techniques/methods , Carbon Dioxide/chemistry , Convection , Cotton Fiber , Diffusion , Humidity , Oxygen/chemistry , Partial Pressure , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL