Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 12(1): 7243, 2021 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-34903713

RESUMEN

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.


Asunto(s)
Cromosomas/metabolismo , Neuronas/metabolismo , Retroelementos/genética , Animales , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Cromosomas/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Retrovirus Endógenos/genética , Evolución Molecular , Amplificación de Genes , Silenciador del Gen , Genes de Partícula A Intracisternal/genética , Genoma Viral/genética , Gliosis/genética , Gliosis/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Ratones , Microglía/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/virología , Provirus/genética , Virión/genética , Virión/metabolismo
2.
Genome Med ; 12(1): 19, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32075678

RESUMEN

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.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Polimorfismo Genético , Sitios de Carácter Cuantitativo , Esquizofrenia/genética , Adipogénesis , Animales , Índice de Masa Corporal , Cromosomas/genética , Cognición , Humanos , Metabolismo de los Lípidos , Mesencéfalo/citología , Mesencéfalo/metabolismo , Ratones , Ratones Endogámicos C57BL , Neurogénesis , Esquizofrenia/metabolismo , Esquizofrenia/patología
3.
Nat Commun ; 10(1): 4112, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31511512

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/genética , Cuerpo Calloso/metabolismo , Epigénesis Genética , Edición Génica , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Semaforinas/genética , Animales , Axones/metabolismo , Epigenoma , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HEK293 , N-Metiltransferasa de Histona-Lisina , Humanos , Ratones Endogámicos C57BL , Red Nerviosa/metabolismo , Neuritas/metabolismo , Fenotipo , Unión Proteica , Proteína Metiltransferasas/metabolismo
4.
Nat Genet ; 49(8): 1239-1250, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28671686

RESUMEN

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.


Asunto(s)
Cromatina/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Neuronas/metabolismo , Animales , Factor de Unión a CCCTC , Cadherinas/genética , Línea Celular , Metilación de ADN , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Masculino , Ratones , Mutación , Conformación de Ácido Nucleico , Unión Proteica , Dominios Proteicos , Proteínas Represoras/metabolismo
5.
Biol Psychiatry ; 80(10): 765-774, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27184921

RESUMEN

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.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/etiología , Conducta Animal , Disfunción Cognitiva/etiología , Metilación de ADN , Epigénesis Genética , Corteza Prefrontal/metabolismo , Desnutrición Proteico-Calórica/complicaciones , Adolescente , Adulto , Animales , Trastorno por Déficit de Atención con Hiperactividad/genética , Barbados , Disfunción Cognitiva/genética , Metilación de ADN/genética , Modelos Animales de Enfermedad , Epigénesis Genética/genética , Estudios de Seguimiento , Humanos , Lactante , Persona de Mediana Edad , Encuestas Nutricionales , Desnutrición Proteico-Calórica/genética , Ratas , Adulto Joven
6.
J Neurosci ; 35(13): 5097-108, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25834037

RESUMEN

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.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Memoria a Corto Plazo/fisiología , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Plasticidad Neuronal/fisiología , Corteza Prefrontal/fisiología , Animales , Conducta Animal/fisiología , Proteínas del Citoesqueleto/metabolismo , Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/efectos de los fármacos , Proteínas de Homeodominio/genética , Masculino , Metilación , Ratones , Ratones Transgénicos , Mutación , Proteínas del Tejido Nervioso/metabolismo , Prosencéfalo/fisiología , Células Piramidales/fisiología
7.
Neuron ; 84(5): 997-1008, 2014 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-25467983

RESUMEN

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.


Asunto(s)
Cromatina/metabolismo , Cognición/fisiología , Regulación de la Expresión Génica/fisiología , Receptores de N-Metil-D-Aspartato/metabolismo , Anciano , Anciano de 80 o más Años , Animales , Animales Recién Nacidos , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Células Cultivadas , Corteza Cerebral/citología , Corteza Cerebral/ultraestructura , Cromatina/efectos de los fármacos , Cognición/efectos de los fármacos , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Persona de Mediana Edad , Neuronas/metabolismo , Neuronas/ultraestructura , Polimorfismo de Nucleótido Simple/genética , Receptores de N-Metil-D-Aspartato/química , Receptores de N-Metil-D-Aspartato/genética , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/genética , Esquizofrenia/patología , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
Neuropsychopharmacology ; 38(1): 183-97, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22643929

RESUMEN

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.


Asunto(s)
Química Encefálica/genética , Epigénesis Genética/fisiología , Metilación de ADN/fisiología , Humanos , Trastornos Mentales/genética , Trastornos Mentales/metabolismo
9.
PLoS Biol ; 10(11): e1001427, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23185133

RESUMEN

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.


Asunto(s)
Metilación de ADN , Histonas/metabolismo , Neuronas/metabolismo , Corteza Prefrontal/citología , Sitio de Iniciación de la Transcripción , Adulto , Animales , Secuencia de Bases , Niño , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Mapeo Cromosómico , Cognición , Contactinas/genética , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/genética , Epigénesis Genética , Evolución Molecular , Redes Reguladoras de Genes , Sitios Genéticos , Histonas/genética , Humanos , Lisina/metabolismo , Macaca/genética , Trastornos Mentales/genética , Neuronas/citología , Pan troglodytes/genética , Filogenia , Proteínas del Grupo Polycomb/metabolismo , Corteza Prefrontal/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Especificidad de la Especie , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...