Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Addict Biol ; 26(2): e12887, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32124535

RESUMEN

Several studies in humans and rodents suggest an association between impulsivity and activity of the stress response on the one hand and addiction vulnerability on the other. The neural cell adhesion molecule (NCAM) has been related to several neuropsychiatric disorders in humans. Constitutively NCAM-deficient (-/-) mice display enhanced novelty-induced behavior and hyperfunction of the hypothalamic-pituitary-adrenal axis. Here we hypothesize that NCAM deficiency causes an altered response to cocaine. Cocaine-induced behaviors of NCAM-/- mice and wild-type (+/+) littermates were analyzed in the conditioned place preference (CPP) test. c-fos mRNA levels were investigated by quantitative polymerase chain reaction (qPCR) to measure neural activation after exposure to the cocaine-associated context. NCAM-/- mice showed an elevated cocaine-induced sensitization, enhanced CPP, impaired extinction, and potentiated cocaine-induced hyperlocomotion and CPP after extinction. NCAM-/- showed no potentiated CPP as compared with NCAM+/+ littermates when a natural rewarding stimulus (ie, an unfamiliar female) was used, suggesting that the behavioral alterations of NCAM-/- mice observed in the CPP test are specific to the effects of cocaine. Activation of the prefrontal cortex and nucleus accumbens induced by the cocaine-associated context was enhanced in NCAM-/- compared with NCAM+/+ mice. Finally, cocaine-induced behavior correlated positively with novelty-induced behavior and plasma corticosterone levels in NCAM-/- mice and negatively with NCAM mRNA levels in the hippocampus and nucleus accumbens in wild-type mice. Our findings indicate that NCAM deficiency affects cocaine-induced CPP in mice and support the view that hyperfunction of the stress response system and reactivity to novelty predict the behavioral responses to cocaine.


Asunto(s)
Cocaína/farmacología , Sistema Hipotálamo-Hipofisario/efectos de los fármacos , Sistema Hipófiso-Suprarrenal/efectos de los fármacos , Animales , Condicionamiento Clásico , Corticosterona/sangre , Genes fos , Locomoción/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Moléculas de Adhesión de Célula Nerviosa , Núcleo Accumbens/efectos de los fármacos , Corteza Prefrontal/efectos de los fármacos , ARN Mensajero
2.
Hum Mol Genet ; 24(5): 1441-56, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25480889

RESUMEN

To investigate epigenetic contributions to Huntington's disease (HD) pathogenesis, we carried out genome-wide mapping of the transcriptional mark, trimethyl-histone H3-lysine 4 (H3K4me3) in neuronal nuclei extracted from prefrontal cortex of HD cases and controls using chromatin immunoprecipitation followed by deep-sequencing. Neuron-specific mapping of the genome-wide distribution of H3K4me3 revealed 136 differentially enriched loci associated with genes implicated in neuronal development and neurodegeneration, including GPR3, TMEM106B, PDIA6 and the Notch signaling genes hairy and enhancer of split 4 (HES4) and JAGGED2, supporting the view that the neuronal epigenome is affected in HD. Importantly, loss of H3K4me3 at CpG-rich sequences on the HES4 promoter was associated with excessive DNA methylation, reduced binding of nuclear proteins to the methylated region and altered expression of HES4 and HES4 targeted genes MASH1 and P21 involved in striatal development. Moreover, hypermethylation of HES4 promoter sequences was strikingly correlated with measures of striatal degeneration and age-of-onset in a cohort of 25 HD brains (r = 0.56, P = 0.006). Lastly, shRNA knockdown of HES4 in human neuroblastoma cells altered MASH1 and P21 mRNA expression and markedly increased mutated HTT-induced aggregates and cell death. These findings, taken together, suggest that epigenetic dysregulation of HES4 could play a critical role in modifying HD disease pathogenesis and severity.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Epigénesis Genética , Proteínas de Homeodominio/metabolismo , Enfermedad de Huntington/genética , Neostriado/patología , Adulto , Autopsia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Estudios de Casos y Controles , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Metilación de ADN , Femenino , Sitios Genéticos , Marcadores Genéticos , Estudio de Asociación del Genoma Completo , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas de Homeodominio/genética , Humanos , Masculino , Neostriado/metabolismo , Neuronas/citología , Neuronas/metabolismo , Filogenia , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Factor de Transcripción HES-1
3.
Adv Exp Med Biol ; 978: 169-183, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28523546

RESUMEN

Major depressive disorder (MDD) is a multifactorial disease, weakly linked to multiple genetic risk factors. In contrast to that, environmental factors and "gene × environment" interaction between specific risk genes and environmental factors, such as severe or early stress exposure, have been strongly linked to MDD vulnerability. Stressors can act on the interface between an organism and the environment, the epigenome. The molecular foundation for the impact of stressors on the risk to develop MDD is based on the hormonal stress response itself: the glucocorticoid receptor (GR, encoded by NR3C1). NR3C1 can directly interact with the epigenome in the cell nucleus. Besides DNA methylation, histone modifications have been reported to be crucial targets for the interaction with the stress response system. Here, we review critical findings on the impact of the most relevant histone modifications, i.e. histone acetylation and methylation, in the context of MDD and related animal models. We discuss new treatment options which have been based on these findings, including histone deacetylase inhibitors (HDACis) and drugs targeting specific histone marks, closely linked to psychiatric disease. In this context we talk about contemporary and future approaches required to fully understand (1) the epigenetics of stress-related disease and (2) the mode of action of potential MDD drugs targeting histone modifications. This includes harnessing the unprecedented potentials of genome-wide analysis of the epigenome and transcriptome, in a cell type-specific manner, and the use of epigenome editing technologies to clearly link epigenetic marks on specific genomic loci to functional relevance.


Asunto(s)
Trastorno Depresivo Mayor/genética , Epigénesis Genética/genética , Código de Histonas/genética , Código de Histonas/fisiología , Animales , Antidepresivos/farmacología , Antidepresivos/uso terapéutico , Encéfalo/metabolismo , Encéfalo/patología , Metilación de ADN , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/metabolismo , Trastorno Depresivo Mayor/fisiopatología , Modelos Animales de Enfermedad , Predicción , Código de Histonas/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Sistema Hipotálamo-Hipofisario/fisiopatología , Ratones , Proteínas del Tejido Nervioso/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/fisiología , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/fisiología , Roedores , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología , Estrés Psicológico/genética , Estrés Psicológico/metabolismo
4.
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
5.
J Neurosci ; 33(29): 11839-51, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23864674

RESUMEN

Little is known about chromosomal loopings involving proximal promoter and distal enhancer elements regulating GABAergic gene expression, including changes in schizophrenia and other psychiatric conditions linked to altered inhibition. Here, we map in human chromosome 2q31 the 3D configuration of 200 kb of linear sequence encompassing the GAD1 GABA synthesis enzyme gene locus, and we describe a loop formation involving the GAD1 transcription start site and intergenic noncoding DNA elements facilitating reporter gene expression. The GAD1-TSS(-50kbLoop) was enriched with nucleosomes epigenetically decorated with the transcriptional mark, histone H3 trimethylated at lysine 4, and was weak or absent in skin fibroblasts and pluripotent stem cells compared with neuronal cultures differentiated from them. In the prefrontal cortex of subjects with schizophrenia, GAD1-TSS(-50kbLoop) was decreased compared with controls, in conjunction with downregulated GAD1 expression. We generated transgenic mice expressing Gad2 promoter-driven green fluorescent protein-conjugated histone H2B and confirmed that Gad1-TSS(-55kbLoop), the murine homolog to GAD1-TSS(-50kbLoop), is a chromosomal conformation specific for GABAergic neurons. In primary neuronal culture, Gad1-TSS(-55kbLoop) and Gad1 expression became upregulated when neuronal activity was increased. We conclude that 3D genome architectures, including chromosomal loopings for promoter-enhancer interactions involved in the regulation of GABAergic gene expression, are conserved between the rodent and primate brain, and subject to developmental and activity-dependent regulation, and disordered in some cases with schizophrenia. More broadly, the findings presented here draw a connection between noncoding DNA, spatial genome architecture, and neuronal plasticity in development and disease.


Asunto(s)
Glutamato Descarboxilasa/genética , Corteza Prefrontal/metabolismo , Esquizofrenia/genética , Animales , Antipsicóticos/farmacología , Células Cultivadas , Cromosomas Humanos Par 2 , Clozapina/farmacología , Metilación de ADN , Regulación hacia Abajo , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Glutamato Descarboxilasa/metabolismo , Haloperidol/farmacología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Esquizofrenia/metabolismo
6.
Sci Signal ; 17(834): eadj6603, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38687825

RESUMEN

The localization, number, and function of postsynaptic AMPA-type glutamate receptors (AMPARs) are crucial for synaptic plasticity, a cellular correlate for learning and memory. The Hippo pathway member WWC1 is an important component of AMPAR-containing protein complexes. However, the availability of WWC1 is constrained by its interaction with the Hippo pathway kinases LATS1 and LATS2 (LATS1/2). Here, we explored the biochemical regulation of this interaction and found that it is pharmacologically targetable in vivo. In primary hippocampal neurons, phosphorylation of LATS1/2 by the upstream kinases MST1 and MST2 (MST1/2) enhanced the interaction between WWC1 and LATS1/2, which sequestered WWC1. Pharmacologically inhibiting MST1/2 in male mice and in human brain-derived organoids promoted the dissociation of WWC1 from LATS1/2, leading to an increase in WWC1 in AMPAR-containing complexes. MST1/2 inhibition enhanced synaptic transmission in mouse hippocampal brain slices and improved cognition in healthy male mice and in male mouse models of Alzheimer's disease and aging. Thus, compounds that disrupt the interaction between WWC1 and LATS1/2 might be explored for development as cognitive enhancers.


Asunto(s)
Hipocampo , Péptidos y Proteínas de Señalización Intracelular , Plasticidad Neuronal , Fosfoproteínas , Proteínas Serina-Treonina Quinasas , Receptores AMPA , Animales , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Masculino , Humanos , Receptores AMPA/metabolismo , Receptores AMPA/genética , Ratones , Plasticidad Neuronal/fisiología , Hipocampo/metabolismo , Vía de Señalización Hippo , Serina-Treonina Quinasa 3 , Transducción de Señal , Memoria/fisiología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Factor de Crecimiento de Hepatocito/metabolismo , Ratones Endogámicos C57BL , Enfermedad de Alzheimer/metabolismo , Fosforilación , Neuronas/metabolismo
7.
Neurobiol Dis ; 56: 104-15, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23639788

RESUMEN

Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common cause of elderly dementia. In an effort to contribute to the potential of molecular approaches to reduce degenerative processes we have tested the possibility that the neural adhesion molecule L1 ameliorates some characteristic cellular and molecular parameters associated with the disease in a mouse model of AD. Three-month-old mice overexpressing mutated forms of amyloid precursor protein and presenilin-1 under the control of a neuron-specific promoter received an injection of adeno-associated virus encoding the neuronal isoform of full-length L1 (AAV-L1) or, as negative control, green fluorescent protein (AAV-GFP) into the hippocampus and occipital cortex. Four months after virus injection, the mice were analyzed for histological and biochemical parameters of AD. AAV-L1 injection decreased the Aß plaque load, levels of Aß42, Aß42/40 ratio and astrogliosis compared with AAV-GFP controls. AAV-L1 injected mice also had increased densities of inhibitory synaptic terminals on pyramidal cells in the hippocampus when compared with AAV-GFP controls. Numbers of microglial cells/macrophages were similar in both groups, but numbers of microglial cells/macrophages per plaque were increased in AAV-L1 injected mice. To probe for a molecular mechanism that may underlie these effects, we analyzed whether L1 would directly and specifically interact with Aß. In a label-free binding assay, concentration dependent binding of the extracellular domain of L1, but not of the close homolog of L1 to Aß40 and Aß42 was seen, with the fibronectin type III homologous repeats 1-3 of L1 mediating this effect. Aggregation of Aß42 in vitro was reduced in the presence of the extracellular domain of L1. The combined observations indicate that L1, when overexpressed in neurons and glia, reduces several histopathological hallmarks of AD in mice, possibly by reduction of Aß aggregation. L1 thus appears to be a candidate molecule to ameliorate the pathology of AD, when applied in therapeutically viable treatment schemes.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Molécula L1 de Adhesión de Célula Nerviosa/uso terapéutico , Enfermedad de Alzheimer/patología , Animales , Western Blotting , Encéfalo/patología , Dependovirus/genética , Ensayo de Inmunoadsorción Enzimática , Gliosis/patología , Proteínas Fluorescentes Verdes , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Microglía/efectos de los fármacos , Lóbulo Occipital/metabolismo , Lóbulo Occipital/patología , Placa Amiloide/patología , Unión Proteica , Células Piramidales/efectos de los fármacos , Receptores CCR2/metabolismo , Fijación del Tejido
8.
Stress ; 16(6): 638-46, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24000815

RESUMEN

The neural cell adhesion molecule (NCAM) plays a crucial role in stress-related brain function, emotional behavior and memory formation. In this study, we investigated the functions of the glucocorticoid and serotonergic systems in mice constitutively deficient for NCAM (NCAM-/- mice). Our data provide evidence for a hyperfunction of the hypothalamic-pituitary-adrenal axis, with enlarged adrenal glands and increased stress-induced corticosterone release, but reduced hippocampal glucocorticoid receptor expression in NCAM-/- mice when compared to NCAM+/+ mice. We also obtained evidence for a hypofunction of 5-HT1A autoreceptors as indicated by increased 8-0H-DPAT-induced hypothermia. These findings suggest a disturbance of both humoral and neural stress systems in NCAM-/- mice. Accordingly, we not only confirmed previously observed hyperarousal of NCAM-/- mice in various anxiety tests, but also observed an increased response to novelty exposure in these animals. Spatial learning deficits of the NCAM-/- mice in a Morris Water maze persisted, even when mice were pretrained to prevent effects of novelty or stress. We suggest that NCAM-mediated processes are involved in both novelty/stress-related emotional behavior and in cognitive function during spatial learning.


Asunto(s)
Hipocampo/metabolismo , Aprendizaje por Laberinto/fisiología , Moléculas de Adhesión de Célula Nerviosa/deficiencia , 8-Hidroxi-2-(di-n-propilamino)tetralin/farmacología , Animales , Ansiedad/inducido químicamente , Corticosterona/sangre , Sistema Hipotálamo-Hipofisario/metabolismo , Hipotermia/inducido químicamente , Memoria a Corto Plazo/fisiología , Ratones , Actividad Motora/fisiología , Sistema Hipófiso-Suprarrenal/metabolismo , Receptores de Glucocorticoides/biosíntesis , Estrés Psicológico/fisiopatología
9.
Epigenetics Chromatin ; 16(1): 42, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37880732

RESUMEN

Cell-cell communication is mediated by membrane receptors and their ligands, such as the Eph/ephrin system, orchestrating cell migration during development and in diverse cancer types. Epigenetic mechanisms are key for integrating external "signals", e.g., from neighboring cells, into the transcriptome in health and disease. Previously, we reported ephrinA5 to trigger transcriptional changes of lncRNAs and protein-coding genes in cerebellar granule cells, a cell model for medulloblastoma. LncRNAs represent important adaptors for epigenetic writers through which they regulate gene expression. Here, we investigate a lncRNA-mediated targeting of DNMT1 to specific gene loci by the combined power of in silico modeling of RNA/DNA interactions and wet lab approaches, in the context of the clinically relevant use case of ephrinA5-dependent regulation of cellular motility of cerebellar granule cells. We provide evidence that Snhg15, a cancer-related lncRNA, recruits DNMT1 to the Ncam1 promoter through RNA/DNA triplex structure formation and the interaction with DNMT1. This mediates DNA methylation-dependent silencing of Ncam1, being abolished by ephrinA5 stimulation-triggered reduction of Snhg15 expression. Hence, we here propose a triple helix recognition mechanism, underlying cell motility regulation via lncRNA-targeted DNA methylation in a clinically relevant context.


Asunto(s)
ARN Largo no Codificante , ARN Largo no Codificante/genética , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , ADN , Movimiento Celular
10.
Sci Adv ; 8(46): eabo1023, 2022 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-36383658

RESUMEN

An adaptive stress response involves various mediators and circuits orchestrating a complex interplay of physiological, emotional, and behavioral adjustments. We identified a population of corticotropin-releasing hormone (CRH) neurons in the lateral part of the interstitial nucleus of the anterior commissure (IPACL), a subdivision of the extended amygdala, which exclusively innervate the substantia nigra (SN). Specific stimulation of this circuit elicits hyperactivation of the hypothalamic-pituitary-adrenal axis, locomotor activation, and avoidance behavior contingent on CRH receptor type 1 (CRHR1) located at axon terminals in the SN, which originate from external globus pallidus (GPe) neurons. The neuronal activity prompting the observed behavior is shaped by IPACLCRH and GPeCRHR1 neurons coalescing in the SN. These results delineate a previously unidentified tripartite CRH circuit functionally connecting extended amygdala and basal ganglia nuclei to drive locomotor activation and avoidance behavior.

11.
J Neurosci ; 30(21): 7152-67, 2010 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-20505083

RESUMEN

Histone methyltransferases specific for the histone H3-lysine 9 residue, including Setdb1 (Set domain, bifurcated 1)/Eset/Kmt1e are associated with repressive chromatin remodeling and expressed in adult brain, but potential effects on neuronal function and behavior remain unexplored. Here, we report that transgenic mice with increased Setdb1 expression in adult forebrain neurons show antidepressant-like phenotypes in behavioral paradigms for anhedonia, despair, and learned helplessness. Chromatin immunoprecipitation in conjunction with DNA tiling arrays (ChIP-chip) revealed that genomic occupancies of neuronal Setdb1 are limited to <1% of annotated genes, which include the NMDA receptor subunit NR2B/Grin2B and other ionotropic glutamate receptor genes. Chromatin conformation capture and Setdb1-ChIP revealed a loop formation tethering the NR2B/Grin2b promoter to the Setdb1 target site positioned 30 kb downstream of the transcription start site. In hippocampus and ventral striatum, two key structures in the neuronal circuitry regulating mood-related behaviors, Setdb1-mediated repressive histone methylation at NR2B/Grin2b was associated with decreased NR2B expression and EPSP insensitivity to pharmacological blockade of NR2B, and accelerated NMDA receptor desensitization consistent with a shift in NR2A/B subunit ratios. In wild-type mice, systemic treatment with the NR2B antagonist, Ro25-6981 [R-(R,S)-alpha-(4-hydroxyphenyl)-beta-methyl-4-(phenylmethyl)-1-piperidine propranol], and hippocampal small interfering RNA-mediated NR2B/Grin2b knockdown resulted in behavioral changes similar to those elicited by the Setdb1 transgene. Together, these findings point to a role for neuronal Setdb1 in the regulation of affective and motivational behaviors through repressive chromatin remodeling at a select set of target genes, resulting in altered NMDA receptor subunit composition and other molecular adaptations.


Asunto(s)
Afecto/fisiología , Conducta Animal/fisiología , Regulación de la Expresión Génica/fisiología , Proteína Metiltransferasas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Adaptación Ocular/efectos de los fármacos , Adaptación Ocular/genética , Afecto/efectos de los fármacos , Factores de Edad , Animales , Animales Recién Nacidos , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Conducta Animal/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Células Cultivadas , Cromatina/metabolismo , Inmunoprecipitación de Cromatina/métodos , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Electrochoque/efectos adversos , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Miedo/efectos de los fármacos , Miedo/fisiología , Preferencias Alimentarias/efectos de los fármacos , Preferencias Alimentarias/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , N-Metiltransferasa de Histona-Lisina , Humanos , Pérdida de Tono Postural/efectos de los fármacos , Pérdida de Tono Postural/fisiología , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuronas/ultraestructura , Técnicas de Placa-Clamp/métodos , Proteína Metiltransferasas/genética , ARN Interferente Pequeño/farmacología , Receptores de N-Metil-D-Aspartato/genética , Sacarosa/administración & dosificación
12.
Hippocampus ; 20(9): 1027-36, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19739230

RESUMEN

The growth arrest specific 5 (gas5) is a noncoding protein gene that hosts small nucleolar RNAs. Based on the observation that gas5 RNA level in the brain is highest in the hippocampus and remarkably enhanced in aged mice, we tested the hypothesis that gas5 is involved in functions controlled by the hippocampus and known to be affected by age, such as spatial learning and novelty-induced behaviors. We show that aged (22-month-old) C57BL/6 male mice have spatial-learning impairments, reduced novelty-induced exploration, and enhanced gas5 RNA levels in the hippocampus compared to young (3-month-old) mice. At both ages, levels of gas5 RNA in the hippocampus negatively correlated with novelty-induced exploration in the open field and elevated-plus maze tests. No correlations were found between gas5 RNA levels in the hippocampus and performance in the water maze test. The expression of gas5 RNA in the rest of the brain did not correlate with any behavioral parameter analyzed. Because variations in novelty-induced behaviors could be caused by stressfull experiences, we analyzed whether gas5 RNA levels in the hippocampus are regulated by acute stressors. We found that gas5 RNA levels in the hippocampus were upregulated by 50% 24 h after a psychogenic stressor (60-min olfactory contact with a rat) but were unchanged after exposure to an unfamiliar environment or after acquisition of new spatial information in a one-trial learning task. The present results suggest that strong psychogenic stressors upregulate gas5 RNA in the hippocampus, which in turn affects novelty-induced responses controlled by this region. We hypothesize that long-life exposure to stressors causes an age-dependent increase in hippocampal gas5 RNA levels, which could be responsible for age-related reduced novelty-induced behaviors, thus suggesting a new mechanism by which ageing and stress affect hippocampal function.


Asunto(s)
Envejecimiento/genética , Conducta Animal/fisiología , Regulación hacia Abajo/genética , Conducta Exploratoria/fisiología , Hipocampo/fisiología , ARN Nucleolar Pequeño/biosíntesis , Estrés Psicológico/genética , Regulación hacia Arriba/genética , Envejecimiento/psicología , Animales , Regulación del Desarrollo de la Expresión Génica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Nucleolar Pequeño/genética
13.
Transl Psychiatry ; 10(1): 115, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32321908

RESUMEN

Neuropsychiatric disorders are a collective of cerebral conditions with a multifactorial and polygenetic etiology. Dysregulation of epigenetic profiles in the brain is considered to play a critical role in the development of neuropsychiatric disorders. SET domain, bifurcate 1 (SETDB1), functioning as a histone H3K9 specific methyltransferase, is not only critically involved in transcriptional silencing and local heterochromatin formation, but also affects genome-wide neuronal epigenetic profiles and is essential for 3D genome integrity. Here, we provide a review of recent advances towards understanding the role of SETDB1 in the central nervous system during early neurodevelopment as well as in the adult brain, with a particular focus on studies that link its functions to neuropsychiatric disorders and related behavioral changes, and the exploration of novel therapeutic strategies targeting SETDB1.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Dominios PR-SET , Encéfalo/metabolismo , Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Histonas/metabolismo
14.
Methods Mol Biol ; 1938: 219-229, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30617983

RESUMEN

Chromatin immunoprecipitation (ChIP) in conjunction with qPCR or next generation sequencing (ChIP-seq) is used to detect protein-DNA interaction. Typically, DNA bound to a protein of interest is captured with an antibody against this protein, and DNA is then purified from DNA-protein complexes. Here, we describe a native Chromatin immunoprecipitation (N-ChIP) approach which is an efficient ChIP method with high resolution for histone modifications and a number of transcription factors. This protocol has been tailored for cultured primary rat astrocytes, and we included the preparation of astrocytic cell cultures in this protocol.


Asunto(s)
Astrocitos/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Inmunoprecipitación de Cromatina , Secuenciación de Nucleótidos de Alto Rendimiento , Animales , Técnicas de Cultivo de Célula , Histonas/metabolismo , Metilación , Ratas
15.
Endocr Connect ; 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30352410

RESUMEN

OBJECTIVE: Patients with non-functioning pituitary adenomas exhibit high morbidity and mortality rates. Growth hormone deficiency and high doses of glucocorticoid substitution therapy have been identified as corresponding risk factors. Interestingly, high levels of endogenous cortisol in, e.g., patients with post-traumatic stress disorder or patients with Cushing's disease have been linked to shorter telomere length. Telomeres are noncoding DNA regions located at the end of chromosomes consisting of repetitive DNA sequences which shorten with ageing and hereby determine cell survival. Therefore, telomere length can serve as a predictor for the onset of disease and mortality in some endocrine disorders (e.g., Cushing's disease). DESIGN/METHODS: We examine telomere length from blood in patients (n = 115) with non-functioning pituitary adenomas (NFPA) in a cross-sectional case control (n = 106, age-, gender- matched) study using qPCR. Linear regression models were used to identify independent predictors of telomere length. RESULTS: We show that patients with NFPA exhibited shorter telomeres than controls. No significant association of indices of growth hormone deficiency (IGF-1-level-SDS, years of unsubstituted growth hormone deficiency etc.) with telomere length was detected. Interestingly, linear regression analysis showed that hydrocortisone replacement dosage in patients with adrenal insufficiency (n = 52) was a significant predictor for shorter telomere length (ß = 0.377; p = 0.018) independent of potential confounders. Median split analysis revealed that higher hydrocortisone intake (> 20 mg) was associated with significantly shorter telomeres. CONCLUSION: These observations strengthen the importance of adjusted glucocorticoid treatment in NFPA patients with respect to morbidity and mortality rates.

16.
Mol Metab ; 17: 1-16, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30174229

RESUMEN

OBJECTIVE: Vulnerability to eating disorders (EDs) is broadly assumed to be associated with early life stress. However, a careful examination of the literature shows that susceptibility to EDs may depend on the type, severity and timing of the stressor and the sex of the individual. We aimed at exploring the link between chronic prenatal stress and predisposition to EDs and metabolic disease. METHODS: We used a chronic variable stress protocol during gestation to explore the metabolic response of male and female offspring to food restriction (FR), activity-based anorexia (ABA), binge eating (BE) and exposure to high fat (HF) diet. RESULTS: Contrary to controls, prenatally stressed (PNS) female offspring showed resistance to ABA and BE and displayed a lower metabolic rate leading to hyperadiposity and obesity on HF diet. Male PNS offspring showed healthy responses to FR and ABA, increased propensity to binge and improved coping with HF compared to controls. We found that long-lasting abnormal responses to metabolic challenge are linked to fetal programming and adult hypothalamic dysregulation in PNS females, resulting from sexually dimorphic adaptations in placental methylation and gene expression. CONCLUSIONS: Our results show that maternal stress may have variable and even opposing effects on ED risk, depending on the ED and the sex of the offspring.


Asunto(s)
Trastornos de Alimentación y de la Ingestión de Alimentos/fisiopatología , Enfermedades Metabólicas/fisiopatología , Animales , Trastorno por Atracón , Peso Corporal/fisiología , Dieta Alta en Grasa , Femenino , Hipotálamo/metabolismo , Insulina , Masculino , Ratones , Ratones Endogámicos ICR , Obesidad/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Factores Sexuales , Estrés Fisiológico/fisiología
17.
Nat Commun ; 9(1): 1596, 2018 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-29686286

RESUMEN

Anorexia nervosa (AN) is a devastating eating disorder characterized by self-starvation that mainly affects women. Its etiology is unknown, which impedes successful treatment options leading to a limited chance of full recovery. Here, we show that gestation is a vulnerable window that can influence the predisposition to AN. By screening placental microRNA expression of naive and prenatally stressed (PNS) fetuses and assessing vulnerability to activity-based anorexia (ABA), we identify miR-340 as a sexually dimorphic regulator involved in prenatal programming of ABA. PNS caused gene-body hypermethylation of placental miR-340, which is associated with reduced miR-340 expression and increased protein levels of several target transcripts, GR, Cry2 and H3F3b. MiR-340 is linked to the expression of several nutrient transporters both in mice and human placentas. Using placenta-specific lentiviral transgenes and embryo transfer, we demonstrate the key role miR-340 plays in the mechanism involved in early life programming of ABA.


Asunto(s)
Anorexia Nerviosa/genética , MicroARNs/metabolismo , Placenta/metabolismo , Efectos Tardíos de la Exposición Prenatal/genética , Adulto , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Transferencia de Embrión , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Intercambio Materno-Fetal , Ratones , Ratones Endogámicos ICR , Ratones Transgénicos , MicroARNs/genética , Actividad Motora , Embarazo , Análisis de Secuencia de ARN , Factores Sexuales
18.
Neuron ; 99(2): 389-403.e9, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30048615

RESUMEN

N6-methyladenosine (m6A) and N6,2'-O-dimethyladenosine (m6Am) are abundant mRNA modifications that regulate transcript processing and translation. The role of both, here termed m6A/m, in the stress response in the adult brain in vivo is currently unknown. Here, we provide a detailed analysis of the stress epitranscriptome using m6A/m-seq, global and gene-specific m6A/m measurements. We show that stress exposure and glucocorticoids region and time specifically alter m6A/m and its regulatory network. We demonstrate that deletion of the methyltransferase Mettl3 or the demethylase Fto in adult neurons alters the m6A/m epitranscriptome, increases fear memory, and changes the transcriptome response to fear and synaptic plasticity. Moreover, we report that regulation of m6A/m is impaired in major depressive disorder patients following glucocorticoid stimulation. Our findings indicate that brain m6A/m represents a novel layer of complexity in gene expression regulation after stress and that dysregulation of the m6A/m response may contribute to the pathophysiology of stress-related psychiatric disorders.


Asunto(s)
Adenosina/análogos & derivados , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Adenosina/genética , Adenosina/metabolismo , Adulto , Animales , Línea Celular Transformada , Humanos , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Estrés Psicológico/psicología
19.
Nat Neurosci ; 21(6): 803-807, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29786085

RESUMEN

The interplay between corticotropin-releasing hormone (CRH) and the dopaminergic system has predominantly been studied in addiction and reward, while CRH-dopamine interactions in anxiety are scarcely understood. We describe a new population of CRH-expressing, GABAergic, long-range-projecting neurons in the extended amygdala that innervate the ventral tegmental area and alter anxiety following chronic CRH depletion. These neurons are part of a distinct CRH circuit that acts anxiolytically by positively modulating dopamine release.


Asunto(s)
Amígdala del Cerebelo/fisiología , Ansiedad/psicología , Hormona Liberadora de Corticotropina/deficiencia , Dopamina/metabolismo , Neuronas GABAérgicas/fisiología , Amígdala del Cerebelo/citología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Hormona Liberadora de Corticotropina/farmacología , Espinas Dendríticas/ultraestructura , Inyecciones , Masculino , Ratones , Ratones Noqueados , Actividad Motora , Optogenética , Percepción del Dolor , Receptores de Hormona Liberadora de Corticotropina/metabolismo , Área Tegmental Ventral/citología , Área Tegmental Ventral/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA