Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Addict Biol ; 26(2): e12887, 2021 03.
Article in English | MEDLINE | ID: mdl-32124535

ABSTRACT

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.


Subject(s)
Cocaine/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Animals , Conditioning, Classical , Corticosterone/blood , Genes, fos , Locomotion/drug effects , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Neural Cell Adhesion Molecules , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , RNA, Messenger
2.
Hum Mol Genet ; 24(5): 1441-56, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25480889

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Epigenesis, Genetic , Homeodomain Proteins/metabolism , Huntington Disease/genetics , Neostriatum/pathology , Adult , Autopsy , Basic Helix-Loop-Helix Transcription Factors/genetics , Case-Control Studies , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Methylation , Female , Genetic Loci , Genetic Markers , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Homeodomain Proteins/genetics , Humans , Male , Neostriatum/metabolism , Neurons/cytology , Neurons/metabolism , Phylogeny , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription Factor HES-1
3.
Adv Exp Med Biol ; 978: 169-183, 2017.
Article in English | MEDLINE | ID: mdl-28523546

ABSTRACT

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.


Subject(s)
Depressive Disorder, Major/genetics , Epigenesis, Genetic/genetics , Histone Code/genetics , Histone Code/physiology , Animals , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use , Brain/metabolism , Brain/pathology , DNA Methylation , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/metabolism , Depressive Disorder, Major/physiopathology , Disease Models, Animal , Forecasting , Histone Code/drug effects , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Hypothalamo-Hypophyseal System/physiopathology , Mice , Nerve Tissue Proteins/metabolism , Pituitary-Adrenal System/physiopathology , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/physiology , Rodentia , Stress, Physiological/genetics , Stress, Physiological/physiology , Stress, Psychological/genetics , Stress, Psychological/metabolism
4.
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
5.
J Neurosci ; 33(29): 11839-51, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23864674

ABSTRACT

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.


Subject(s)
Glutamate Decarboxylase/genetics , Prefrontal Cortex/metabolism , Schizophrenia/genetics , Animals , Antipsychotic Agents/pharmacology , Cells, Cultured , Chromosomes, Human, Pair 2 , Clozapine/pharmacology , DNA Methylation , Down-Regulation , Fibroblasts/metabolism , Gene Expression Regulation , Glutamate Decarboxylase/metabolism , Haloperidol/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Histones/genetics , Histones/metabolism , Humans , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Schizophrenia/metabolism
6.
Sci Signal ; 17(834): eadj6603, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687825

ABSTRACT

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.


Subject(s)
Hippocampus , Intracellular Signaling Peptides and Proteins , Neuronal Plasticity , Phosphoproteins , Protein Serine-Threonine Kinases , Receptors, AMPA , Animals , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Male , Humans , Receptors, AMPA/metabolism , Receptors, AMPA/genetics , Mice , Neuronal Plasticity/physiology , Hippocampus/metabolism , Hippo Signaling Pathway , Serine-Threonine Kinase 3 , Signal Transduction , Memory/physiology , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/genetics , Hepatocyte Growth Factor/metabolism , Mice, Inbred C57BL , Alzheimer Disease/metabolism , Phosphorylation , Neurons/metabolism
7.
Neurobiol Dis ; 56: 104-15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23639788

ABSTRACT

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.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecule L1/therapeutic use , Alzheimer Disease/pathology , Animals , Blotting, Western , Brain/pathology , Dependovirus/genetics , Enzyme-Linked Immunosorbent Assay , Gliosis/pathology , Green Fluorescent Proteins , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Microglia/drug effects , Occipital Lobe/metabolism , Occipital Lobe/pathology , Plaque, Amyloid/pathology , Protein Binding , Pyramidal Cells/drug effects , Receptors, CCR2/metabolism , Tissue Fixation
8.
Stress ; 16(6): 638-46, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24000815

ABSTRACT

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.


Subject(s)
Hippocampus/metabolism , Maze Learning/physiology , Neural Cell Adhesion Molecules/deficiency , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Anxiety/chemically induced , Corticosterone/blood , Hypothalamo-Hypophyseal System/metabolism , Hypothermia/chemically induced , Memory, Short-Term/physiology , Mice , Motor Activity/physiology , Pituitary-Adrenal System/metabolism , Receptors, Glucocorticoid/biosynthesis , Stress, Psychological/physiopathology
9.
Epigenetics Chromatin ; 16(1): 42, 2023 10 26.
Article in English | MEDLINE | ID: mdl-37880732

ABSTRACT

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.


Subject(s)
RNA, Long Noncoding , RNA, Long Noncoding/genetics , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , DNA , Cell Movement
10.
Sci Adv ; 8(46): eabo1023, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36383658

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-20505083

ABSTRACT

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.


Subject(s)
Affect/physiology , Behavior, Animal/physiology , Gene Expression Regulation/physiology , Protein Methyltransferases/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Adaptation, Ocular/drug effects , Adaptation, Ocular/genetics , Affect/drug effects , Age Factors , Animals , Animals, Newborn , Avoidance Learning/drug effects , Avoidance Learning/physiology , Behavior, Animal/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cells, Cultured , Chromatin/metabolism , Chromatin Immunoprecipitation/methods , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Electroshock/adverse effects , Excitatory Amino Acid Agents/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Fear/drug effects , Fear/physiology , Food Preferences/drug effects , Food Preferences/physiology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Hippocampus/cytology , Histone-Lysine N-Methyltransferase , Humans , Immobility Response, Tonic/drug effects , Immobility Response, Tonic/physiology , Maze Learning/drug effects , Maze Learning/physiology , Membrane Potentials/drug effects , Membrane Potentials/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Neurons/drug effects , Neurons/physiology , Neurons/ultrastructure , Patch-Clamp Techniques/methods , Protein Methyltransferases/genetics , RNA, Small Interfering/pharmacology , Receptors, N-Methyl-D-Aspartate/genetics , Sucrose/administration & dosage
12.
Hippocampus ; 20(9): 1027-36, 2010 Sep.
Article in English | MEDLINE | ID: mdl-19739230

ABSTRACT

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.


Subject(s)
Aging/genetics , Behavior, Animal/physiology , Down-Regulation/genetics , Exploratory Behavior/physiology , Hippocampus/physiology , RNA, Small Nucleolar/biosynthesis , Stress, Psychological/genetics , Up-Regulation/genetics , Aging/psychology , Animals , Gene Expression Regulation, Developmental/physiology , Male , Mice , Mice, Inbred C57BL , RNA, Small Nucleolar/genetics
13.
Transl Psychiatry ; 10(1): 115, 2020 04 22.
Article in English | MEDLINE | ID: mdl-32321908

ABSTRACT

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.


Subject(s)
Histone-Lysine N-Methyltransferase , PR-SET Domains , Brain/metabolism , Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/metabolism
14.
Methods Mol Biol ; 1938: 219-229, 2019.
Article in English | MEDLINE | ID: mdl-30617983

ABSTRACT

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.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Chromatin Immunoprecipitation , High-Throughput Nucleotide Sequencing , Animals , Cell Culture Techniques , Histones/metabolism , Methylation , Rats
15.
Endocr Connect ; 2018 08 01.
Article in English | MEDLINE | ID: mdl-30352410

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-30174229

ABSTRACT

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.


Subject(s)
Feeding and Eating Disorders/physiopathology , Metabolic Diseases/physiopathology , Animals , Binge-Eating Disorder , Body Weight/physiology , Diet, High-Fat , Female , Hypothalamus/metabolism , Insulin , Male , Mice , Mice, Inbred ICR , Obesity/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/physiopathology , Sex Factors , Stress, Physiological/physiology
17.
Nat Commun ; 9(1): 1596, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29686286

ABSTRACT

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.


Subject(s)
Anorexia Nervosa/genetics , MicroRNAs/metabolism , Placenta/metabolism , Prenatal Exposure Delayed Effects/genetics , Adult , Animals , Cell Line, Tumor , Disease Models, Animal , Embryo Transfer , Female , Gene Expression Regulation, Developmental/genetics , Genetic Predisposition to Disease , Humans , Male , Maternal-Fetal Exchange , Mice , Mice, Inbred ICR , Mice, Transgenic , MicroRNAs/genetics , Motor Activity , Pregnancy , Sequence Analysis, RNA , Sex Factors
18.
Neuron ; 99(2): 389-403.e9, 2018 07 25.
Article in English | MEDLINE | ID: mdl-30048615

ABSTRACT

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.


Subject(s)
Adenosine/analogs & derivatives , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism , Adenosine/genetics , Adenosine/metabolism , Adult , Animals , Cell Line, Transformed , Humans , Male , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Stress, Psychological/psychology
19.
Nat Neurosci ; 21(6): 803-807, 2018 06.
Article in English | MEDLINE | ID: mdl-29786085

ABSTRACT

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.


Subject(s)
Amygdala/physiology , Anxiety/psychology , Corticotropin-Releasing Hormone/deficiency , Dopamine/metabolism , GABAergic Neurons/physiology , Amygdala/cytology , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/physiology , Corticotropin-Releasing Hormone/pharmacology , Dendritic Spines/ultrastructure , Injections , Male , Mice , Mice, Knockout , Motor Activity , Optogenetics , Pain Perception , Receptors, Corticotropin-Releasing Hormone/metabolism , Ventral Tegmental Area/cytology , Ventral Tegmental Area/physiology
SELECTION OF CITATIONS
SEARCH DETAIL