Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Neuropsychopharmacology ; 49(8): 1276-1284, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38332016

RESUMEN

DNA cytosine methylation has been documented as a potential epigenetic mechanism of transcriptional regulation underlying opioid use disorder. However, methylation of RNA cytosine residues, which would drive another level of biological influence as an epitranscriptomic mechanism of gene and protein regulation has not been studied in the context of addiction. Here, we probed whether chronic morphine exposure could alter tRNA cytosine methylation (m5C) and resulting expression levels in the medial prefrontal cortex (mPFC), a brain region crucial for reward processing and executive function that exhibits opioid-induced molecular restructuring. We identified dynamic changes in glycine tRNA (tRNAGlyGCC) cytosine methylation, corresponding to altered expression levels of this tRNA at multiple timepoints following 15 days of daily morphine. Additionally, a robust increase in methylation, coupled with decreased expression, was present after 30 days of withdrawal, suggesting that repeated opioid administration produces changes to the tRNA regulome long after discontinuation. Furthermore, forebrain-wide knockout of neuronal Nsun2, a tRNA methyltransferase, was associated with disruption of opioid conditioned place preference, and this effect was recapitulated by regional mPFC Nsun2 knockout. Taken together, these studies provide a foundational link between the regulation of tRNA cytosine methylation and opioid reward and highlight the tRNA machinery as a potential therapeutic target in addiction.


Asunto(s)
Ratones Endogámicos C57BL , Morfina , Corteza Prefrontal , ARN de Transferencia , Recompensa , Síndrome de Abstinencia a Sustancias , Animales , Masculino , Ratones , Morfina/farmacología , Morfina/administración & dosificación , Morfina/efectos adversos , Síndrome de Abstinencia a Sustancias/genética , Corteza Prefrontal/metabolismo , Corteza Prefrontal/efectos de los fármacos , ARN de Transferencia/genética , Epigénesis Genética/efectos de los fármacos , Analgésicos Opioides/farmacología , Analgésicos Opioides/administración & dosificación , Ratones Noqueados , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Comportamiento de Búsqueda de Drogas/fisiología
2.
Acta Neuropathol ; 145(1): 29-48, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36357715

RESUMEN

Epitranscriptomic regulation adds a layer of post-transcriptional control to brain function during development and adulthood. The identification of RNA-modifying enzymes has opened the possibility of investigating the role epitranscriptomic changes play in the disease process. NOP2/Sun RNA methyltransferase 2 (NSun2) is one of the few known brain-enriched methyltransferases able to methylate mammalian non-coding RNAs. NSun2 loss of function due to autosomal-recessive mutations has been associated with neurological abnormalities in humans. Here, we show NSun2 is expressed in adult human neurons in the hippocampal formation and prefrontal cortex. Strikingly, we unravel decreased NSun2 protein expression and an increased ratio of pTau/NSun2 in the brains of patients with Alzheimer's disease (AD) as demonstrated by Western blotting and immunostaining, respectively. In a well-established Drosophila melanogaster model of tau-induced toxicity, reduction of NSun2 exacerbated tau toxicity, while overexpression of NSun2 partially abrogated the toxic effects. Conditional ablation of NSun2 in the mouse brain promoted a decrease in the miR-125b m6A levels and tau hyperphosphorylation. Utilizing human induced pluripotent stem cell (iPSC)-derived neuronal cultures, we confirmed NSun2 deficiency results in tau hyperphosphorylation. We also found that neuronal NSun2 levels decrease in response to amyloid-beta oligomers (AßO). Notably, AßO-induced tau phosphorylation and cell toxicity in human neurons could be rescued by overexpression of NSun2. Altogether, these results indicate that neuronal NSun2 deficiency promotes dysregulation of miR-125b and tau phosphorylation in AD and highlights a novel avenue for therapeutic targeting.


Asunto(s)
Enfermedad de Alzheimer , Células Madre Pluripotentes Inducidas , MicroARNs , Ratones , Animales , Humanos , Adulto , Metiltransferasas/genética , Fosforilación/genética , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , MicroARNs/genética , Proteínas tau/metabolismo , Mamíferos/metabolismo
3.
Mol Psychiatry ; 27(8): 3204-3213, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35505091

RESUMEN

Transfer (t)RNAs are 70-90 nucleotide small RNAs highly regulated by 43 different types of epitranscriptomic modifications and requiring aminoacylation ('charging') for mRNA decoding and protein synthesis. Smaller cleavage products of mature tRNAs, or tRNA fragments, have been linked to a broad variety of noncanonical functions, including translational inhibition and modulation of the immune response. Traditionally, knowledge about tRNA regulation in brain is derived from phenotypic exploration of monogenic neurodevelopmental and neurodegenerative diseases associated with rare mutations in tRNA modification genes. More recent studies point to the previously unrecognized potential of the tRNA regulome to affect memory, synaptic plasticity, and affective states. For example, in mature cortical neurons, cytosine methylation sensitivity of the glycine tRNA family (tRNAGly) is coupled to glycine biosynthesis and codon-specific alterations in ribosomal translation together with robust changes in cognition and depression-related behaviors. In this Review, we will discuss the emerging knowledge of the neuronal tRNA landscape, with a focus on epitranscriptomic tRNA modifications and downstream molecular pathways affected by alterations in tRNA expression, charging levels, and cleavage while mechanistically linking these pathways to neuropsychiatric disease and provide insight into future areas of study for this field.


Asunto(s)
Biosíntesis de Proteínas , ARN de Transferencia , Biosíntesis de Proteínas/genética , ARN de Transferencia/genética , ARN de Transferencia/metabolismo , Codón , ARN Mensajero/genética , ARN Mensajero/metabolismo , Glicina/genética
4.
Front Neurol ; 12: 745330, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777213

RESUMEN

Traumatic brain injury (TBI) affects millions of Americans each year, with extremely high prevalence in the Veteran community, and sleep disturbance is one of the most commonly reported symptoms. Reduction in the quality and amount of sleep can negatively impact recovery and result in a wide range of behavioral and physiological symptoms, such as impaired cognition, mood and anxiety disorders, and cardiovascular effects. Thus, to improve long-term patient outcomes and develop novel treatments, it is essential to understand the molecular mechanisms involved in sleep disturbance following TBI. In this effort, we performed transcriptional profiling in an established rodent model of penetrating ballistic brain injury (PBBI) in conjunction with continuous sleep/wake EEG/EMG recording of the first 24 h after injury. Rats subjected to PBBI showed profound differences in sleep architecture. Injured animals spent significantly more time in slow wave sleep and less time in REM sleep compared to sham control animals. To identify PBBI-related transcriptional differences, we then performed transcriptome-wide gene expression profiling at 24 h post-injury, which identified a vast array of immune- related genes differentially expressed in the injured cortex as well as sleep-related genes. Further, transcriptional changes associated with total time spent in various sleep stages were identified. Such molecular changes may underlie the pathology and symptoms that emerge following TBI, including neurodegeneration, sleep disturbance, and mood disorders.

5.
Front Behav Neurosci ; 14: 160, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33192359

RESUMEN

The short and long-term neurological and psychological consequences of traumatic brain injury (TBI), and especially mild TBI (mTBI) are of immense interest to the Veteran community. mTBI is a common and detrimental result of combat exposure and results in various deleterious outcomes, including mood and anxiety disorders, cognitive deficits, and post-traumatic stress disorder (PTSD). In the current study, we aimed to further define the behavioral and molecular effects of blast-related mTBI using a well-established (3 × 75 kPa, one per day on three consecutive days) repeated blast overpressure (rBOP) model in rats. We exposed adult male rats to the rBOP procedure and conducted behavioral tests for anxiety and fear conditioning at 1-1.5 months (sub-acute) or 12-13 months (chronic) following blast exposure. We also used next-generation sequencing to measure transcriptome-wide gene expression in the amygdala of sham and blast-exposed animals at the sub-acute and chronic time points. Results showed that blast-exposed animals exhibited an anxiety-like phenotype at the sub-acute timepoint but this phenotype was diminished by the chronic time point. Conversely, gene expression analysis at both sub-acute and chronic timepoints demonstrated a large treatment by timepoint interaction such that the most differentially expressed genes were present in the blast-exposed animals at the chronic time point, which also corresponded to a Bdnf-centric gene network. Overall, the current study identified changes in the amygdalar transcriptome and anxiety-related phenotypic outcomes dependent on both blast exposure and aging, which may play a role in the long-term pathological consequences of mTBI.

6.
Mol Immunol ; 125: 172-177, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32707536

RESUMEN

Recent studies suggest that microbiome derived 3(3,4-dihydroxy-phenyl) propionic acid (DHCA) attenuates IL-6 cytokine production through downregulation of the epigenetic modifier DNA Methyltransferase 1 (DNMT1) expression and inhibition of DNA methylation at the 5'-C-phosphate-G-3' (CpG)-rich IL6 sequence introns 1 and 3 in a mouse model of depression. In this study, we extended the investigation of DHCA epigenetic mechanisms in IL-6 expression in human peripheral blood mononuclear cells (PBMC). Using Lucia Luciferase reporter gene system we identified CpG-rich sequences in which of methylation is influenced by DHCA similar to what observed in response to treatment with the DNA methylation inhibitor 5-aza-2'-deoxycytidine. Correlation study showed that DNA methylation at select CpG motifs in the IL-6 promoter correlates with IL-6 gene expression. Our study suggests that DHCA is effective in reducing IL-6 expression in human PBMCs, in part, by regulation of methylation in the IL-6 promoter region.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Monocitos/efectos de los fármacos , Propionatos/farmacología , Células Cultivadas , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Regulación hacia Abajo , Regulación de la Expresión Génica/genética , Humanos , Interleucina-6/biosíntesis , Microbiota , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/genética
7.
Mol Nutr Food Res ; 62(8): e1700722, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29473292

RESUMEN

SCOPE: Stress is a known contributor to various forms of disease in humans and animals, although mechanisms are still unknown. In animals, psychosocial stress-induced depression/anxiety phenotypes are coincidental with increased inflammation in both brain and blood. The authors recently showed that a novel treatment with a select bioactive polyphenol preparation promotes resilience to stress-mediated depression/anxiety phenotypes mice. Moreover, selective bioactive phenolic compounds within the polyphenol preparation are identified that are effective in mitigating the behavioral effects of bone marrow transplantation from stressed mice. METHODS AND RESULTS: Here, an animal model of adult stress and bone marrow transplantation is used to identify an epigenetic signature of repeated social defeat stress (RSDS) that is passed through bone marrow hematopoietic progenitor cells to naïve mice, revealing the maintenance of epigenetic memory following stress both centrally and peripherally. Further, polyphenols are administered to naïve and stress-susceptible mice, demonstrating that polyphenol treatment in mice from both susceptible and naïve donors alters global DNA methylation in the central nervous system and periphery and likewise has an effect on human blood cells after immune challenge. CONCLUSIONS: Findings highlight the enduring molecular memory of stress and the possible mechanism by which select bioactive polyphenols may promote resiliency to stress. Polyphenols may be an efficacious alternative to traditional pharmacological treatments in psychiatry.


Asunto(s)
Antocianinas/uso terapéutico , Antidepresivos/uso terapéutico , Ácidos Cafeicos/uso terapéutico , Metilación de ADN , Depresión/prevención & control , Suplementos Dietéticos , Modelos Animales de Enfermedad , Glucósidos/uso terapéutico , Adulto , Animales , Antocianinas/metabolismo , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/uso terapéutico , Antidepresivos/metabolismo , Conducta Animal , Trasplante de Médula Ósea/efectos adversos , Trasplante de Médula Ósea/psicología , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Ácidos Cafeicos/metabolismo , Células Cultivadas , Depresión/inmunología , Depresión/metabolismo , Depresión/psicología , Epigénesis Genética , Glucósidos/metabolismo , Humanos , Inmunidad Celular , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Masculino , Ratones Endogámicos C57BL , Neuronas/inmunología , Neuronas/metabolismo , Neuronas/patología , Conducta Social , Estrés Psicológico/inmunología , Estrés Psicológico/patología , Estrés Psicológico/fisiopatología , Estrés Psicológico/psicología
8.
Dev Psychobiol ; 59(6): 703-714, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28767135

RESUMEN

Early life adversity is known to disrupt behavioral trajectories and many rodent models have been developed to characterize these stress-induced outcomes. One example is the scarcity-adversity model of low nesting resources. This model employs resource scarcity (i.e., low nesting materials) to elicit adverse caregiving conditions (including maltreatment) toward rodent neonates. Our lab utilizes a version of this model wherein caregiving exposures occur outside the home cage during the first postnatal week. The aim of this study was to determine adolescent and adult phenotypic outcomes associated with this model, including assessment of depressive- and anxiety-like behaviors and performance in different cognitive domains. Exposure to adverse caregiving had no effect on adolescent behavioral performance whereas exposure significantly impaired adult behavioral performance. Further, adult behavioral assays revealed substantial differences between sexes. Overall, data demonstrate the ability of repeated exposure to brief bouts of maltreatment outside the home cage in infancy to impact the development of several behavioral domains later in life.


Asunto(s)
Conducta Animal/fisiología , Conducta de Elección/fisiología , Conducta Materna/fisiología , Estrés Psicológico/fisiopatología , Animales , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Femenino , Vivienda para Animales , Masculino , Ratas , Ratas Long-Evans , Caracteres Sexuales
9.
Dev Psychopathol ; 29(2): 477-489, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28401839

RESUMEN

Negative experiences with a caregiver during infancy can result in long-term changes in brain function and behavior, but underlying mechanisms are not well understood. It is our central hypothesis that brain and behavior changes are conferred by early childhood adversity through epigenetic changes involving DNA methylation. Using a rodent model of early-life caregiver maltreatment (involving exposure to an adverse caregiving environment for postnatal days 1-7), we have previously demonstrated abnormal methylation of DNA associated with the brain-derived neurotrophic factor (Bdnf) gene in the medial prefrontal cortex (mPFC) of adult rats. The aim of the current study was to characterize Bdnf DNA methylation in specific cell populations within the mPFC. In the prefrontal cortex, there is approximately twice as many neurons as glia, and studies have recently shown differential and distinctive DNA methylation patterns in neurons versus nonneurons. Here, we extracted nuclei from the mPFC of adult animals that had experienced maltreatment and used fluorescence-activated cell sorting to isolate cell types before performing bisulfite sequencing to estimate methylation of cytosine-guanine sites. Our data indicate that early-life stress induced methylation of DNA associated with Bdnf IV in a cell-type and sex-specific manner. Specifically, females that experienced early-life maltreatment exhibited greater neuronal cytosine-guanine methylation compared to controls, while no changes were detected in Bdnf methylation in males regardless of cell type. These changes localize the specificity of our previous findings to mPFC neurons and highlight the capacity of maltreatment to cause methylation changes that are likely to have functional consequences for neuronal function.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Metilación de ADN , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Estrés Psicológico/metabolismo , Adultos Sobrevivientes del Maltrato a los Niños , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Corteza Prefrontal/citología , Ratas , Ratas Long-Evans , Factores Sexuales
10.
Int J Dev Neurosci ; 62: 56-62, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28330827

RESUMEN

DNA methylation (addition of methyl groups to cytosines) and changes in telomere length (TTAGGG repeats on the ends of chromosomes) are two molecular modifications that result from stress and could contribute to the long-term effects of intrauterine exposure to maternal stress on offspring behavior. Here, we measured methylation of DNA associated with the Brain-derived neurotrophic factor (Bdnf) gene, a gene important in development and plasticity, and telomere length in the brains of adult rat male and female offspring whose mothers were exposed to unpredictable and variable stressors throughout gestation. Males exposed to prenatal stress had greater methylation (Bdnf IV) in the medial prefrontal cortex (mPFC) compared to non-stressed male controls and stressed females. Further, prenatally-stressed animals had shorter telomeres than controls in the mPFC. Together findings indicate a long-term impact of prenatal stress on brain DNA methylation and telomere biology with relevance for behavioral and health outcomes, and contribute to a growing literature linking stress to intergenerational molecular changes.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Encéfalo/metabolismo , Metilación de ADN/genética , Epigénesis Genética/fisiología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Estrés Psicológico , Homeostasis del Telómero/fisiología , Análisis de Varianza , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Femenino , Masculino , Embarazo , Resultado del Embarazo , Ratas , Ratas Sprague-Dawley , Factores Sexuales , Estrés Psicológico/genética , Estrés Psicológico/metabolismo , Estrés Psicológico/patología
11.
Dev Psychobiol ; 58(2): 176-84, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26394891

RESUMEN

The present study investigated whether repeated early postnatal exposure to the predator odor 2,5-dihydro-2,4,5-trimethylthiazoline (TMT) alters behavioral responses to the stimulus later in life, at postnatal day (PN30). Long-Evans rat pups with their mothers were exposed for 20 min daily to TMT, water, or a noxious odor, butyric acid (BTA), during the first three weeks of life. Mothers exposed to TMT displayed more crouching and nursing behavior than those exposed to BTA, and TMT exposed pups emitted more ultrasonic vocalizations than BTA exposed pups. At PN30, rats were tested for freezing to TMT, water, or BTA. Rats exposed to TMT during the postnatal period displayed less freezing to TMT than rats exposed postnatally to water or BTA. Our data indicate that early-life experience with a predator cue has a significant impact on later fear responses to that same cue, highlighting the programming capacity of the postnatal environment on the development of behavior.


Asunto(s)
Conducta Animal/fisiología , Señales (Psicología) , Miedo/fisiología , Reacción Cataléptica de Congelación/fisiología , Conducta Materna/fisiología , Odorantes , Vocalización Animal/fisiología , Animales , Animales Recién Nacidos , Ácido Butírico , Femenino , Masculino , Ratas , Ratas Long-Evans , Tiazoles , Agua
12.
Stress ; 18(6): 607-15, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26305287

RESUMEN

Infant-caregiver experiences are major contributing factors to neural and behavioral development. Research indicates that epigenetic mechanisms provide a way in which infant-caregiver experiences affect gene activity and other downstream processes in the brain that influence behavioral development. Our laboratory previously demonstrated in a rodent model that exposure to maltreatment alters methylation of DNA associated with the brain-derived neurotrophic factor (bdnf) and reelin genes as well as mRNA of key epigenetic regulatory genes in the medial prefrontal cortex (mPFC). In the current study, we characterized patterns of histone acetylation at bdnf and reelin gene loci after our caregiver manipulations. Using a within-litter design (n = 8-10/group from eight litters), pups were exposed to adverse (maltreatment condition: exposure to a stressed caregiver) or nurturing (cross-foster condition: exposure to a nurturing caregiver) caregiving environments outside the home cage for 30 min daily during the first postnatal week. Remaining pups in a litter were left with the biological mother during each session (providing normal care controls). We then used chromatin immunoprecipitation (ChIP) and quantitative RT-PCR to measure histone 3 lysine 9/14 acetylation associated with bdnf promoters I and IV and the reelin promoter in the adult mPFC. Maltreated females had decreased acetylation at bdnf IV, while neither males nor females exhibited histone acetylation alterations at bdnf I or reelin. These data demonstrate the ability of maltreatment to have long-term consequences on histone acetylation in the mPFC, and provide further evidence of the epigenetic susceptibility of bdnf IV to the quality of infant-caregiver experiences.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Epigénesis Genética , Histonas/metabolismo , Corteza Prefrontal/metabolismo , Acetilación , Animales , Moléculas de Adhesión Celular Neuronal/genética , Moléculas de Adhesión Celular Neuronal/metabolismo , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Histonas/genética , Masculino , Conducta Materna , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , Ratas , Proteína Reelina , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo
13.
Front Behav Neurosci ; 9: 79, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25904853

RESUMEN

Early childhood is a sensitive period in which infant-caregiver experiences have profound effects on brain development and behavior. Clinical studies have demonstrated that infants who experience stress and adversity in the context of caregiving are at an increased risk for the development of psychiatric disorders. Animal models have helped to elucidate some molecular substrates of these risk factors, but a complete picture of the biological basis remains unknown. Studies continue to indicate that environmentally-driven epigenetic modifications may be an important mediator between adverse caregiving environments and psychopathology. Epigenetic modifications such as DNA methylation, which normally represses gene transcription, and microRNA processing, which interferes with both transcription and translation, show long-term changes throughout the brain and body following adverse caregiving. Recent evidence has also shown that telomeres (TTAGGG nucleotide repeats that cap the ends of DNA) exhibit long-term changes in the brain and in the periphery following exposure to adverse caregiving environments. Interestingly, telomeric enzymes and subtelomeric regions are subject to epigenetic modifications-a factor which may play an important role in regulating telomere length and contribute to future mental health. This review will focus on clinical and animal studies that highlight the long-term epigenetic and telomeric changes produced by adverse caregiving in early-life.

14.
Semin Cell Dev Biol ; 43: 76-84, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25917771

RESUMEN

While it is well-known that stress during development and adulthood can confer long-term neurobiological and behavioral consequences, investigators have only recently begun to assess underlying epigenetic modifications. In this review, we highlight clinical research and work from animal models that provide evidence of the impact of stressful experiences either during the perinatal period or adulthood on DNA methylation and behavior. Additionally, we explore the more controversial concept of transgenerational inheritance, including that associated with preconception stress experienced by the mother or father. Finally, we discuss challenges associated with the idea of transgenerational epigenetics and for the field of epigenetics in general.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética/genética , Patrón de Herencia/genética , Estrés Fisiológico/genética , Estrés Psicológico/genética , Animales , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Modelos Animales de Enfermedad , Epigenómica , Femenino , Humanos , Masculino , Ratones , Ratas
15.
Dev Psychobiol ; 56(8): 1755-63, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24752649

RESUMEN

We have previously shown in infant rats that brief and repeated experiences with a stressed dam outside the homecage (maltreatment) alters methylation of DNA associated with the brain-derived neurotrophic factor (bdnf) gene within the developing and adult prefrontal cortex. BDNF is a key mediator of activity-dependent processes that have a profound influence on neural development and plasticity. Here we examined whether maltreatment also alters bdnf DNA methylation in two additional regions known to be prominently affected by diverse forms of early life adversity in humans- the hippocampus and amygdala. We found significant bdnf DNA methylation modifications present within the adult hippocampus (dorsal and ventral) and amygdala (central/basolateral complex). We observed that the nature of change differed between sexes, gene locus (bdnf I vs. IV), and brain region. Furthermore, a manipulation that did not produce any obvious behavior difference in infants (brief and repeated experiences with a nurturing foster dam) also had long-term effects on methylation. These data provide further empirical support of DNA methylation modifications as biological consequences of caregiving environments.


Asunto(s)
Amígdala del Cerebelo/metabolismo , Animales Recién Nacidos/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Metilación de ADN/fisiología , Ambiente , Hipocampo/metabolismo , Animales , Femenino , Masculino , Ratas
16.
Int J Dev Neurosci ; 31(8): 804-10, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24120634

RESUMEN

Quality of maternal care experienced during infancy is a key factor that can confer vulnerability or resilience to psychiatric disorders later in life. Research continues to indicate that early-life experiences can affect developmental trajectories through epigenetic alterations capable of affecting gene regulation and neural plasticity. Previously, our lab has shown that experiences within an adverse caregiving environment (i.e. maltreatment) produce aberrant DNA methylation patterns at various gene loci in the medial prefrontal cortex (mPFC) of developing and adult rats. This study aimed to determine whether caregiver maltreatment likewise affects expression levels of several genes important in regulating DNA methylation patterns (Dnmt1, Dnmt3a, MeCP2, Gadd45b, and Hdac1). While we observed minimal changes in gene expression within the mPFC of developing rats, we observed expression changes for all genes in adult animals. Specifically, exposure to maltreatment produced a significant decrease in mRNA levels of all epigenetic regulators in adult males and a significant decrease in Gadd45b in adult females. Our results here provide further empirical support for the long-term and sex-specific epigenetic consequences of caregiver maltreatment on the mPFC.


Asunto(s)
Cuidadores , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Corteza Prefrontal/metabolismo , Factores de Edad , Análisis de Varianza , Animales , Animales Recién Nacidos , Antígenos de Diferenciación , Cuidadores/psicología , ADN (Citosina-5-)-Metiltransferasas , ADN Metiltransferasa 3A , Dinaminas , Femenino , Histona Desacetilasa 1 , Masculino , Proteína 2 de Unión a Metil-CpG , Ratas , Ratas Long-Evans
17.
Dev Neurosci ; 35(4): 306-16, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23751776

RESUMEN

Quality of maternal care in infancy is an important contributing factor in the development of behavior and psychopathology. One way maternal care could affect behavioral trajectories is through environmentally induced epigenetic alterations within brain regions known to play prominent roles in cognition, emotion regulation, and stress responsivity. Whereas such research has largely focused on the hippocampus or hypothalamus, the prefrontal cortex (PFC) has only just begun to receive attention. The current study was designed to determine whether exposure to maltreatment or nurturing care is associated with differential methylation of candidate gene loci (bdnf and reelin) within the medial PFC (mPFC) of developing and adult rats. Using a within-litter design, infant male and female rats were exposed to an adverse or nurturing caregiving environment outside their home cage for 30 min per day during the first postnatal week. Additional littermates remained with their biological caregiver within the home cage during the manipulations. We observed that infant rats subjected to caregiver maltreatment emitted more audible and ultrasonic vocalizations than littermates subjected to nurturing care either within or outside of the home cage. While we found no maltreatment-induced changes in bdnf DNA methylation present in infancy, sex-specific alterations were present in the mPFC of adolescents and adults that had been exposed to maltreatment. Furthermore, while maltreated females showed differences in reelin DNA methylation that were transient, males exposed to maltreatment and both males and females exposed to nurturing care outside the home cage showed differences in reelin methylation that emerged by adulthood. Our results demonstrate the ability of infant-caregiver interactions to epigenetically mark genes known to play a prominent role in cognition and psychiatric disorders within the mPFC. Furthermore, our data indicate the remarkable complexity of alterations that can occur, with both transient and later emerging DNA methylation differences that could shape developmental trajectories and underlie gender differences in outcomes.


Asunto(s)
Maltrato a los Niños , Metilación de ADN/genética , Conducta Materna , Corteza Prefrontal/metabolismo , Animales , Animales Recién Nacidos , Conducta Animal/fisiología , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/genética , Moléculas de Adhesión Celular Neuronal/biosíntesis , Moléculas de Adhesión Celular Neuronal/genética , Niño , Epigénesis Genética/fisiología , Proteínas de la Matriz Extracelular/biosíntesis , Proteínas de la Matriz Extracelular/genética , Femenino , Expresión Génica , Humanos , Masculino , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal/genética , Plasticidad Neuronal/fisiología , Corteza Prefrontal/crecimiento & desarrollo , Embarazo , Ratas , Ratas Long-Evans , Proteína Reelina , Serina Endopeptidasas/biosíntesis , Serina Endopeptidasas/genética , Caracteres Sexuales
18.
Wiley Interdiscip Rev Cogn Sci ; 4(1): 105-115, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26304177

RESUMEN

Recent discoveries have associated epigenetic mechanisms, including DNA methylation, histone modifications, and microRNA (miRNA) processing, with activity-dependent changes in gene expression necessary to drive long-term memory formation. Here, we discuss the current interpretation of epigenetic mechanisms in the context of memory and sustained behavioral change. One of the two emerging viewpoints is that epigenetic mechanisms subserve information storage in the central nervous system (CNS), a notion supported by rodent studies of fear, recognition and spatial memories, and stress. The second viewpoint is that epigenetics serves as a mechanism for passing on acquired information across generations, a provocative notion now supported by several lines of work using developing and adult rodents. Continued research on such mechanisms promises to advance our understanding of biological pathways linking experiences to long-term and even multigenerational trajectories in neurobiology and behavior. WIREs Cogn Sci 2013, 4:105-115. doi: 10.1002/wcs.1205 For further resources related to this article, please visit the WIREs website.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...