Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Biochem Cell Biol ; 97(4): 375-379, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30398926

RESUMEN

Mouse models of fetal alcohol spectrum disorders (FASD) have repeatedly identified genes with long-term changes in expression, DNA methylation, noncoding RNA, and histone modifications in response to neurodevelopmental alcohol exposure. Articulation of FASD is achieved via alcohol's effect on gene expression, likely involving epigenetic regulation. The list of genes affected is large and heterogeneous, depending on experimental protocol. We present reanalysis and synthesis of results highlighting the Wnt transcription factor 7 like 2 (Tcf7l2) gene as uniquely compatible with hippocampal DNA methylation, histone modifications, and gene expression changes in a coordinated response to neurodevelopmental alcohol exposure. We data-mined the literature for Tcf7l2 alterations in response to prenatal alcohol exposure. Four studies identified changes in brain Tcf7l2 expression in different FASD models. Further, we performed an in silico TCF7L2 binding site analysis for FASD mouse model data sets. Seven of these published gene lists were significantly enriched for TCF7L2 binding, indicating potential functional relationships. Finally, TCF7L2 is involved in regulation of hundreds of genes, with a role in brain development, myelination, and neuronal function. Tcf7l2 may be involved in neurological defects associated with alcohol exposure via dysregulation of many genes through Wnt signaling. Further functional work is warranted to validate this model for FASD.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Vía de Señalización Wnt , Animales , Ratones , Proteína 2 Similar al Factor de Transcripción 7/genética , Vía de Señalización Wnt/genética
3.
Neurosci Biobehav Rev ; 153: 105350, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37544390

RESUMEN

In numerous brain structures, insulin signaling modulates the homeostatic processes, sensitivity to reward pathways, executive function, memory, and cognition. Through human studies and animal models, mounting evidence implicates central insulin signaling in the metabolic, physiological, and psychological consequences of early life adversity. In this review, we describe the consequences of early life adversity in the brain where insulin signaling is a key factor and how insulin may moderate the effects of adversity on psychiatric and cardio-metabolic health outcomes. Further understanding of how early life adversity and insulin signaling impact specific brain regions and mental and physical health outcomes will assist in prevention, diagnosis, and potential intervention following early life adversity.


Asunto(s)
Experiencias Adversas de la Infancia , Salud Mental , Animales , Humanos , Insulina , Encéfalo , Función Ejecutiva
4.
Front Mol Neurosci ; 14: 671891, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34149355

RESUMEN

Neurodevelopment in humans is a long, elaborate, and highly coordinated process involving three trimesters of prenatal development followed by decades of postnatal development and maturation. Throughout this period, the brain is highly sensitive and responsive to the external environment, which may provide a range of inputs leading to positive or negative outcomes. Fetal alcohol spectrum disorders (FASD) result from prenatal alcohol exposure (PAE). Although the molecular mechanisms of FASD are not fully characterized, they involve alterations to the regulation of gene expression via epigenetic marks. As in the prenatal stages, the postnatal period of neurodevelopment is also sensitive to environmental inputs. Often this sensitivity is reflected in children facing adverse conditions, such as maternal separation. This exposure to early life stress (ELS) is implicated in the manifestation of various behavioral abnormalities. Most FASD research has focused exclusively on the effect of prenatal ethanol exposure in isolation. Here, we review the research into the effect of prenatal ethanol exposure and ELS, with a focus on the continuum of epigenomic and transcriptomic alterations. Interestingly, a select few experiments have assessed the cumulative effect of prenatal alcohol and postnatal maternal separation stress. Regulatory regions of different sets of genes are affected by both treatments independently, and a unique set of genes are affected by the combination of treatments. Notably, epigenetic and gene expression changes converge at the clustered protocadherin locus and oxidative stress pathway. Functional studies using epigenetic editing may elucidate individual contributions of regulatory regions for hub genes and further profiling efforts may lead to the development of non-invasive methods to identify children at risk. Taken together, the results favor the potential to improve neurodevelopmental outcomes by epigenetic management of children born with FASD using favorable postnatal conditions with or without therapeutic interventions.

5.
Front Genet ; 11: 70, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32174962

RESUMEN

Fetal alcohol spectrum disorder (FASD) is characterized by developmental and behavioral deficits caused by maternal drinking during pregnancy. Children born with FASD often face additional stresses, including maternal separation, that add yet additional deficits. The mechanism associated with this interaction is not known. We have used a mouse model for prenatal ethanol exposure and maternal separation to demonstrate that the combination of the two treatments results in more than additive deficits. Furthermore, the behavioral deficits are associated with changes in hippocampal gene expression that persist into adulthood. What initiates and maintains these changes remains to be established and forms the focus of this report. Specifically, MeDIP-Seq was used to assess if changes in promoter DNA methylation are affected by exposure to prenatal ethanol and maternal separation including its relationship to gene expression. The novel results show that different sets of genes implicated by promoter DNA methylation are affected by both treatments independently, and a relatively unique set of genes are affected by the combination of the two treatments. Prenatal ethanol exposure leads to altered promoter DNA methylation at genes important for transcriptional regulation. Maternal separation leads to changes at genes important for histone methylation and immune response, and the combination of two treatments results in DNA methylation changes at genes important for neuronal migration and immune response. Our dual results from the same hippocampal samples suggest there is minimal complementarity between changes in promoter DNA methylation and gene expression, although genes involved tend to be critical for brain development and function. While remaining to be validated, such results argue that mechanisms beyond promoter DNA methylation must be involved in lasting gene expression alterations leading to behavioral deficits implicated in FASD. They may facilitate early and reliable diagnosis, as well as novel strategies for the amelioration of FASD-related deficits.

6.
J Neurodev Disord ; 12(1): 15, 2020 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-32416732

RESUMEN

BACKGROUND: Fetal alcohol spectrum disorders (FASD) are common, seen in 1-5% of the population in the USA and Canada. Children diagnosed with FASD are not likely to remain with their biological parents, facing early maternal separation and foster placements throughout childhood. METHODS: We model FASD in mice via prenatal alcohol exposure and further induce early life stress through maternal separation. We use RNA-seq followed by clustering of expression profiles through weighted gene co-expression network analysis (WGCNA) to analyze transcriptomic changes that result from the treatments. We use reverse transcription qPCR to validate these changes in the mouse hippocampus. RESULTS: We report an association between adult hippocampal gene expression and prenatal ethanol exposure followed by postnatal separation stress that is related to behavioral changes. Expression profile clustering using WGCNA identifies a set of transcripts, module 19, associated with anxiety-like behavior (r = 0.79, p = 0.002) as well as treatment group (r = 0.68, p = 0.015). Genes in this module are overrepresented by genes involved in transcriptional regulation and other pathways related to neurodevelopment. Interestingly, one member of this module, Polr2a, polymerase (RNA) II (DNA directed) polypeptide A, is downregulated by the combination of prenatal ethanol and postnatal stress in an RNA-Seq experiment and qPCR validation (q = 2e-12, p = 0.004, respectively). CONCLUSIONS: Together, transcriptional control in the hippocampus is implicated as a potential underlying mechanism leading to anxiety-like behavior via environmental insults. Further research is required to elucidate the mechanism involved and use this insight towards early diagnosis and amelioration strategies involving children born with FASD.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal/genética , Perfilación de la Expresión Génica , Hipocampo/metabolismo , Privación Materna , Procesamiento Postranscripcional del ARN , Consumo de Bebidas Alcohólicas/genética , Animales , Animales Recién Nacidos , Ansiedad , Canadá , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Etanol/metabolismo , Femenino , Trastornos del Espectro Alcohólico Fetal/metabolismo , Regulación de la Expresión Génica , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Efectos Tardíos de la Exposición Prenatal/metabolismo , Transcriptoma
7.
Behav Brain Res ; 308: 94-103, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27102339

RESUMEN

Prenatal alcohol exposure (PAE) can result in fetal alcohol spectrum disorder (FASD), characterized by developmental disability. As children with FASD are often raised in suboptimal conditions, we have investigated the combination of PAE via maternal preference consumption of 10% ethanol in water with early life stress (ELS) via daily 3h maternal separation and isolation. Our results focus on development and behavioral features, including activity, anxiety-like behavior, as well as learning and memory. PAE influenced the number of pups surviving to postnatal day 2 and 70, with fewer surviving pups associated with the severity of ethanol exposure. PAE and ELS both had effects on pup weight at postnatal day 21, with amount of ethanol exposure positively correlating with pup weight. We found females were more active than males in a novel open field environment, but not following PAE. In addition, PAE resulted in overall increased exploratory behavior in the open field. Further, PAE and ELS both resulted in overnight hypoactivity in a home cage environment, as well as learning deficits that were influenced by sex in the Barnes Maze for learning and memory. These results are attributed to environmental interactions involving PAE and ELS.


Asunto(s)
Ansiedad/etiología , Síntomas Conductuales/etiología , Discapacidades del Desarrollo/etiología , Trastornos del Espectro Alcohólico Fetal/fisiopatología , Privación Materna , Análisis de Varianza , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Etanol/toxicidad , Conducta Exploratoria , Femenino , Relaciones Interpersonales , Tamaño de la Camada , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Comportamiento de Nidificación
8.
PLoS One ; 11(5): e0154836, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27136348

RESUMEN

The molecular basis of Fetal Alcohol Spectrum Disorders (FASD) is poorly understood; however, epigenetic and gene expression changes have been implicated. We have developed a mouse model of FASD characterized by learning and memory impairment and persistent gene expression changes. Epigenetic marks may maintain expression changes over a mouse's lifetime, an area few have explored. Here, mice were injected with saline or ethanol on postnatal days four and seven. At 70 days of age gene expression microarray, methylated DNA immunoprecipitation microarray, H3K4me3 and H3K27me3 chromatin immunoprecipitation microarray were performed. Following extensive pathway analysis of the affected genes, we identified the top affected gene expression pathway as "Free radical scavenging". We confirmed six of these changes by droplet digital PCR including the caspase Casp3 and Wnt transcription factor Tcf7l2. The top pathway for all methylation-affected genes was "Peroxisome biogenesis"; we confirmed differential DNA methylation in the Acca1 thiolase promoter. Altered methylation and gene expression in oxidative stress pathways in the adult hippocampus suggests a novel interface between epigenetic and oxidative stress mechanisms in FASD.


Asunto(s)
Metilación de ADN/genética , Etanol/toxicidad , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Histonas/metabolismo , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Femenino , Radicales Libres/metabolismo , Regulación del Desarrollo de la Expresión Génica , Metilación/efectos de los fármacos , Ratones , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Embarazo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
9.
Psychiatr Genet ; 26(6): 229-257, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27606929

RESUMEN

The XXIIIrd World Congress of Psychiatric Genetics meeting, sponsored by the International Society of Psychiatric Genetics, was held in Toronto, ON, Canada, on 16-20 October 2015. Approximately 700 participants attended to discuss the latest state-of-the-art findings in this rapidly advancing and evolving field. The following report was written by trainee travel awardees. Each was assigned one session as a rapporteur. This manuscript represents the highlights and topics that were covered in the plenary sessions, symposia, and oral sessions during the conference, and contains major notable and new findings.


Asunto(s)
Trastornos Mentales/genética , Estudio de Asociación del Genoma Completo , Humanos , Salud Mental
10.
Front Pediatr ; 2: 90, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25207264

RESUMEN

Brain development in mammals is long lasting. It begins early during embryonic growth and is finalized in early adulthood. This progression represents a delicate choreography of molecular, cellular, and physiological processes initiated and directed by the fetal genotype in close interaction with environment. Not surprisingly, most aberrations in brain functioning including intellectual disability (ID) are attributed to either gene(s), or environment or the interaction of the two. The ensuing complexity has made the assessment of this choreography, ever challenging. A model to assess this complexity has used a mouse model (C57BL/6J or B6) that is subjected to prenatal alcohol exposure. The resulting pups show learning and memory deficits similar to patients with fetal alcohol spectrum disorder (FASD), which is associated with life-long changes in gene expression. Interestingly, this change in gene expression underlies epigenetic processes including DNA methylation and miRNAs. This paradigm is applicable to ethanol exposure at different developmental times (binge at trimesters 1, 2, and 3 as well as continuous preference drinking (70%) of 10% alcohol by B6 females during pregnancy). The exposure leads to life-long changes in neural epigenetic marks, gene expression, and a variety of defects in neurodevelopment and CNS function. We argue that this cascade may be reversed postnatally via drugs, chemicals, and environment including maternal care. Such conclusions are supported by two sets of results. First, antipsychotic drugs that are used to treat ID including psychosis function via changes in DNA methylation, a major epigenetic mark. Second, post-natal environment may improve (with enriched environments) or worsen (with negative and maternal separation stress) the cognitive ability of pups that were prenatally exposed to ethanol as well as their matched controls. In this review, we will discuss operational epigenetic mechanisms involved in the development of intellectual ability/disability in response to alcohol during prenatal or post-natal development. In doing so, we will explore the potential of epigenetic manipulation in the treatment of FASD and related disorders implicated in ID.

11.
Front Genet ; 5: 161, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24917881

RESUMEN

There is abundant evidence that prenatal alcohol exposure leads to a range of behavioral and cognitive impairments, categorized under the term fetal alcohol spectrum disorders (FASDs). These disorders are pervasive in Western cultures and represent the most common preventable source of neurodevelopmental disabilities. The genetic and epigenetic etiology of these phenotypes, including those factors that may maintain these phenotypes throughout the lifetime of an affected individual, has become a recent topic of investigation. This review integrates recent data that has progressed our understanding FASD as a continuum of molecular events, beginning with cellular stress response and ending with a long-term "footprint" of epigenetic dysregulation across the genome. It reports on data from multiple ethanol-treatment paradigms in mouse models that identify changes in gene expression that occur with respect to neurodevelopmental timing of exposure and ethanol dose. These studies have identified patterns of genomic alteration that are dependent on the biological processes occurring at the time of ethanol exposure. This review also adds to evidence that epigenetic processes such as DNA methylation, histone modifications, and non-coding RNA regulation may underlie long-term changes to gene expression patterns. These may be initiated by ethanol-induced alterations to DNA and histone methylation, particularly in imprinted regions of the genome, affecting transcription which is further fine-tuned by altered microRNA expression. These processes are likely complex, genome-wide, and interrelated. The proposed model suggests a potential for intervention, given that epigenetic changes are malleable and may be altered by postnatal environment. This review accentuates the value of mouse models in deciphering the molecular etiology of FASD, including those processes that may provide a target for the ammelioration of this common yet entirely preventable disorder.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA