Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Child Psychiatry Hum Dev ; 54(3): 672-682, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-34727279

RESUMEN

Shorter telomeres mark cellular aging and are linked to chronic stress exposure as well as negative physical and psychological outcomes. However, it is unclear whether telomere length mediates associations between early stress exposure and later externalizing problems, or whether boys and girls differ in pathways to these concerns. We therefore examined associations between telomere length, early stress via negative caregiving, and children's externalizing symptom development over time in 409 three-year-old children and their parents. Telomere length mediated the association between early parental intrusiveness and later rule-breaking behavior; however, this association was moderated by children's biological sex such that parent intrusiveness was related only to boys' rule-breaking. Findings support the notion that children's telomere length may mark individual differences in responses to negative early caregiving, and highlight a potential mechanism contributing to the development of rule-breaking problems in boys.


Asunto(s)
Actuación (Psicología) , Conducta Infantil , Responsabilidad Parental , Padres , Telómero , Telómero/metabolismo , Padres/psicología , Conducta Infantil/psicología , Responsabilidad Parental/psicología , Humanos , Masculino , Femenino , Preescolar , Niño , Experiencias Adversas de la Infancia/psicología , Caracteres Sexuales , Estrés Psicológico/psicología , Adulto , Atención , Agresión , Trastornos de la Conducta Infantil/psicología
2.
Science ; 360(6386): 327-331, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29674594

RESUMEN

The genetic basis of autism spectrum disorder (ASD) is known to consist of contributions from de novo mutations in variant-intolerant genes. We hypothesize that rare inherited structural variants in cis-regulatory elements (CRE-SVs) of these genes also contribute to ASD. We investigated this by assessing the evidence for natural selection and transmission distortion of CRE-SVs in whole genomes of 9274 subjects from 2600 families affected by ASD. In a discovery cohort of 829 families, structural variants were depleted within promoters and untranslated regions, and paternally inherited CRE-SVs were preferentially transmitted to affected offspring and not to their unaffected siblings. The association of paternal CRE-SVs was replicated in an independent sample of 1771 families. Our results suggest that rare inherited noncoding variants predispose children to ASD, with differing contributions from each parent.


Asunto(s)
Trastorno del Espectro Autista/genética , Predisposición Genética a la Enfermedad , Variación Genética , Herencia Paterna , Regiones Promotoras Genéticas/genética , Exones , Regulación de la Expresión Génica , Genoma Humano , Humanos , Mutación , Linaje , ARN no Traducido/genética , Selección Genética , Eliminación de Secuencia , Factores de Transcripción/genética
3.
Dev Neurosci ; 36(6): 499-519, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25278313

RESUMEN

The developing brain is remarkably sensitive to alcohol exposure, resulting in the wide range of cognitive and neurobehavioral characteristics categorized under the term fetal alcohol spectrum disorders (FASD). The brain is particularly susceptible to alcohol during synaptogenesis, a process that occurs heavily during the third trimester and is characterized by the establishment and pruning of neural circuitry; however, the molecular response of the brain to ethanol during synaptogenesis has not been documented. To model a binge-like exposure during the third-trimester neurodevelopmental equivalent, neonate mice were given a high (5 g/kg over 2 h) dose of ethanol at postnatal day 7. Acute transcript changes within the brain were assessed using expression arrays and analyzed for associations with gene ontology functional categories, canonical pathways, and gene network interactions. The short-term effect of ethanol was characterized by an acute stress response and a downregulation of energetically costly cellular processes. Further, alterations to a number of genes with roles in synaptic transmission and hormonal signaling, particularly those associated with the neuroendocrine development and function, were evident. Ethanol exposure during synaptogenesis was also associated with altered histone deacetylase and microRNA transcript levels, suggesting that abnormal epigenetic patterning may maintain some of the persistent molecular consequences of developmental ethanol exposure. The results shed insight into the sensitivity of the brain to ethanol during the third-trimester equivalent and outline how ethanol-induced alterations to genes associated with neural connectivity may contribute to FASD phenotypes.


Asunto(s)
Encéfalo/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Etanol/efectos adversos , Trastornos del Espectro Alcohólico Fetal/metabolismo , Expresión Génica/efectos de los fármacos , Transducción de Señal/genética , Estrés Fisiológico , Transmisión Sináptica/genética , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Etanol/administración & dosificación , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL
4.
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.

5.
Clin Epigenetics ; 5(1): 14, 2013 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-23915435

RESUMEN

BACKGROUND: Prenatal alcohol exposure is known to result in fetal alcohol spectrum disorders, a continuum of physiological, behavioural, and cognitive phenotypes that include increased risk for anxiety and learning-associated disorders. Prenatal alcohol exposure results in life-long disorders that may manifest in part through the induction of long-term gene expression changes, potentially maintained through epigenetic mechanisms. FINDINGS: Here we report a decrease in the expression of Canabinoid receptor 1 (Cnr1) and an increase in the expression of the regulatory microRNA miR-26b in the brains of adult mice exposed to ethanol during neurodevelopment. Furthermore, we show that miR-26b has significant complementarity to the 3'-UTR of the Cnr1 transcript, giving it the potential to bind and reduce the level of Cnr1 expression. CONCLUSIONS: These findings elucidate a mechanism through which some genes show long-term altered expression following prenatal alcohol exposure, leading to persistent alterations to cognitive function and behavioural phenotypes observed in fetal alcohol spectrum disorders.

6.
Dis Model Mech ; 6(4): 977-92, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23580197

RESUMEN

Fetal alcohol spectrum disorders (FASDs) are characterized by life-long changes in gene expression, neurodevelopment and behavior. What mechanisms initiate and maintain these changes are not known, but current research suggests a role for alcohol-induced epigenetic changes. In this study we assessed alterations to adult mouse brain tissue by assaying DNA cytosine methylation and small noncoding RNA (ncRNA) expression, specifically the microRNA (miRNA) and small nucleolar RNA (snoRNA) subtypes. We found long-lasting alterations in DNA methylation as a result of fetal alcohol exposure, specifically in the imprinted regions of the genome harboring ncRNAs and sequences interacting with regulatory proteins. A large number of major nodes from the identified networks, such as Pten signaling, contained transcriptional repressor CTCF-binding sites in their promoters, illustrating the functional consequences of alcohol-induced changes to DNA methylation. Next, we assessed ncRNA expression using two independent array platforms and quantitative PCR. The results identified 34 genes that are targeted by the deregulated miRNAs. Of these, four (Pten, Nmnat1, Slitrk2 and Otx2) were viewed as being crucial in the context of FASDs given their roles in the brain. Furthermore, ≈ 20% of the altered ncRNAs mapped to three imprinted regions (Snrpn-Ube3a, Dlk1-Dio3 and Sfmbt2) that showed differential methylation and have been previously implicated in neurodevelopmental disorders. The findings of this study help to expand on the mechanisms behind the long-lasting changes in the brain transcriptome of FASD individuals. The observed changes could contribute to the initiation and maintenance of the long-lasting effect of alcohol.


Asunto(s)
Metilación de ADN/genética , Trastornos del Espectro Alcohólico Fetal/genética , ARN no Traducido/genética , Envejecimiento/genética , Animales , Sitios de Unión/genética , Encéfalo/metabolismo , Encéfalo/patología , Factor de Unión a CCCTC , Análisis por Conglomerados , Biología Computacional , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes/genética , Genoma/genética , Masculino , Exposición Materna/efectos adversos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Modelos Genéticos , Embarazo , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , ARN no Traducido/metabolismo , Proteínas Represoras/metabolismo , Reproducibilidad de los Resultados , Factores de Tiempo
7.
J Neurodev Disord ; 5(1): 6, 2013 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-23497526

RESUMEN

BACKGROUND: Maternal alcohol consumption is known to adversely affect fetal neurodevelopment. While it is known that alcohol dose and timing play a role in the cognitive and behavioral changes associated with prenatal alcohol exposure, it is unclear what developmental processes are disrupted that may lead to these phenotypes. METHODS: Mice (n=6 per treatment per developmental time) were exposed to two acute doses of alcohol (5 g/kg) at neurodevelopmental times representing the human first, second, or third trimester equivalent. Mice were reared to adulthood and changes to their adult brain transcriptome were assessed using expression arrays. These were then categorized based on Gene Ontology annotations, canonical pathway associations, and relationships to interacting molecules. RESULTS: The results suggest that ethanol disrupts biological processes that are actively occurring at the time of exposure. These include cell proliferation during trimester one, cell migration and differentiation during trimester two, and cellular communication and neurotransmission during trimester three. Further, although ethanol altered a distinct set of genes depending on developmental timing, many of these show interrelatedness and can be associated with one another via 'hub' molecules and pathways such as those related to huntingtin and brain-derived neurotrophic factor. CONCLUSIONS: These changes to brain gene expression represent a 'molecular footprint' of neurodevelopmental alcohol exposure that is long-lasting and correlates with active processes disrupted at the time of exposure. This study provides further support that there is no neurodevelopmental time when alcohol cannot adversely affect the developing brain.

8.
Brain Res ; 1458: 18-33, 2012 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-22560501

RESUMEN

Many women continue to consume low to moderate quantities of alcohol during pregnancy, which can result in the variable neurobehavioural effects in the absence of physiological abnormalities that characterize fetal alcohol spectrum disorders (FASD). Previously, we reported that a mouse model for FASD based on voluntary maternal ethanol consumption throughout gestation resulted in offspring that showed mild developmental delay, anxiety-related traits, and deficits in spatial learning. Here, we extend this model by evaluating the gene expression changes that occur in the adult brain of C57BL/6J mice prenatally exposed to ethanol via maternal preference drinking. The results of two independent expression array experiments indicate that ethanol induces subtle but consistent changes to global gene expression. Gene enrichment analysis showed over-represented gene ontology classifications of cellular, embryonic, and nervous system development. Molecular network analysis supported these classifications, with significant networks related to cellular and tissue development, free radical scavenging, and small molecule metabolism. Further, a number of genes identified have previously been implicated in FASD-relevant neurobehavioural phenotypes such as cognitive function (Ache, Bcl2, Cul4b, Dkc1, Ebp, Lcat, Nsdh1, Sstr3), anxiety (Bcl2), attention deficit hyperactivity disorder (Nsdh1), and mood disorders (Bcl2, Otx2, Sstr3). The results suggest a complex residual "footprint" of neurodevelopmental ethanol exposure that may provide a new perspective for identifying mechanisms that underlie the life-long persistence of FASD-related cognitive and behavioural alterations, including potential targets for treatment.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Química Encefálica/efectos de los fármacos , Química Encefálica/genética , Modelos Animales de Enfermedad , Trastornos del Espectro Alcohólico Fetal/genética , Efectos Tardíos de la Exposición Prenatal/genética , Transcriptoma/efectos de los fármacos , Consumo de Bebidas Alcohólicas/metabolismo , Consumo de Bebidas Alcohólicas/patología , Animales , Animales Recién Nacidos , Etanol/administración & dosificación , Etanol/efectos adversos , Femenino , Trastornos del Espectro Alcohólico Fetal/metabolismo , Redes Reguladoras de Genes/efectos de los fármacos , Redes Reguladoras de Genes/fisiología , Masculino , Conducta Materna/efectos de los fármacos , Conducta Materna/fisiología , Ratones , Ratones Endogámicos C57BL , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Factores de Tiempo , Transcriptoma/genética
9.
Behav Brain Res ; 223(2): 376-87, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21601595

RESUMEN

Fetal alcohol spectrum disorders (FASD) remain the most common preventable cause of behavioural abnormalities and cognitive deficits, yet little is known about the biological mechanisms involved in FASD pathology. Maternal voluntary ethanol consumption in mice may be a useful model for establishing the biological basis of moderate ethanol exposure phenotypes, which make up the majority of FASD cases. We have employed a two-bottle choice paradigm of maternal ethanol consumption throughout gestation and the early postnatal period in C57BL/6J mice. We assessed the efficacy of this model to produce a range of FASD-relevant phenotypes and evaluated gene expression changes in the adult offspring. Results showed stable maternal consumption and lack of maternal care differences between ethanol-consuming and water-only dams. Ethanol-exposed offspring showed delays in neonatal reflex and coordination development. Further, ethanol-exposed adolescent mice showed decreased activity in a novel environment that appeared to be the result of novelty-induced anxiety, and acquisition learning deficits. Evaluation of the neurotransmitter-associated genes Gabra6, Glra1, and Grin2c revealed significant down-regulation of Glra1 and Grin2c in the brains of ethanol-exposed young adult males. These results suggest that this model is able to produce a range of behavioural phenotypes consistent with prenatal ethanol exposure and may be used to evaluate resulting long-term genetic changes. Given the range of genetic resources available for inbred mouse strains, the model described here may prove to be a useful tool in evaluating the molecular basis of FASD.


Asunto(s)
Consumo de Bebidas Alcohólicas/patología , Trastornos del Espectro Alcohólico Fetal/patología , Animales , Animales Recién Nacidos , Química Encefálica/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Fuerza de la Mano/fisiología , Masculino , Conducta Materna , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Actividad Motora/fisiología , Sistema Nervioso/crecimiento & desarrollo , Equilibrio Postural/efectos de los fármacos , Embarazo , ARN/biosíntesis , ARN/genética , Receptores de GABA-A/genética , Receptores de Glicina/biosíntesis , Receptores de Glicina/genética , Receptores de N-Metil-D-Aspartato/genética , Reflejo/fisiología , Reflejo de Sobresalto/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sobrevida , Aumento de Peso/efectos de los fármacos
10.
Behav Genet ; 40(5): 660-71, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20496163

RESUMEN

The genetic factors that increase risk for alcohol and nicotine addiction have been elusive, although the frequent co-abuse of these drugs suggests they may act on a common biological pathway. A site of action for both nicotine and alcohol effects in the brain are neuronal nicotinic acetylcholine receptors (nAChR). This report explores the association between six nAChR subunit genes (Chrna3, Chrna4, Chrnb4, Chrnb2, Chrna5, and Chrna7) with alcohol preference (AP) using co-segregation of AP with nAChR subunit genotypes in a F(2) population produced from reciprocal crosses of alcohol-preferring C57BL/6J (B6) and alcohol-avoiding DBA/2J (D2) strains of mice. Polymorphisms located within the Chrna5-Chrna3-Chrnb4 cluster on mouse chromosome 9 were found to co-segregate with AP, with high-drinking F(2) mice carrying B6 alleles and low-drinking F(2) mice carrying D2 alleles. Further, the Chrnb4 and Chrna5 genes showed expression differences between B6 and D2 mice, which is compatible with their involvement in AP in mice and, potentially, alcohol abuse in humans.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Proteínas del Tejido Nervioso/genética , Receptores Nicotínicos/genética , Consumo de Bebidas Alcohólicas/metabolismo , Animales , Encéfalo/metabolismo , Cromosomas de los Mamíferos/genética , Cruzamientos Genéticos , Etanol/farmacología , Femenino , Genotipo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Polimorfismo Genético , Receptor Nicotínico de Acetilcolina alfa 7
11.
Genomics ; 93(4): 358-66, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19101625

RESUMEN

The Spsb family of genes encode well-conserved proteins of unknown function. Mammalian Spsb genes are likely the result of three separate duplication and divergence events during vertebrate evolution. The phylogenetic relationship along with expression and regulation of Spsb genes may offer insight into the evolution and function of this gene family in vertebrates. We have established that Spsb genes are expressed in numerous tissues, however their pattern and level of expression is tissue-dependent. Further, only Spsb1 is responsive to stress caused by ethanol exposure in the mouse brain, which suggests that Spsb genes have acquired different regulatory mechanisms. Analysis of cis-regulatory elements supports this, but also reveals some common regulatory modules involved in cell proliferation and stress response. Our results contribute to the growing body of data on the expression and function of Spsb genes, which serve as a model for studies on the origin, divergence and specialization of eukaryotic gene families.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica , Variación Genética , Familia de Multigenes , Proteínas Supresoras de la Señalización de Citocinas/genética , Secuencia de Aminoácidos , Animales , Evolución Molecular , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Datos de Secuencia Molecular , Filogenia , Regiones Promotoras Genéticas , Canal Liberador de Calcio Receptor de Rianodina/química , Proteínas Supresoras de la Señalización de Citocinas/química , Proteínas Supresoras de la Señalización de Citocinas/clasificación , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factores de Transcripción/metabolismo
12.
Genome ; 50(10): 877-97, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18059552

RESUMEN

Most familial behavioral phenotypes result from the complex interaction of multiple genes. Studies of such phenotypes involving human subjects are often inconclusive owing to complexity of causation and experimental limitations. Studies of animal models argue for the use of established genetic strains as a powerful tool for genetic dissection of behavioral disorders and have led to the identification of rare genes and genetic mechanisms implicated in such phenotypes. We have used microarrays to study global gene expression in adult brains of four genetic strains of mice (C57BL/6J, DBA/2J, A/J, and BALB/c). Our results demonstrate that different strains show expression differences for a number of genes in the brain, and that closely related strains have similar patterns of gene expression as compared with distantly related strains. In addition, among the 24 000 genes and ESTs on the microarray, 77 showed at least a 1.5-fold increase in the brains of C57BL/6J mice as compared with those of DBA/2J mice. These genes fall into such functional categories as gene regulation, metabolism, cell signaling, neurotransmitter transport, and DNA/RNA binding. The importance of these findings as a novel genetic resource and their use and application in the genetic analysis of complex behavioral phenotypes, susceptibilities, and responses to drugs and chemicals are discussed.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Conducta , Expresión Génica , Genómica , Ratones , Modelos Animales , Animales , Encéfalo/metabolismo , Perfilación de la Expresión Génica , Humanos , Ratones Endogámicos , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...