Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 78(6): 3045-3055, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33313982

RESUMEN

Excess maternal fat intake and obesity increase offspring susceptibility to conditions such as chronic anxiety and substance abuse. We hypothesised that environmentally modulated DNA methylation changes (5mC/5hmC) in regulatory regions of the genome that modulate mood and consumptive behaviours could contribute to susceptibility to these conditions. We explored the effects of environmental factors on 5mC/5hmC levels within the GAL5.1 enhancer that controls anxiety-related behaviours and alcohol intake. We first observed that 5mC/5hmC levels within the GAL5.1 enhancer differed significantly in different parts of the brain. Moreover, we noted that early life stress had no significant effect of 5mC/5hmC levels within GAL5.1. In contrast, we identified that allowing access of pregnant mothers to high-fat diet (> 60% calories from fat) had a significant effect on 5mC/5hmC levels within GAL5.1 in hypothalamus and amygdala of resulting male offspring. Cell transfection-based studies using GAL5.1 reporter plasmids showed that 5mC has a significant repressive effect on GAL5.1 activity and its response to known stimuli, such as EGR1 transcription factor expression and PKC agonism. Intriguingly, CRISPR-driven disruption of GAL5.1 from the mouse genome, although having negligible effects on metabolism or general appetite, significantly decreased intake of high-fat diet suggesting that GAL5.1, in addition to being epigenetically modulated by high-fat diet, also actively contributes to the consumption of high-fat diet suggesting its involvement in an environmentally influenced regulatory loop. Furthermore, considering that GAL5.1 also controls alcohol preference and anxiety these studies may provide a first glimpse into an epigenetically controlled mechanism that links maternal high-fat diet with transgenerational susceptibility to alcohol abuse and anxiety.


Asunto(s)
Alcoholismo/patología , Ansiedad/patología , Dieta Alta en Grasa , Elementos de Facilitación Genéticos/genética , 5-Metilcitosina/metabolismo , Alcoholismo/genética , Amígdala del Cerebelo/metabolismo , Animales , Ansiedad/genética , Línea Celular Tumoral , Metilación de ADN , Modelos Animales de Enfermedad , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Epigénesis Genética , Femenino , Humanos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa C/química , Proteína Quinasa C/metabolismo
2.
Nutr Neurosci ; 23(4): 321-334, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30032721

RESUMEN

A high-fat diet induces hypothalamic inflammation in rodents which, in turn, contributes to the development of obesity by eliciting both insulin and leptin resistance. However, the mechanism by which long-chain saturated fatty acids trigger inflammation is still contentious. To elucidate this mechanism, the effect of fatty acids on the expression of the pro-inflammatory cytokines IL-6 and TNFα was investigated in the mHypoE-N42 hypothalamic cell line (N42). N42 cells were treated with lauric acid (LA) and palmitic acid (PA). PA challenge was carried out in the presence of either a TLR4 inhibitor, a ceramide synthesis inhibitor (L-cycloserine), oleic acid (OA) or eicosapentaenoic acid (EPA). Intracellular ceramide accumulation was quantified using LC-ESI-MS/MS. PA but not LA upregulated IL-6 and TNFα. L-cycloserine, OA and EPA all counteracted PA-induced intracellular ceramide accumulation leading to a downregulation of IL-6 and TNFα. However, a TLR4 inhibitor failed to inhibit PA-induced upregulation of pro-inflammatory cytokines.In conclusion, PA induced the expression of IL-6 and TNFα in N42 neuronal cells independently of TLR4 but, partially, via ceramide synthesis with OA and EPA being anti-inflammatory by decreasing PA-induced intracellular ceramide build-up. Thus, ceramide accumulation represents one on the mechanisms by which PA induces inflammation in neurons.


Asunto(s)
Ceramidas/biosíntesis , Encefalitis/metabolismo , Hipotálamo/metabolismo , Ácido Palmítico/administración & dosificación , Ácido Palmítico/metabolismo , Receptor Toll-Like 4/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular , Encefalitis/inducido químicamente , Hipotálamo/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Ratas Sprague-Dawley
3.
Psychoneuroendocrinology ; 109: 104407, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31445429

RESUMEN

The cannabinoid-1 receptor (CB1) plays a critical role in a number of biological processes including nutrient intake, addiction and anxiety-related behaviour. Numerous studies have shown that expression of the gene encoding CB1 (CNR1) is highly dynamic with changes in the tissue specific expression of CNR1 associated with brain homeostasis and disease progression. However, little is known of the mechanisms regulating this dynamic expression. To gain a better understanding of the genomic mechanisms modulating the expression of CNR1 in health and disease we characterised the role of a highly conserved regulatory sequence (ECR1) in CNR1 intron 2 that contained a polymorphism in linkage disequilibrium with disease associated SNPs. We used CRISPR/CAS9 technology to disrupt ECR1 within the mouse genome. Disruption of ECR1 significantly reduced CNR1 expression in the hippocampus but not in the hypothalamus. These mice also displayed an altered sex-specific anxiety-related behavioural profile (open field test), reduced ethanol intake and a reduced hypothermic response following CB1 agonism. However, no significant changes in feeding patterns were detected. These data suggest that, whilst not all of the expression of CNR1 is modulated by ECR1, this highly conserved enhancer is required for appropriate physiological responses to a number of stimuli. The combination of comparative genomics and CRISPR/CAS9 disruption used in our study to determine the functional effects of genetic and epigenetic changes on the activity of tissue-specific regulatory elements at the CNR1 locus represent an important first step in gaining a mechanistic understanding of cannabinoid regulatory pharmacogenetics.


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Conducta Adictiva/genética , Receptor Cannabinoide CB1/genética , Animales , Ansiedad/genética , Trastornos de Ansiedad/genética , Encéfalo/metabolismo , Cannabinoides/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Genotipo , Hipocampo/metabolismo , Hipotálamo/metabolismo , Intrones/genética , Desequilibrio de Ligamiento/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple/genética , Receptor Cannabinoide CB1/metabolismo
4.
J Biol Chem ; 287(16): 12828-34, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22362764

RESUMEN

Polymorphisms within intron 2 of the CNR1 gene, which encodes cannabinoid receptor 1 (CB(1)), have been associated with addiction, obesity, and brain volume deficits. We used comparative genomics to identify a polymorphic (rs9444584-C/T) sequence (ECR1) in intron 2 of the CNR1 gene that had been conserved for 310 million years. The C-allele of ECR1 (ECR1(C)) acted as an enhancer in hypothalamic and dorsal root ganglia cells and responded to MAPK activation through the MEKK pathway but not in hippocampal cells. However, ECR1(T) was significantly more active in hypothalamic and dorsal root ganglia cells but, significantly, and in contrast to ECR1(C), was highly active in hippocampal cells where it also responded strongly to activation of MAPK. Intriguingly, rs9444584 is in strong linkage disequilibrium with two other SNPs (rs9450898 (r(2) = 0.841) and rs2023239 (r(2) = 0.920)) that have been associated with addiction, obesity (rs2023239), and reduced fronto-temporal white matter volumes in schizophrenia patients as a result of cannabis misuse (rs9450898). Considering their high linkage disequilibrium and the increased response of ECR1(T) to MAPK signaling when compared with ECR1(C), it is possible that the functional effects of the different alleles of rs9444584 may play a role in the conditions associated with rs9450898 and rs2023239. Further analysis of the different alleles of ECR1 may lead to a greater understanding of the role of CNR1 gene misregulation in these conditions as well as chronic inflammatory pain.


Asunto(s)
Ganglios Espinales/fisiología , Hipocampo/fisiología , Hipotálamo/fisiología , Receptor Cannabinoide CB1/genética , Esquizofrenia/genética , Alelos , Animales , Secuencia de Bases , Pollos , Dolor Crónico/genética , Secuencia Conservada , Elementos de Facilitación Genéticos/genética , Ganglios Espinales/citología , Hipocampo/citología , Humanos , Hipotálamo/citología , Intrones/genética , Desequilibrio de Ligamiento , Sistema de Señalización de MAP Quinasas/genética , Datos de Secuencia Molecular , Obesidad/genética , Polimorfismo de Nucleótido Simple/genética , Cultivo Primario de Células , Ratas , Especificidad de la Especie
5.
Neuropsychopharmacology ; 36(11): 2211-21, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21716262

RESUMEN

The expression of the galanin gene (GAL) in the paraventricular nucleus (PVN) and in the amygdala of higher vertebrates suggests the requirement for highly conserved, but unidentified, regulatory sequences that are critical to allow the galanin gene to control alcohol and fat intake and modulate mood. We used comparative genomics to identify a highly conserved sequence that lay 42 kb 5' of the human GAL transcriptional start site that we called GAL5.1. GAL5.1 activated promoter activity in neurones of the PVN, arcuate nucleus and amygdala that also expressed the galanin peptide. Analysis in neuroblastoma cells demonstrated that GAL5.1 acted as an enhancer of promoter activity after PKC activation. GAL5.1 contained two polymorphisms; rs2513280(C/G) and rs2513281(A/G), that occurred in two allelic combinations (GG or CA) where the dominant GG alelle occurred in 70-83 % of the human population. Intriguingly, both SNPs were found to be in LD (R(2) of 0.687) with another SNP (rs2156464) previously associated with major depressive disorder (MDD). Recreation of these alleles in reporter constructs and subsequent magnetofection into primary rat hypothalamic neurones showed that the CA allele was 40 % less active than the GG allele. This is consistent with the hypothesis that the weaker allele may affect food and alcohol preference. The linkage of the SNPs analysed in this study with a SNP previously associated with MDD together with the functioning of GAL5.1 as a PVN and amygdala specific enhancer represent a significant advance in our ability to understand alcoholism, obesity and major depressive disorder.


Asunto(s)
Alcoholismo/genética , Amígdala del Cerebelo/metabolismo , Depresión/genética , Galanina/genética , Hipotálamo/metabolismo , Obesidad/genética , Animales , Secuencia de Bases , Línea Celular Tumoral , Células Cultivadas , Elementos de Facilitación Genéticos/fisiología , Galanina/biosíntesis , Regulación de la Expresión Génica , Variación Genética/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Datos de Secuencia Molecular , Neuroblastoma/genética , Polimorfismo de Nucleótido Simple/fisiología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA