Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Cell ; 186(20): 4404-4421.e20, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37774679

RESUMO

Persistent DNA double-strand breaks (DSBs) in neurons are an early pathological hallmark of neurodegenerative diseases including Alzheimer's disease (AD), with the potential to disrupt genome integrity. We used single-nucleus RNA-seq in human postmortem prefrontal cortex samples and found that excitatory neurons in AD were enriched for somatic mosaic gene fusions. Gene fusions were particularly enriched in excitatory neurons with DNA damage repair and senescence gene signatures. In addition, somatic genome structural variations and gene fusions were enriched in neurons burdened with DSBs in the CK-p25 mouse model of neurodegeneration. Neurons enriched for DSBs also had elevated levels of cohesin along with progressive multiscale disruption of the 3D genome organization aligned with transcriptional changes in synaptic, neuronal development, and histone genes. Overall, this study demonstrates the disruption of genome stability and the 3D genome organization by DSBs in neurons as pathological steps in the progression of neurodegenerative diseases.


Assuntos
Quebras de DNA de Cadeia Dupla , Doenças Neurodegenerativas , Animais , Humanos , Camundongos , Doença de Alzheimer/genética , DNA , Reparo do DNA/genética , Doenças Neurodegenerativas/genética , Neurônios/fisiologia , Análise de Célula Única , Análise de Sequência de RNA , Instabilidade Genômica
2.
Mol Psychiatry ; 28(3): 1112-1127, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36577841

RESUMO

Williams syndrome (WS) is a neurodevelopmental disorder caused by a heterozygous micro-deletion in the WS critical region (WSCR) and is characterized by hyper-sociability and neurocognitive abnormalities. Nonetheless, whether and to what extent WSCR deletion leads to epigenetic modifications in the brain and induces pathological outcomes remains largely unknown. By examining DNA methylation in frontal cortex, we revealed genome-wide disruption in the methylome of individuals with WS, as compared to typically developed (TD) controls. Surprisingly, differentially methylated sites were predominantly annotated as introns and intergenic loci and were found to be highly enriched around binding sites for transcription factors that regulate neuronal development, plasticity and cognition. Moreover, by utilizing enhancer-promoter interactome data, we confirmed that most of these loci function as active enhancers in the human brain or as target genes of transcriptional networks associated with myelination, oligodendrocyte (OL) differentiation, cognition and social behavior. Cell type-specific methylation analysis revealed aberrant patterns in the methylation of active enhancers in neurons and OLs, and important neuron-glia interactions that might be impaired in individuals with WS. Finally, comparison of methylation profiles from blood samples of individuals with WS and healthy controls, along with other data collected in this study, identified putative targets of endophenotypes associated with WS, which can be used to define brain-risk loci for WS outside the WSCR locus, as well as for other associated pathologies. In conclusion, our study illuminates the brain methylome landscape of individuals with WS and sheds light on how these aberrations might be involved in social behavior and physiological abnormalities. By extension, these results may lead to better diagnostics and more refined therapeutic targets for WS.


Assuntos
Síndrome de Williams , Humanos , Síndrome de Williams/genética , Síndrome de Williams/patologia , Neurônios/metabolismo , Metilação de DNA , Oligodendroglia/patologia , DNA
3.
FASEB J ; 36(7): e22406, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35713935

RESUMO

The question of whether behavioral traits are heritable is under debate. An obstacle in demonstrating transgenerational inheritance in mammals originates from the maternal environment's effect on offspring phenotype. Here, we used in ovo embryonic heat conditioning (EHC) of first-generation chicks, demonstrating heredity of both heat and immunological resilience, confirmed by a reduced fibril response in their untreated offspring to either heat or LPS challenge. Concordantly, transcriptome analysis confirmed that EHC induces changes in gene expression in the anterior preoptic hypothalamus (APH) that contribute to these phenotypes in the offspring. To study the association between epigenetic mechanisms and trait heritability, DNA-methylation patterns in the APH of offspring of control versus EHC fathers were evaluated. Genome-wide analysis revealed thousands of differentially methylated sites (DMSs), which were highly enriched in enhancers and CCCTC-binding factor (CTCF) sites. Overlap analysis revealed 110 differentially expressed genes that were associated with altered methylation, predominantly on enhancers. Gene-ontology analysis shows pathways associated with immune response, chaperone-mediated protein folding, and stress response. For the proof of concept, we focused on HSP25 and SOCS3, modulators of heat and immune responses, respectively. Chromosome conformational capture (3C) assay identified interactions between their promoters and methylated enhancers, with the strongest frequency on CTCF binding sites. Furthermore, gene expression corresponded with the differential methylation patterns, and presented increased CTCF binding in both hyper- and hypomethylated DMSs. Collectively, we demonstrate that EHC induces transgenerational thermal and immunological resilience traits. We propose that one of the mechanisms underlying inheritance depends on three-dimensional (3D) chromatin reorganization.


Assuntos
Epigênese Genética , Temperatura Alta , Animais , Galinhas , Metilação de DNA , Padrões de Herança , Mamíferos , Processamento de Proteína Pós-Traducional
4.
Glia ; 70(4): 619-633, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34904755

RESUMO

Stressful environmental events in early life have long-lasting consequences on later stress responses. We previously showed that heat conditioning of 3-day-old chicks during the critical period of heat-response development leads to heat vulnerability later in life. Here we assessed the role of early-life heat stress on the inflammatory response in the chick anterior hypothalamus (AH), focusing on hypothalamic microglia. We identified the microglial cell population in the chick AH using anti-KUL01 and anti-CD45 antibodies. Specific microglial features were also confirmed by expression of their signature genes. Under normal environmental conditions, hypothalamic microglia isolated from lipopolysaccharide (LPS)-injected chicks displayed a classical activated proinflammatory profile accompanied by a decreased homeostatic signature, demonstrating similarity of immune response with mammalian microglial cells. In accordance with our previous observations, conditioning of 3-day-old chicks under high ambient temperature decreased the number of newborn cells in the AH, among them microglial precursors. Although heat exposure did not affect microglial cell viability, it had a long-term impact on LPS-induced inflammatory response. Exposure to harsh heat led to heat vulnerability, and attenuated recruitment of peripheral monocytes and T cells into the AH following LPS challenge. Moreover, heat conditioning altered microglial reactivity, manifested as suppressed microglial activation in response to LPS. Innate immune memory developed by heat conditioning might underlie suppression of the microglial response to LPS challenge. We describe alterations in genome-wide CpG methylation profile of hypothalamic microglia, demonstrating probable epigenetic involvement in the reprogramming of microglial function, leading to heat-induced inflammatory cross-tolerance.


Assuntos
Hipotálamo , Microglia , Animais , Galinhas , Temperatura Alta , Lipopolissacarídeos/toxicidade
5.
Addict Biol ; 22(1): 163-171, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26419743

RESUMO

Binge eating (BE) and drug seeking share similar behavioral features, including loss of control over consumption and compulsive seeking of the craved substance. Previous studies in animal models have demonstrated a complex interaction between 'state' BE, produced by intermittent access to a palatable diet, and 'trait' BE, a phenotypical proneness towards overeating. In the present study, we examined the relationship between state and trait BE and cocaine seeking. We used Otsuka Long Evans Tokushima Fatty rats, a genetic model for obesity that demonstrates BE-like behavior, and Long Evans Tokushima Otsuka controls. They received a schedule of limited access to a palatable diet (3 days/week or 5 days/week access to Ensure for a month). Next, they underwent cocaine self-administration training (1 mg/kg, 1 hour/day for 10 days) followed by extinction sessions (7 days). We found that the degree of BE-like behavior and the state and trait BE combination predicted cocaine craving patterns. Lower levels of dopamine D2 receptors in the prefrontal cortex were correlated with increased drug craving. Moreover, restricted access to an attractive diet was found to be a risk factor for heightened cocaine craving, particularly in trait binge eaters, as rats on the 3 days/week access schedule persistently failed to cease cocaine seeking throughout extinction. Hence, we postulate a joint role of state and trait BE as risk factors for heightened cocaine craving.


Assuntos
Comportamento Aditivo/fisiopatologia , Comportamento Animal/fisiologia , Transtorno da Compulsão Alimentar/fisiopatologia , Cocaína/administração & dosagem , Fissura/fisiologia , Comportamento de Procura de Droga/fisiologia , Animais , Modelos Animais de Doenças , Inibidores da Captação de Dopamina/administração & dosagem , Masculino , Ratos , Ratos Long-Evans
6.
FASEB J ; 28(9): 4148-57, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24928196

RESUMO

This study aimed to determine whether epigenetic malprogramming induced by high-fat diet (HFD) has an obesogenic effect on nonmated and mated female rats and their offspring. Further, it aimed to reprogram offspring's epigenetic malprogramming and phenotype by providing normal diet after weaning. Body weight (BW) was measured, and plasma and hypothalamic arcuate nuclei were collected for analysis of hormones, mRNA, and DNA CpG methylation of the promoter of Pomc, a key factor in control of food intake. In nonmated females, HFD decreased Pomc/leptin ratio by ∼38%. This finding was associated with Pomc promoter hypermethylation. While heavier during pregnancy, during lactation HFD dams showed sharper BW decrease (2.5-fold) and loss of Pomc promoter hypermethylation. Moreover, their weight loss was correlated with demethylation (r=-0.707) and with gadd45b mRNA expression levels (r=0.905). Even though offspring of HFD dams ate standard chow from weaning, they displayed increased BW, Pomc promoter hypermethylation, and vulnerability to HFD challenge (3-fold kilocalorie intake increase). These findings demonstrate a long-term effect of maternal HFD on CpG methylation of the Pomc promoter in the offspring, which was not reprogrammed by standard chow from weaning. Further, the results suggest a possible mechanism of demethylation of the Pomc promoter following pregnancy and lactation.


Assuntos
Adiposidade/genética , Ilhas de CpG/genética , Metilação de DNA , Dieta Hiperlipídica/efeitos adversos , Sobrepeso/genética , Regiões Promotoras Genéticas/genética , Pró-Proteína Convertases/genética , Animais , Antígenos de Diferenciação/genética , Peso Corporal , Ingestão de Alimentos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Lactação , Leptina/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Obesidade/etiologia , Gravidez , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Desmame
7.
Front Endocrinol (Lausanne) ; 14: 1121829, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36817590

RESUMO

Introduction: Although dieting is a key factor in improving physiological functions associated with obesity, the role by which histone methylation modulates satiety/hunger regulation of the hypothalamus through weight loss remains largely elusive. Canonically, H3K9me2 is a transcriptional repressive post-translational epigenetic modification that is involved in obesity, however, its role in the hypothalamic arcuate nucleus (ARC) has not been thoroughly explored. Here we explore the role that KDM4D, a specific demethylase of residue H3K9, plays in energy balance by directly modulating the expression of AgRP, a key neuropeptide that regulates hunger response. Methods: We used a rodent model of diet-induced obesity (DIO) to assess whether histone methylation malprogramming impairs energy balance control and how caloric restriction may reverse this phenotype. Using ChIP-qPCR, we assessed the repressive modification of H3K9me2 at the site of AgRP. To elucidate the functional role of KDM4D in reversing obesity via dieting, a pharmacological agent, JIB-04 was used to inhibit the action of KDM4D in vivo. Results: In DIO, downregulation of Kdm4d mRNA results in both enrichment of H3K9me2 on the AgRP promoter and transcriptional repression of AgRP. Because epigenetic modifications are dynamic, it is possible for some of these modifications to be reversed when external cues are altered. The reversal phenomenon was observed in calorically restricted rats, in which upregulation of Kdm4d mRNA resulted in demethylation of H3K9 on the AgRP promoter and transcriptional increase of AgRP. In order to verify that KDM4D is necessary to reverse obesity by dieting, we demonstrated that in vivo inhibition of KDM4D activity by pharmacological agent JIB-04 in naïve rats resulted in transcriptional repression of AgRP, decreasing orexigenic signaling, thus inhibiting hunger. Discussion: We propose that the action of KDM4D through the demethylation of H3K9 is critical in maintaining a stable epigenetic landscape of the AgRP promoter, and may offer a target to develop new treatments for obesity.


Assuntos
Histonas , Obesidade , Ratos , Animais , Histonas/metabolismo , Metilação , Proteína Relacionada com Agouti/metabolismo , Obesidade/metabolismo , Hipotálamo/metabolismo , RNA Mensageiro/metabolismo
8.
Commun Biol ; 6(1): 1269, 2023 12 14.
Artigo em Inglês | MEDLINE | ID: mdl-38097729

RESUMO

Gtf2i encodes the general transcription factor II-I (TFII-I), with peak expression during pre-natal and early post-natal brain development stages. Because these stages are critical for proper brain development, we studied at the single-cell level the consequences of Gtf2i's deletion from excitatory neurons, specifically on mitochondria. Here we show that Gtf2i's deletion resulted in abnormal morphology, disrupted mRNA related to mitochondrial fission and fusion, and altered autophagy/mitophagy protein expression. These changes align with elevated reactive oxygen species levels, illuminating Gtf2i's importance in neurons mitochondrial function. Similar mitochondrial issues were demonstrated by Gtf2i heterozygous model, mirroring the human condition in Williams syndrome (WS), and by hemizygous neuronal Gtf2i deletion model, indicating Gtf2i's dosage-sensitive role in mitochondrial regulation. Clinically relevant, we observed altered transcript levels related to mitochondria, hypoxia, and autophagy in frontal cortex tissue from WS individuals. Our study reveals mitochondrial and autophagy-related deficits shedding light on WS and other Gtf2i-related disorders.


Assuntos
Fatores de Transcrição TFIII , Síndrome de Williams , Humanos , Autofagia/genética , Heterozigoto , Neurônios/metabolismo , Fatores de Transcrição TFIII/genética , Fatores de Transcrição TFIII/metabolismo , Síndrome de Williams/genética , Síndrome de Williams/metabolismo
9.
Cell Stem Cell ; 29(1): 116-130.e7, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34995493

RESUMO

Down syndrome (DS) is a genetic disorder driven by the triplication of chromosome 21 (T21) and characterized by a wide range of neurodevelopmental and physical disabilities. Transcriptomic analysis of tissue samples from individuals with DS has revealed that T21 induces a genome-wide transcriptional disruption. However, the consequences of T21 on the nuclear architecture and its interplay with the transcriptome remain unknown. In this study, we find that unlike human induced pluripotent stem cells (iPSCs), iPSC-derived neural progenitor cells (NPCs) exhibit genome-wide "chromosomal introversion," disruption of lamina-associated domains, and global chromatin accessibility changes in response to T21, consistent with the transcriptional and nuclear architecture changes characteristic of senescent cells. Treatment of T21-harboring NPCs with senolytic drugs alleviates the transcriptional, molecular, and cellular dysfunctions associated with DS. Our findings provide a mechanistic link between T21 and global transcriptional disruption and indicate that senescence-associated phenotypes may play a key role in the neurodevelopmental pathogenesis of DS.


Assuntos
Síndrome de Down , Células-Tronco Pluripotentes Induzidas , Células-Tronco Neurais , Perfilação da Expressão Gênica , Humanos , Transcriptoma/genética
10.
Nat Neurosci ; 23(12): 1606-1617, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33020654

RESUMO

The epigenome and three-dimensional (3D) genomic architecture are emerging as key factors in the dynamic regulation of different transcriptional programs required for neuronal functions. In this study, we used an activity-dependent tagging system in mice to determine the epigenetic state, 3D genome architecture and transcriptional landscape of engram cells over the lifespan of memory formation and recall. Our findings reveal that memory encoding leads to an epigenetic priming event, marked by increased accessibility of enhancers without the corresponding transcriptional changes. Memory consolidation subsequently results in spatial reorganization of large chromatin segments and promoter-enhancer interactions. Finally, with reactivation, engram neurons use a subset of de novo long-range interactions, where primed enhancers are brought in contact with their respective promoters to upregulate genes involved in local protein translation in synaptic compartments. Collectively, our work elucidates the comprehensive transcriptional and epigenomic landscape across the lifespan of memory formation and recall in the hippocampal engram ensemble.


Assuntos
Epigenômica , Hipocampo/fisiologia , Memória/fisiologia , Rememoração Mental/fisiologia , Transcriptoma , Animais , Mapeamento Encefálico , Consolidação da Memória/fisiologia , Camundongos , Camundongos Transgênicos , Neurônios/fisiologia , Sinapses/metabolismo , Sinapses/fisiologia , Regulação para Cima/fisiologia
11.
Am J Physiol Regul Integr Comp Physiol ; 297(6): R1749-60, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19793959

RESUMO

Understanding the early factors affecting obesity development in males and females may help to prevent obesity and may lead to the discovery of more effective treatments for those already obese. The Otsuka Long-Evans Tokushima Fatty (OLETF) rat model of obesity is characterized by hyperphagia-induced obesity, due to a spontaneous lack of CCK(1) receptors. In the present study, we focused on the behavioral and physiological aspects of obesity development from weaning to adulthood. We examined body weight, feeding efficiency, fat pad [brown, retroperitoneal, inguinal and epydidimal (in males)] weight, inguinal adipocyte size and number, leptin and oxytocin levels, body mass index, waist circumference, and females' estrous cycle structure. In the males, central hypothalamic gene expression was also examined. OLETF rats presented overall higher fat and leptin levels, larger adipocytes, and increased waist circumference and BMI from weaning until adulthood, compared with controls. Analysis of developmental patterns of gene expression for hypothalamic neuropeptides revealed peptide-specific patterns that may underlie or be a consequence of the obesity development. Analysis of the developmental trajectories toward obesity within the OLETF strain revealed that OLETF females developed obesity in a more gradual manner than the males, presenting delayed obesity-related "turning points," with reduced adipocyte size but larger postweaning fat pads and increased adipocyte hyperplasia compared with the males. Intake decrease in estrus vs. proestrus was significantly less in OLETF vs. Long-Evans Tokushima Otsuka females. The findings highlight the importance of using different sex-appropriate approaches to increase the efficacy of therapeutic interventions in the treatment and prevention of chronic early-onset obesity.


Assuntos
Envelhecimento , Comportamento Alimentar , Hiperfagia/fisiopatologia , Obesidade/fisiopatologia , Ratos Endogâmicos OLETF , Adipócitos/patologia , Tecido Adiposo/patologia , Tecido Adiposo/fisiopatologia , Fatores Etários , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Glicemia/metabolismo , Índice de Massa Corporal , Peso Corporal , Doença Crônica , Modelos Animais de Doenças , Progressão da Doença , Ingestão de Alimentos , Estro , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Hiperfagia/genética , Hiperfagia/metabolismo , Hiperfagia/psicologia , Hipotálamo/metabolismo , Hipotálamo/fisiopatologia , Leptina/sangue , Masculino , Neuropeptídeos/genética , Obesidade/genética , Obesidade/metabolismo , Obesidade/psicologia , Ocitocina/sangue , RNA Mensageiro/metabolismo , Ratos , Receptor de Colecistocinina A/deficiência , Receptor de Colecistocinina A/genética
12.
Neuron ; 102(5): 929-943.e8, 2019 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-31076275

RESUMO

Neuronal and synaptic loss is characteristic in many neurodegenerative diseases, such as frontotemporal dementia and Alzheimer's disease. Recently, we showed that inducing gamma oscillations with visual stimulation (gamma entrainment using sensory stimuli, or GENUS) reduced amyloid plaques and phosphorylated tau in multiple mouse models. Whether GENUS can affect neurodegeneration or cognitive performance remains unknown. Here, we demonstrate that GENUS can entrain gamma oscillations in the visual cortex, hippocampus, and prefrontal cortex in Tau P301S and CK-p25 mouse models of neurodegeneration. Tau P301S and CK-p25 mice subjected to chronic, daily GENUS from the early stages of neurodegeneration showed a preservation of neuronal and synaptic density across multiple brain areas and modified cognitive performance. Our transcriptomic and phosphoproteomic data suggest that chronic GENUS shifts neurons to a less degenerative state, improving synaptic function, enhancing neuroprotective factors, and reducing DNA damage in neurons while also reducing inflammatory response in microglia.


Assuntos
Ritmo Gama/fisiologia , Hipocampo/fisiopatologia , Doenças Neurodegenerativas/fisiopatologia , Neurônios/patologia , Neuroproteção/fisiologia , Estimulação Luminosa/métodos , Córtex Pré-Frontal/fisiopatologia , Córtex Visual/fisiopatologia , Animais , Dano ao DNA , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Hipocampo/metabolismo , Hipocampo/patologia , Inflamação , Camundongos , Microglia/imunologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Fosfoproteínas/metabolismo , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Proteômica , Aprendizagem Espacial/fisiologia , Memória Espacial/fisiologia , Sinapses/metabolismo , Sinapses/patologia , Córtex Visual/metabolismo , Córtex Visual/patologia
14.
Endocrinology ; 158(4): 842-851, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28324105

RESUMO

Maternal obesity is a risk factor for offspring obesity. The melanocortin 4 receptor (Mc4r) is one of the mediators of food intake and energy balance. The present study examined the epigenetic mechanisms underlying altered Mc4r levels in the hypothalamic paraventricular nucleus in the offspring of high-fat diet (HFD)-induced obese dams and sought to elucidate the role of thyroid hormones in epigenetic regulation and tagging of their nucleosome at the Mc4r promoter. Female Wistar rats were fed an HFD or standard chow from weaning through gestation and lactation. Epigenetic alterations were analyzed in the offspring on postnatal day 21 at the Mc4r promoter using chromatin immunoprecipitation and bisulfite sequencing. To study the role of triiodothyronine (T3) in Mc4r downregulation, dams received methimazole (MMI), an inhibitor of thyroid hormone production. Offspring of HFD-fed dams had a greater body weight, elevated plasma T3 concentrations, and lower Mc4r messenger RNA levels than controls. At the Mc4r promoter, offspring of HFD-fed mothers demonstrated increased histone 3 lysine 27 acetylation (H3K27ac) with a greater association to thyroid hormone receptor-ß (TRß), an inhibitor of Mc4r transcription. Moreover, TRß coimmunoprecipitated with H3K27ac, supporting their presence in the same complex. Maternal MMI administration prevented the HFD reduction in Mc4r levels, the increase in TRß, and the increase in the TRß-H3K27ac association, providing further support for the role of T3 in downregulating Mc4r levels. These findings demonstrate that a perinatal HFD environment affects Mc4r regulation through a T3 metabolic pathway involving histone acetylation of its promoter.


Assuntos
Epigênese Genética , Receptor Tipo 4 de Melanocortina/metabolismo , Tri-Iodotironina/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Feminino , Fenômenos Fisiológicos da Nutrição Materna , Metimazol/farmacologia , Obesidade/metabolismo , Gravidez , Regiões Promotoras Genéticas , Ratos , Ratos Wistar , Receptor Tipo 4 de Melanocortina/genética
15.
Diabetes ; 65(8): 2258-67, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27217481

RESUMO

A maternal high-fat diet (HFD) alters the offspring's feeding regulation, leading to obesity. This phenomenon is partially mediated by aberrant expression of the hypothalamic anorexigenic neuropeptide proopiomelanocortin (POMC). Nevertheless, although some individual offspring suffer from morbid obesity, others escape the malprogramming. It is suggested that this difference is due to epigenetic programming. In this study, we report that in lean offspring of non-HFD-fed dams, essential promoter regions for Pomc expression were enriched with 5-hydroxymethylcytosine (5hmC) together with a reduction in the level of 5-methylcytosine (5mC). Moreover, 5hmC was negatively correlated whereas 5mC was positively correlated with body weight in offspring from both HFD- and control-fed dams. We further found that Pomc expression in obese offspring is determined by a two-step epigenetic inhibitory mechanism in which CpG methylation is linked with histone posttranslational modifications. An increase in CpG methylation at the Poxmc promoter enables binding of methyl-binding domain 1 (MBD1) to 5mC, but not to its derivative 5hmC. MBD1 then interacts with SET domain bifurcated 1 methyltransferase to promote bimethylation on the histone 3 lysine 9 residue, reducing Pomc mRNA expression. These results suggest an epigenetic regulatory mechanism that affects obesity-prone or resilient traits.


Assuntos
5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Histonas/metabolismo , Pró-Opiomelanocortina/genética , Regiões Promotoras Genéticas/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Ilhas de CpG/genética , Metilação de DNA/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Obesidade/metabolismo , Ratos , Ratos Wistar , Sulfitos/farmacologia , Magreza/metabolismo
16.
Psychoneuroendocrinology ; 38(12): 2844-53, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23958347

RESUMO

Impaired response of the brain to the leptin signal leads to a persisting dysregulation of food intake and energy balance. High plasma leptin or insulin should activate proopiomelanocortin (POMC), the precursor of the anorexigenic neuropeptide α-melanocyte-stimulating hormone (α-MSH) in the hypothalamic arcuate nucleus (ARC). Nevertheless, in obesity, this signal transduction pathway might be impaired. In this study we investigated whether chronic high fat (HF) diet consumption from post-weaning to adulthood increases CpG methylation of the Pomc promoter. The hypothesis that this would disrupt the essential binding of the transcription factor Sp1 to the Pomc promoter was tested. Male rats were raised from postnatal day 21 till 90 on either HF or standard diet. As a result HF fed rats were significantly heavier, with high leptin and insulin levels in their plasma but almost no changes in ARC mRNA expression levels of Pomc. The Pomc promoter area in the HF-treated rats was found to be hypermethylated. Furthermore, there was a direct correlation in individual rats between CpG methylation at specific sites that affect Sp1 binding and plasma leptin levels and/or body weight. Although, as expected the HF diet resulted in up-regulation of Sp1, the binding of Sp1 to the hypermethylated Pomc promoter was significantly reduced. Therefore, we suggest that hypermethylation on the promoter region of the Pomc gene can emerge at post-lactation periods and interfere with transcription factor binding, thus blocking the effects of high leptin levels, leading to obesity.


Assuntos
Metilação de DNA/fisiologia , Dieta Hiperlipídica/efeitos adversos , Obesidade/etiologia , Pró-Opiomelanocortina/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Animais , Western Blotting , Peso Corporal/fisiologia , Imunoprecipitação da Cromatina , Ilhas de CpG , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Insulina/sangue , Leptina/sangue , Leptina/fisiologia , Masculino , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição Sp1/genética
17.
Neuropharmacology ; 62(8): 2447-54, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22401956

RESUMO

Given that food is a natural reinforcement, deficits in the reward system can lead to disordered eating behavior, inducing or worsening an already existing pre-obese phenotype. In order to evaluate developmental, food-reward-related measures we used the OLETF rat, an animal model of early-onset overeating-induced obesity, and a natural CCK-1 receptor knockout. Dopamine-like-receptor type 1 (D1R) and D2R levels were examined in a reward-related brain area (Nac shell) and sucrose preference was assessed at selected time points from weaning to adulthood (postnatal day [PND]90). In addition, a group of OLETF was pair fed (PF) to the amount of food consumed by same-age LETO controls (from weaning to PND 90) to examine the contribution of overweight to the alteration in DR expression. In addition, we examined food "craving"-like behavior by analyzing microstructural patterns of licking a palatable liquid diet. OLETF rats expressed significantly lower D2R levels than LETO controls only on PND 90. In PF OLETF, weight and D2R levels were normalized. In addition, OLETF presented exaggerated preference for the high sucrose concentration. After 30-day abstinence, OLETF rats presented significant increased initial rate of licking, suggesting food "craving". Thus, adult OLETF rats demonstrated altered D2R signaling similar to drug-induced sensitization, suggesting a link with their avidity for sucrose and their abnormal craving response. However, the current findings of a late deficit appearance and the novel PF results suggest that deficits in the motivation/regulatory systems of the OLETF rat are a developing process (at least from weaning and on) depending on the overeating and obese phenotype of the rats and not only on the CCK mutation.


Assuntos
Peso Corporal/fisiologia , Comportamento Alimentar/fisiologia , Hiperfagia/metabolismo , Núcleo Accumbens/metabolismo , Obesidade/metabolismo , Animais , Modelos Animais de Doenças , Ingestão de Alimentos/fisiologia , Hiperfagia/genética , Obesidade/genética , Ratos , Ratos Endogâmicos OLETF , Receptor de Colecistocinina A/genética , Receptor de Colecistocinina A/metabolismo , Receptores de Dopamina D1/genética , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Recompensa
18.
Behav Neurosci ; 123(6): 1251-60, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20001108

RESUMO

Ontogenetic trajectories from weaning to adulthood and sex differences in feeding patterns were examined in the Otsuka Long-Evans Tokushima Fatty (OLETF) rat, an animal model of early onset overeating-induced obesity, and a natural cholecystokinin-1 receptor knockout. Overnight patterns of licking a palatable liquid diet (Ensure) were analyzed on Postnatal Days 22, 38, 60, and 90. Because different microstructure profiles may reflect alterations in the influence of positive and negative signals, we examined meal parameters to uncover developing mechanisms underlying eating behavior in this strain. OLETF rats displayed significantly greater caloric intake, larger meals (in number of licks), and more (within-meal) clusters of feeding (which were shorter in duration and contained fewer licks per cluster) than did Long-Evans Tokushima Ohtsuka (LETO) strain controls. OLETF rats also had significantly lower satiety ratios than LETO rats. Moreover, we identified sex differences in the age of emergence of microstructural patterns of obesity-related overeating, suggesting that systems other than cholecystokinin may be disrupted, possibly worsening the OLETF strain's obesity phenotype.


Assuntos
Dieta , Ingestão de Energia/fisiologia , Comportamento Alimentar/fisiologia , Receptor de Colecistocinina A/genética , Fatores Etários , Análise de Variância , Animais , Peso Corporal/genética , Peso Corporal/fisiologia , Ingestão de Alimentos/genética , Ingestão de Alimentos/fisiologia , Ingestão de Energia/genética , Feminino , Hiperfagia/genética , Masculino , Mutação , Ratos , Ratos Endogâmicos OLETF , Ratos Long-Evans , Fatores Sexuais , Especificidade da Espécie
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA