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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neuroendocrinology ; 113(10): 1076-1090, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37517388

RESUMO

INTRODUCTION: In the hippocampus, clock gene expression is important for memory and mood; however, the signaling mechanism controlling clock gene expression in the hippocampus is unknown. Recent findings suggest that circadian glucocorticoid rhythms driven by the suprachiasmatic nucleus (SCN) control rhythmic clock gene expression in neurons; in addition, dexamethasone modulates hippocampal clock gene expression. We therefore hypothesized that oscillations of clock genes in the hippocampus could be driven by SCN-controlled circadian rhythms in glucocorticoids. METHODS: Temporal profiles of hippocampal clock gene expression were established by quantitative reverse-transcription real-time PCR on rat hippocampi, while cellular distribution was established by in situ hybridization. To determine the effect of rhythmic glucocorticoids on hippocampal clock gene expression, the SCN was lesioned, adrenal glands removed and a 24 h exogenous corticosterone rhythm at physiological levels was reestablished by use of a programmable infusion pump. RESULTS: Daily rhythms were detected for Per1, Per2, Bmal1, Nr1d1, and Dbp, while clock gene products were confirmed in both the hippocampus proper and the dentate gyrus. In sham controls, differential hippocampal expression of Per1 and Dbp between ZT3 and ZT15 was detectable. This rhythm was abolished by SCN lesion; however, reestablishing the natural rhythm in corticosterone restored differential rhythmic expression of both Per1 and Dbp. Further, a 6 h phase delay in the corticosterone profile caused a predictable shift in expression of Nr1d1. CONCLUSION: Our data show that rhythmic corticosterone can drive hippocampal clock gene rhythms suggesting that the SCN regulates the circadian oscillator of the hippocampus by controlling the circadian rhythm in circulating glucocorticoids.


Assuntos
Relógios Circadianos , Ratos , Animais , Relógios Circadianos/genética , Corticosterona/farmacologia , Corticosterona/metabolismo , Ritmo Circadiano/genética , Núcleo Supraquiasmático/metabolismo , Glucocorticoides/farmacologia , Glucocorticoides/metabolismo , Hipocampo/metabolismo
2.
J Pineal Res ; 75(2): e12893, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37349875

RESUMO

Circadian clock gene expression in the suprachiasmatic nucleus (SCN) controls 24 h rhythms in body functions, but clock genes are also expressed in extra-hypothalamic tissues, including the melatonin-producing pineal gland. The nocturnal increase in pineal melatonin synthesis is a hallmark in circadian biology, but the role of local clock gene oscillations in the mammalian pineal gland is unknown. The aim of this work is to determine the role of clock genes in endocrine function of the pineal gland with focus on the Aanat transcript encoding the rhythm-generating enzyme of melatonin synthesis. Using the rat as a model, we here established 24 h expression patterns of clock genes in the pineal gland in vivo. Lesion studies showed that rhythmic clock gene expression in the pineal gland to a large extent depends on the SCN; further, clock gene rhythms could be re-established in cultured pineal cells synchronized by rhythmic stimulation with norepinephrine in 12 h pulses, suggesting that pineal cells house a slave oscillator controlled by adrenergic signaling in the gland. Histological analyses showed that clock genes are expressed in pinealocytes and colocalize with Aanat transcripts, thus potentially enabling clock gene products to control cellular melatonin production. To test this, cultured pineal cells were transfected using small interfering RNA to knock down clock gene expression. While successful knockdown of Per1 had a minor effect on Aanat, Clock knockdown produced a marked overexpression of Aanat in the pinealocytes. Our study suggests that SCN-dependent rhythmic Clock gene expression in the pinealocytes regulates the daily profile of Aanat expression.


Assuntos
Relógios Circadianos , Melatonina , Glândula Pineal , Ratos , Animais , Melatonina/metabolismo , Glândula Pineal/metabolismo , Fatores de Transcrição/metabolismo , Núcleo Supraquiasmático/metabolismo , Ritmo Circadiano/genética , Mamíferos/metabolismo
3.
J Pineal Res ; 75(4): e12905, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37649242

RESUMO

Melatonin is synthesized in the pineal gland during nighttime in response to nocturnal increase in the activity of the enzyme aralkylamine N-acetyltransferase (AANAT), the transcription of which is modulated by several homeodomain transcription factors. Recent work suggests that the homeodomain transcription factor ISL LIM homeobox 2 (ISL2) is expressed in the pineal gland, but its role is currently unknown. With the purpose of identifying the mechanisms that control pineal expression of Isl2 and the possible function of Isl2 in circadian pineal biology, we report that Isl2 is specifically expressed in the pinealocytes of the rat pineal gland. Its expression exhibits a 24 h rhythm with high transcript and protein levels during the day and a trough in the second half of the night. This rhythm persists in darkness, and lesion studies reveal that it requires intact function of the suprachiasmatic nuclei, suggesting intrinsic circadian regulation. In vivo and in vitro experiments show that pineal Isl2 expression is repressed by adrenergic signaling acting via cyclic AMP; further, Isl2 is negatively regulated by the nocturnal transcription factor cone-rod homeobox. During development, pineal Isl2 expression is detectable from embryonic day 19, preceding Aanat by several days. In vitro knockdown of Isl2 is accompanied by an increase in Aanat transcript levels suggesting that ISL2 represses its daytime expression. Thus, rhythmic expression of ISL2 in pinealocytes is under the control of the suprachiasmatic nucleus acting via adrenergic signaling in the gland to repress nocturnal expression, while ISL2 itself negatively regulates daytime pineal expression of Aanat and thereby suggestively enhances the circadian rhythm in melatonin synthesis.

4.
J Pineal Res ; 71(2): e12753, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34129741

RESUMO

Homeobox genes encode transcription factors controlling development; however, a number of homeobox genes are expressed postnatally specifically in melatonin-producing pinealocytes of the pineal gland and photoreceptors of the retina along with transcripts devoted to melatonin synthesis and phototransduction. Homeobox genes regulate melatonin synthesis in pinealocytes, but some homeobox genes also seem to be involved in regulation of retinal phototransduction. Due to the lack of photoreceptor models, we here introduce the rat pinealocyte culture as an in vitro model for studying retinal phototransduction. Systematic qPCR analyses were performed on the rat retina and pineal gland in 24 hour in vivo series and on primary cultures of rat pinealocytes: All homeobox genes and melatonin synthesis components, as well as nine out of ten phototransduction genes, were readily detectable in all three experimental settings, confirming molecular similarity between cultured pinealocytes and in vivo retinal tissue. 24 hours circadian expression was mostly confined to transcripts in the pineal gland, including a novel rhythm in arrestin (Sag). Individual knockdown of the homeobox genes orthodenticle homeobox 2 (Otx2), cone-rod homeobox (Crx) and LIM homeobox 4 (Lhx4) in pinealocyte culture using siRNA resulted in specific downregulation of transcripts representing all levels of phototransduction; thus, all phototransduction genes studied in culture were affected by one or several siRNA treatments. Histological colocalization of homeobox and phototransduction transcripts in the rat retinal photoreceptor was confirmed by RNAscope in situ hybridization, thus suggesting that homeobox gene-encoded transcription factors control postnatal expression of phototransduction genes in the retinal photoreceptor.


Assuntos
Melatonina , Glândula Pineal , Animais , Genes Homeobox , Transdução de Sinal Luminoso , Melatonina/metabolismo , Glândula Pineal/metabolismo , Ratos , Fatores de Transcrição/metabolismo
5.
Addict Biol ; 22(2): 411-422, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26632340

RESUMO

The drive to eat is regulated by two compensatory brain pathways termed as homeostatic and hedonic. Hypothalamic orexinergic (ORX) neurons regulate metabolism, feeding and reward, thus controlling physiological and hedonic appetite. Circadian regulation of feeding, metabolism and rhythmic activity of ORX cells are driven by the brain suprachiasmatic clock. How the circadian clock impacts on ORX signalling and feeding-reward rhythms is, however, unknown. Here we used mice lacking the nuclear receptor REV-ERBα, a transcription repressor and a key component of the molecular clockwork, to study food-reward behaviour. Rev-Erbα mutant mice showed highly motivated behaviours to obtain palatable food, an increase in the intake and preference for tasty diets, and in the expression of the ORX protein in the hypothalamus. Palatable food intake was inhibited in animals treated with the ORX1R antagonist. Analyzing the Orx promoter, we found Retinoic acid-related Orphan receptor Response Element binding sites for Rev-Erbα. Furthermore, Rev-Erbα dampened the activation of Orx in vitro and in vivo. Our data provide evidence for a possible repressive role of Rev-Erbα in the regulation of ORX signalling, highlighting an implication of the circadian clockwork in modulating food-reward behaviours with an important impact for the central regulation of overeating.


Assuntos
Ingestão de Alimentos/genética , Comportamento Alimentar/fisiologia , Hipotálamo/metabolismo , Neurônios/metabolismo , Membro 1 do Grupo D da Subfamília 1 de Receptores Nucleares/genética , Orexinas/metabolismo , Animais , Ritmo Circadiano , Ingestão de Alimentos/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Antagonistas dos Receptores de Orexina/farmacologia , Receptores de Orexina/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais
6.
Appetite ; 117: 263-269, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28687372

RESUMO

The suprachiasmatic nucleus (SCN) times the daily rhythms of behavioral processes including feeding. Beyond the SCN, the hypothalamic arcuate nucleus (ARC), involved in feeding regulation and metabolism, and the epithalamic lateral habenula (LHb), implicated in reward processing, show circadian rhythmic activity. These brain oscillators are functionally coupled to coordinate the daily rhythm of food intake. In rats, a free choice high-fat high-sugar (fcHFHS) diet leads to a rapid increase of calorie intake and body weight gain. Interestingly, under a fcHFHS condition, rats ingest a similar amount of sugar during day time (rest phase) as during night time (active phase), but keep the rhythmic intake of regular chow-food. The out of phase between feeding patterns of regular (chow) and highly rewarding food (sugar) may involve alterations of brain circadian oscillators regulating feeding. Here, we report that the fcHFHS diet is a successful model to induce calorie intake, body weight gain and fat tissue accumulation in mice, extending its effectiveness as previously reported in rats. Moreover, we observed that whereas in the SCN the day-night difference in the PER2 clock protein expression was similar between chow-fed and fcHFHS-fed animals, in the LHb, this day-night difference was altered in fcHFHS-exposed animals compared to control chow mice. These findings confirm previous observations in rats showing disrupted daily patterns of feeding behavior under a fcHFHS diet exposure, and extend our insights on the effects of the diet on circadian gene expression in brain clocks.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Dieta Ocidental/efeitos adversos , Comportamento Alimentar , Preferências Alimentares , Regulação da Expressão Gênica , Habenula/metabolismo , Proteínas Circadianas Period/metabolismo , Fatores de Transcrição ARNTL/genética , Adiposidade , Animais , Comportamento Animal , Comportamento de Escolha , Ritmo Circadiano , Habenula/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Especificidade de Órgãos , Proteínas Circadianas Period/genética , Distribuição Aleatória , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/patologia , Aumento de Peso
7.
Neuropharmacology ; 256: 110007, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-38795953

RESUMO

Feeding, like many other biological functions, displays a daily rhythm. This daily rhythmicity is controlled by the circadian timing system of which the central master clock is located in the hypothalamic suprachiasmatic nucleus (SCN). Other brain areas and tissues throughout the body also display rhythmic functions and contain the molecular clock mechanism known as peripheral oscillators. To generate the daily feeding rhythm, the SCN signals to different hypothalamic areas with the lateral hypothalamus, paraventricular nucleus and arcuate nucleus being the most prominent. With respect to the rewarding aspects of feeding behavior, the dopaminergic system is also under circadian influence. However the SCN projects only indirectly to the different reward regions, such as the ventral tegmental area where dopamine neurons are located. In addition, high palatable, high caloric diets have the potential to disturb the normal daily rhythms of physiology and have been shown to alter for example meal patterns. Around a meal several hormones and peptides are released that are also under circadian influence. For example, the release of postprandial insulin and glucagon-like peptide following a meal depend on the time of the day. Finally, we review the effect of deletion of different clock genes on feeding behavior. The most prominent effect on feeding behavior has been observed in Clock mutants, whereas deletion of Bmal1 and Per1/2 only disrupts the day-night rhythm, but not overall intake. Data presented here focus on the rodent literature as only limited data are available on the mechanisms underlying daily rhythms in human eating behavior.


Assuntos
Ritmo Circadiano , Comportamento Alimentar , Animais , Comportamento Alimentar/fisiologia , Ritmo Circadiano/fisiologia , Humanos , Núcleo Supraquiasmático/fisiologia , Núcleo Supraquiasmático/metabolismo
8.
Methods Mol Biol ; 2550: 113-121, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36180684

RESUMO

Suspension primary cultures of rat pineal cells have been used for decades to determine biochemical regulatory mechanisms of pineal melatonin synthesis, but more recently, RNA interference technology has made the study of the role of specific genes in this melatonin-proficient model system possible. We here present a protocol for preparing rat pineal cell cultures and efficiently knock down gene expression by use of synthetic siRNA.


Assuntos
Melatonina , Glândula Pineal , Animais , Células Cultivadas , Regulação para Baixo , Expressão Gênica , Melatonina/genética , Melatonina/metabolismo , Glândula Pineal/metabolismo , RNA de Cadeia Dupla/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley
9.
J Neuroendocrinol ; 31(5): e12718, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30958590

RESUMO

Humans have engineered a dietary environment that has driven the global prevalence of obesity and several other chronic metabolic diseases to pandemic levels. To prevent or treat obesity and associated comorbidities, it is crucial that we understand how our dietary environment, especially in combination with a sedentary lifestyle and/or daily-life stress, can dysregulate energy balance and promote the development of an obese state. Substantial mechanistic insight into the maladaptive adaptations underlying caloric overconsumption and excessive weight gain has been gained by analysing brains from rodents that were eating prefabricated nutritionally-complete pellets of high-fat diet (HFD). Although long-term consumption of HFDs induces chronic metabolic diseases, including obesity, they do not model several important characteristics of the modern-day human diet. For example, prefabricated HFDs ignore the (effects of) caloric consumption from a fluid source, do not appear to model the complex interplay in humans between stress and preference for palatable foods, and, importantly, lack any aspect of choice. Therefore, our laboratory uses an obesogenic free-choice high-fat high-sucrose (fc-HFHS) diet paradigm that provides rodents with the opportunity to choose from several diet components, varying in palatability, fluidity, texture, form and nutritive content. Here, we review recent advances in our understanding how the fc-HFHS diet disrupts peripheral metabolic processes and produces adaptations in brain circuitries that govern homeostatic and hedonic components of energy balance. Current insight suggests that the fc-HFHS diet has good construct and face validity to model human diet-induced chronic metabolic diseases, including obesity, because it combines the effects of food palatability and energy density with the stimulating effects of variety and choice. We also highlight how behavioural, physiological and molecular adaptations might differ from those induced by prefabricated HFDs that lack an element of choice. Finally, the advantages and disadvantages of using the fc-HFHS diet for preclinical studies are discussed.


Assuntos
Dieta Hiperlipídica , Modelos Animais de Doenças , Ingestão de Energia , Doenças Metabólicas/fisiopatologia , Obesidade/fisiopatologia , Animais , Comportamento de Escolha , Açúcares da Dieta/administração & dosagem , Metabolismo Energético , Humanos , Doenças Metabólicas/etiologia , Doenças Metabólicas/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Estresse Psicológico
11.
Artigo em Inglês | MEDLINE | ID: mdl-29686649

RESUMO

Under normal light-dark conditions, nocturnal rodents consume most of their food during the dark period. Diets high in fat and sugar, however, may affect the day-night feeding rhythm resulting in a higher light phase intake. In vitro and in vivo studies showed that nutrients affect clock-gene expression. We therefore hypothesized that overconsuming fat and sugar alters clock-gene expression in brain structures important for feeding behavior. We determined the effects of a free-choice high-fat high-sugar (fcHFHS) diet on clock-gene expression in rat brain areas related to feeding and reward and compared them with chow-fed rats. Consuming a fcHFHS diet for 6 weeks disrupted day-night differences in Per2 mRNA expression in the nucleus accumbens (NAc) and lateral hypothalamus but not in the suprachiasmatic nucleus, habenula, and ventral tegmental area. Furthermore, short-term sugar drinking, but not fat feeding, upregulates Per2 mRNA expression in the NAc. The disruptions in day-night differences in NAc Per2 gene expression were not accompanied by altered day-night differences in the mRNA expression of peptides related to food intake. We conclude that the fcHFHS diet and acute sugar drinking affect Per2 gene expression in areas involved in food reward; however, this is not sufficient to alter the day-night pattern of food intake.

12.
Front Neurosci ; 11: 23, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28223912

RESUMO

Feeding behavior shows a rhythmic daily pattern, which in nocturnal rodents is observed mainly during the dark period. This rhythmicity is under the influence of the hypothalamic suprachiasmatic nucleus (SCN), the main biological clock. Nevertheless, various studies have shown that in rodent models of obesity, using high-energy diets, the general locomotor activity and feeding rhythms can be disrupted. Here, we review the data on the effects of diet-induced obesity (DIO) on locomotor activity and feeding patterns, as well as the effect on the brain sites within the neural circuitry involved in metabolic and rewarding feeding behavior. In general, DIO may alter locomotor activity by decreasing total activity. On the other hand, DIO largely alters eating patterns, producing increased overall ingestion and number of eating bouts that can extend to the resting period. Furthermore, within the hypothalamic areas, little effect has been reported on the molecular circadian mechanism in DIO animals with ad libitum hypercaloric diets and little or no data exist so far on its effects on the reward system areas. We further discuss the possibility of an uncoupling of metabolic and reward systems in DIO and highlight a gap of circadian and metabolic research that may help to better understand the implications of obesity.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA