Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters










Publication year range
1.
Front Microbiol ; 15: 1392016, 2024.
Article in English | MEDLINE | ID: mdl-38746744

ABSTRACT

Consumption of dietary fiber has been linked to several health benefits. Among these, dietary fiber breakdown through the process of anaerobic fermentation by the colonic microbiota leads to the production of beneficial metabolites, mainly short-chain fatty acids (acetate, propionate, and butyrate), which have been implicated in reduced calorie intake. Nevertheless, the link between gut microbiota and obesity remains unclear. We investigated the effects of dietary fibers on food intake and body weight gain in two independent but similarly designed studies in rats. In the first study, the inclusion of 10% w/w pectin, fructooligosaccharides or beta-glucan (n = 10/group) in the diets each significantly reduced body weight gain ('responders') compared to the cellulose control whereas, in a closely matched, but not fully identical study (n = 8/group), no effect of dietary fiber on body weight ('non-responders') was observed. The aim of this work was to explore the basis of this differential response between the two similarly designed and comparable studies, with a focus on the potential role of the gut microbiota in the control of food intake and body weight.

2.
iScience ; 26(8): 107373, 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37599827

ABSTRACT

Vitamin A is a micronutrient essential for vertebrate animals maintained in homeostatic balance in the body; however, little is known about the control of this balance. This study investigated whether the hypothalamus, a key integrative brain region, regulates vitamin A levels in the liver and circulation. Vitamin A in the form of retinol or retinoic acid was stereotactically injected into the 3rd ventricle of the rat brain. Alternatively, retinoids in the mouse hypothalamus were altered through retinol-binding protein 4 (Rbp4) gene knockdown. This led to rapid change in the liver proteins controlling vitamin A homeostasis as well as vitamin A itself in liver and the circulation. Prolonged disruption of Rbp4 in the region of the arcuate nucleus of the mouse hypothalamus altered retinol levels in the liver. This supports the concept that the brain may sense retinoids and influence whole-body vitamin A homeostasis with a possible "vitaminostatic" role.

3.
Financ Innov ; 9(1): 101, 2023.
Article in English | MEDLINE | ID: mdl-37325238

ABSTRACT

This study investigates how financial literacy and behavioral traits affect the adoption of electronic payment (ePayment) services in Japan. We construct a financial literacy index using a representative sample of 25,000 individuals from the Bank of Japan's 2019 Financial Literacy Survey. We then analyze the relationship between this index and the extensive and intensive usage of two types of payment services: electronic money (e-money) and mobile payment apps. Using an instrumental variable approach, we find that higher financial literacy is positively associated with a higher likelihood of adopting ePayment services. The empirical results suggest that individuals with higher financial literacy use payment services more frequently. We also find that risk-averse people are less likely to adopt and use ePayment services, whereas people with herd behavior tend to adopt and use ePayment services more. Our empirical results also suggest that the effects of financial literacy on the adoption and use of ePayment differ among people with different behavioral traits. Supplementary Information: The online version contains supplementary material available at 10.1186/s40854-023-00504-3.

4.
Cell Metab ; 34(10): 1472-1485.e6, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36087576

ABSTRACT

Morning loaded calorie intake in humans has been advocated as a dietary strategy to improve weight loss. This is also supported by animal studies suggesting time of eating can prevent weight gain. However, the underlying mechanisms through which timing of eating could promote weight loss in humans are unclear. In a randomized crossover trial (NCT03305237), 30 subjects with obesity/overweight underwent two 4-week calorie-restricted but isoenergetic weight loss diets, with morning loaded or evening loaded calories (45%:35%:20% versus 20%:35%:45% calories at breakfast, lunch, and dinner, respectively). We demonstrate no differences in total daily energy expenditure or resting metabolic rate related to the timing of calorie distribution, and no difference in weight loss. Participants consuming the morning loaded diet reported significantly lower hunger. Thus, morning loaded intake (big breakfast) may assist with compliance to weight loss regime through a greater suppression of appetite.


Subject(s)
Appetite , Hunger , Animals , Diet, Reducing , Energy Intake/physiology , Energy Metabolism , Healthy Volunteers , Humans , Obesity/metabolism , Weight Loss
5.
J Clin Endocrinol Metab ; 107(2): e708-e715, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34473293

ABSTRACT

CONTEXT: Daily variation in the thermic effect of food (TEF) is commonly reported and proposed as a contributing factor to weight gain with late eating. However, underlying circadian variability in resting metabolic rate (RMR) is an overlooked factor when calculating TEF associated with eating at different times of the day. OBJECTIVE: This work aimed to determine whether methodological approaches to calculating TEF contribute to the reported phenomena of daily variation in TEF. METHODS: Fourteen overweight to obese but otherwise healthy individuals had their resting and postprandial energy expenditure (EE) measured over 15.5 hours at a clinical research unit. TEF was calculated for breakfast, lunch, and dinner using standard methods (above a baseline and premeal RMR measure) and compared to a method incorporating a circadian RMR by which RMR was derived from a sinusoid curve model and TEF was calculated over and above the continuously changing RMR. Main outcome measures were TEF at breakfast, lunch, and dinner calculated by different methods. RESULTS: Standard methods of calculating TEF above a premeal measured RMR showed that morning TEF (60.8 kcal ±â€…5.6) (mean ±â€…SEM) was 1.6 times greater than TEF at lunch (36.3 kcal ±â€…8.4) and 2.4 times greater than dinner TEF (25.2 kcal ±â€…9.6) (P = .022). However, adjusting for modeled circadian RMR nullified any differences between breakfast (54.1 kcal ±â€…30.8), lunch (49.5 kcal ±â€…29.4), and dinner (49.1 kcal ±â€…25.7) (P = .680). CONCLUSION: Differences in TEF between morning and evening can be explained by the underlying circadian resting EE, which is independent of an acute effect of eating.


Subject(s)
Basal Metabolism/physiology , Circadian Rhythm/physiology , Obesity/metabolism , Overweight/metabolism , Thermogenesis/physiology , Adult , Calorimetry, Indirect , Energy Intake , Female , Humans , Male , Middle Aged , Postprandial Period/physiology , Time Factors , Young Adult
6.
Aging Cell ; 20(9): e13455, 2021 09.
Article in English | MEDLINE | ID: mdl-34409748

ABSTRACT

Intracellular amyloid beta oligomer (iAßo) accumulation and neuronal hyperexcitability are two crucial events at early stages of Alzheimer's disease (AD). However, to date, no mechanism linking iAßo with an increase in neuronal excitability has been reported. Here, the effects of human AD brain-derived (h-iAßo) and synthetic (iAßo) peptides on synaptic currents and action potential firing were investigated in hippocampal neurons. Starting from 500 pM, iAßo rapidly increased the frequency of synaptic currents and higher concentrations potentiated the AMPA receptor-mediated current. Both effects were PKC-dependent. Parallel recordings of synaptic currents and nitric oxide (NO)-associated fluorescence showed that the increased frequency, related to pre-synaptic release, was dependent on a NO-mediated retrograde signaling. Moreover, increased synchronization in NO production was also observed in neurons neighboring those dialyzed with iAßo, indicating that iAßo can increase network excitability at a distance. Current-clamp recordings suggested that iAßo increased neuronal excitability via AMPA-driven synaptic activity without altering membrane intrinsic properties. These results strongly indicate that iAßo causes functional spreading of hyperexcitability through a synaptic-driven mechanism and offers an important neuropathological significance to intracellular species in the initial stages of AD, which include brain hyperexcitability and seizures.


Subject(s)
Amyloid beta-Peptides/metabolism , Synapses/metabolism , Animals , Female , Humans , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Rats, Wistar
7.
J Neuroendocrinol ; 32(7): e12886, 2020 07.
Article in English | MEDLINE | ID: mdl-32662577

ABSTRACT

Circadian rhythms play a critical role in the physiological processes involved in energy metabolism and energy balance (EB). A large array of metabolic processes, including the expression of many energy-regulating endocrine hormones, display temporal rhythms that are driven by both the circadian clock and food intake. Mealtime has been shown to be a compelling zeitgeber in peripheral tissue rhythms. Inconsistent signalling to the periphery, because of mismatched input from the central clock vs time of eating, results in circadian disruption in which central and/or peripheral rhythms are asynchronously time shifted or their amplitudes reduced. A growing body of evidence supports the negative health effects of circadian disruption, with strong evidence in murine models that mealtime-induced circadian disruption results in various metabolic consequences, including energy imbalance and weight gain. Increased weight gain has been reported to occur even without differences in energy intake, indicating an effect of circadian disruption on energy expenditure. However, the translation of these findings to humans is not well established because the ability to undertake rigorously controlled dietary studies that explore the chronic effects on energy regulation is challenging. Establishing the neuroendocrine changes in response to both acute and chronic variations in mealtime, along with observations in populations with routinely abnormal mealtimes, may provide greater insight into underlying mechanisms that influence long-term weight management under different meal patterns. Human studies should explore mechanisms through relevant biomarkers; for example, cortisol, leptin, ghrelin and other energy-regulating neuroendocrine factors. Mistiming between aggregate hormonal signals, or between hormones with their receptors, may cause reduced signalling intensity and hormonal resistance. Understanding how mealtimes may impact on the coordination of endocrine factors is essential for untangling the complex regulation of EB. Here a review is provided on current evidence of the impacts of mealtime on energy metabolism and the underlying neuroendocrine mechanisms, with a specific focus on human research.


Subject(s)
Circadian Rhythm/physiology , Energy Metabolism/physiology , Feeding Behavior/physiology , Animals , Circadian Clocks/physiology , Energy Intake/physiology , Humans , Meals/physiology , Mice , Time Factors
8.
J Neuroendocrinol ; 31(3): e12680, 2019 03.
Article in English | MEDLINE | ID: mdl-30585661

ABSTRACT

Animals have evolved diverse seasonal variations in physiology and reproduction to accommodate yearly changes in environmental and climatic conditions. These changes in physiology are initiated by changes in photoperiod (daylength) and are mediated through melatonin, which relays photoperiodic information to the pars tuberalis of the pituitary gland. Melatonin drives thyroid-stimulating hormone transcription and synthesis in the pars tuberalis, which, in turn, regulates thyroid hormone and retinoic acid synthesis in the tanycytes lining the third ventricle of the hypothalamus. Seasonal variation in central thyroid hormone signalling is conserved among photoperiodic animals. Despite this, different species adopt divergent phenotypes to cope with the same seasonal changes. A common response amongst different species is increased hypothalamic cell proliferation/neurogenesis in short photoperiod. That cell proliferation/neurogenesis may be important for seasonal timing is based on (i) the neurogenic potential of tanycytes; (ii) the fact that they are the locus of striking seasonal morphological changes; and (iii) the similarities to mechanisms involved in de novo neurogenesis of energy balance neurones. We propose that a decrease in hypothalamic thyroid hormone and retinoic acid signalling initiates localised neurodegeneration and apoptosis, which leads to a reduction in appetite and body weight. Neurodegeneration induces compensatory cell proliferation from the neurogenic niche in tanycytes and new cells are born under short photoperiod. Because these cells have the potential to differentiate into a number of different neuronal phenotypes, this could provide a mechanistic basis to explain the seasonal regulation of energy balance, as well as reproduction. This cycle can be achieved without changes in thyroid hormone/retinoic acid and explains recent data obtained from seasonal animals held in natural conditions. However, thyroid/retinoic acid signalling is required to synchronise the cycles of apoptosis, proliferation and differentiation. Thus, hypothalamic neurogenesis provides a framework to explain diverse photoperiodic responses.


Subject(s)
Adaptation, Physiological , Body Weight/physiology , Chronobiology Phenomena , Models, Neurological , Pituitary Gland/metabolism , Reproduction , Animals , Appetite Regulation , Energy Metabolism , Ependymoglial Cells/metabolism , Humans , Melatonin/metabolism , Neurogenesis , Photoperiod , Seasons , Thyroid Hormones/metabolism
9.
Sci Rep ; 8(1): 15648, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30353127

ABSTRACT

Hypothalamic inflammation is thought to contribute to obesity. One potential mechanism is via gut microbiota derived bacterial lipopolysaccharide (LPS) entering into the circulation and activation of Toll-like receptor-4. This is called metabolic endotoxemia. Another potential mechanism is systemic inflammation arising from sustained exposure to high-fat diet (HFD) over more than 12 weeks. In this study we show that mice fed HFD over 8 weeks become obese and show elevated plasma LPS binding protein, yet body weight gain and adiposity is not attenuated in mice lacking Tlr4 or its co-receptor Cd14. In addition, caecal microbiota composition remained unchanged by diet. Exposure of mice to HFD over a more prolonged period (20 weeks) to drive systemic inflammation also caused obesity. RNAseq used to assess hypothalamic inflammation in these mice showed increased hypothalamic expression of Serpina3n and Socs3 in response to HFD, with few other genes altered. In situ hybridisation confirmed increased Serpina3n and Socs3 expression in the ARC and DMH at 20-weeks, but also at 8-weeks and increased SerpinA3N protein could be detected as early as 1 week on HFD. Overall these data show lack of hypothalamic inflammation in response to HFD and that metabolic endotoxemia does not link HFD to obesity.


Subject(s)
Acute-Phase Proteins/genetics , Diet, High-Fat/adverse effects , Endotoxemia/complications , Obesity/etiology , Serpins/genetics , Toll-Like Receptor 4/immunology , Up-Regulation , Animals , Endotoxemia/genetics , Endotoxemia/immunology , Endotoxemia/pathology , Gastrointestinal Microbiome , Gene Expression Regulation , Genotype , Hypothalamus/immunology , Hypothalamus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/immunology , Obesity/pathology , Signal Transduction , Toll-Like Receptor 4/genetics
10.
Cell Rep ; 21(6): 1521-1533, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29117558

ABSTRACT

Evidence suggests that altered gut microbiota composition may be involved in the development of obesity. Studies using mice made obese with refined high-fat diets have supported this; however, these have commonly used chow as a control diet, introducing confounding factors from differences in dietary composition that have a key role in shaping microbiota composition. We compared the effects of feeding a refined high-fat diet with those of feeding either a refined low-fat diet or a chow diet on gut microbiota composition and host physiology. Feeding both refined low- or high-fat diets resulted in large alterations in the gut microbiota composition, intestinal fermentation, and gut morphology, compared to a chow diet. However, body weight, body fat, and glucose intolerance only increased in mice fed the refined high-fat diet. The choice of control diet can dissociate broad changes in microbiota composition from obesity, raising questions about the previously proposed relationship between gut microbiota and obesity.


Subject(s)
Gastrointestinal Microbiome/physiology , Obesity/etiology , Animals , Bacteroidetes/genetics , Bacteroidetes/growth & development , Bacteroidetes/isolation & purification , Blood Glucose/analysis , Body Weight , Cecum/microbiology , Chromatography, High Pressure Liquid , Diet, High-Fat , Fatty Acids, Volatile/analysis , Fatty Acids, Volatile/metabolism , Feces/microbiology , Firmicutes/genetics , Firmicutes/growth & development , Firmicutes/isolation & purification , Glucose Intolerance/metabolism , Glucose Intolerance/microbiology , Glucose Intolerance/pathology , Ileum/microbiology , Male , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/microbiology , RNA, Ribosomal, 16S/chemistry , RNA, Ribosomal, 16S/isolation & purification , RNA, Ribosomal, 16S/metabolism , Sequence Analysis, DNA
11.
Dis Model Mech ; 9(11): 1245-1255, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27821603

ABSTRACT

A multi-dimensional strategy to tackle the global obesity epidemic requires an in-depth understanding of the mechanisms that underlie this complex condition. Much of the current mechanistic knowledge has arisen from preclinical research performed mostly, but not exclusively, in laboratory mouse and rat strains. These experimental models mimic certain aspects of the human condition and its root causes, particularly the over-consumption of calories and unbalanced diets. As with human obesity, obesity in rodents is the result of complex gene-environment interactions. Here, we review the traditional monogenic models of obesity, their contemporary optogenetic and chemogenetic successors, and the use of dietary manipulations and meal-feeding regimes to recapitulate the complexity of human obesity. We critically appraise the strengths and weaknesses of these different models to explore the underlying mechanisms, including the neural circuits that drive behaviours such as appetite control. We also discuss the use of these models for testing and screening anti-obesity drugs, beneficial bio-actives, and nutritional strategies, with the goal of ultimately translating these findings for the treatment of human obesity.


Subject(s)
Biomedical Research , Obesity/pathology , Animals , Diet , Disease Models, Animal , Humans , Models, Genetic , Obesity/genetics , Translational Research, Biomedical
12.
Sci Rep ; 6: 26830, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27225311

ABSTRACT

Long-term and reversible changes in body weight are typical of seasonal animals. Thyroid hormone (TH) and retinoic acid (RA) within the tanycytes and ependymal cells of the hypothalamus have been implicated in the photoperiodic response. We investigated signalling downstream of RA and how this links to the control of body weight and food intake in photoperiodic F344 rats. Chemerin, an inflammatory chemokine, with a known role in energy metabolism, was identified as a target of RA. Gene expression of chemerin (Rarres2) and its receptors were localised within the tanycytes and ependymal cells, with higher expression under long (LD) versus short (SD) photoperiod, pointing to a physiological role. The SD to LD transition (increased food intake) was mimicked by 2 weeks of ICV infusion of chemerin into rats. Chemerin also increased expression of the cytoskeletal protein vimentin, implicating hypothalamic remodelling in this response. By contrast, acute ICV bolus injection of chemerin on a 12 h:12 h photoperiod inhibited food intake and decreased body weight with associated changes in hypothalamic neuropeptides involved in growth and feeding after 24 hr. We describe the hypothalamic ventricular zone as a key site of neuroendocrine regulation, where the inflammatory signal, chemerin, links TH and RA signaling to hypothalamic remodeling.


Subject(s)
Chemokines/physiology , Energy Metabolism/physiology , Hypothalamus/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Photoperiod , Animals , Body Weight/drug effects , Chemokines/administration & dosage , Chemokines/pharmacology , Eating/drug effects , Energy Metabolism/drug effects , Ependyma/cytology , Ependyma/metabolism , Ependymoglial Cells/metabolism , Humans , Hypothalamus/cytology , Hypothalamus/drug effects , Injections, Intraventricular , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Neuronal Plasticity/drug effects , Random Allocation , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Receptors, Chemokine/analysis , Receptors, Chemokine/drug effects , Signal Transduction/drug effects , Thyroid Hormones/physiology
13.
Glia ; 64(3): 425-39, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26527258

ABSTRACT

Thyroid hormone (TH) is essential for adult brain function and its actions include several key roles in the hypothalamus. Although TH controls gene expression via specific TH receptors of the nuclear receptor class, surprisingly few genes have been demonstrated to be directly regulated by TH in the hypothalamus, or the adult brain as a whole. This study explored the rapid induction by TH of retinaldehyde dehydrogenase 1 (Raldh1), encoding a retinoic acid (RA)-synthesizing enzyme, as a gene specifically expressed in hypothalamic tanycytes, cells that mediate a number of actions of TH in the hypothalamus. The resulting increase in RA may then regulate gene expression via the RA receptors, also of the nuclear receptor class. In vivo exposure of the rat to TH led to a significant and rapid increase in hypothalamic Raldh1 within 4 hours. That this may lead to an in vivo increase in RA is suggested by the later induction by TH of the RA-responsive gene Cyp26b1. To explore the actions of RA in the hypothalamus as a potential mediator of TH control of gene regulation, an ex vivo hypothalamic rat slice culture method was developed in which the Raldh1-expressing tanycytes were maintained. These slice cultures confirmed that TH did not act on genes regulating energy balance but could induce Raldh1. RA has the potential to upregulate expression of genes involved in growth and appetite, Ghrh and Agrp. This regulation is acutely sensitive to epigenetic changes, as has been shown for TH action in vivo. These results indicate that sequential triggering of two nuclear receptor signalling systems has the capability to mediate some of the functions of TH in the hypothalamus.


Subject(s)
Ependymoglial Cells/drug effects , Hypothalamus/cytology , Retinal Dehydrogenase/metabolism , Thyroid Hormones/pharmacology , Tretinoin/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Animals, Newborn , Cells, Cultured , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Gene Expression Regulation/drug effects , In Vitro Techniques , Male , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Organ Culture Techniques , Pro-Opiomelanocortin/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Receptors, Retinoic Acid/metabolism , Retinal Dehydrogenase/genetics , Species Specificity , Vimentin/metabolism
14.
Brain Struct Funct ; 221(6): 3315-26, 2016 07.
Article in English | MEDLINE | ID: mdl-26374207

ABSTRACT

Retinoic acid (RA) is a potent regulator of gene transcription via its activation of a set of nuclear receptors controlling transcriptional activation. Precise maintenance of where and when RA is generated is essential and achieved by local expression of synthetic and catabolic enzymes. The catabolic enzymes Cyp26a1 and Cyp26b1 have been studied in detail in the embryo, where they limit gradients of RA that form patterns of gene expression, crucial for morphogenesis. This paracrine role of RA has been assumed to occur in most tissues and that the RA synthetic enzymes release RA at a site distant from the catabolic enzymes. In contrast to the embryonic CNS, relatively little is known about RA metabolism in the adult brain. This study investigated the distribution of Cyp26a1 and Cyp26b1 transcripts in the rat brain, identifying several novel regions of expression, including the cerebral cortex for both enzymes and striatum for Cyp26b1. In vivo use of a new and potent inhibitor of the Cyp26 enzymes, ser 2-7, demonstrated a function for endogenous Cyp26 in the brain and that hippocampal RA levels can be raised by ser 2-7, altering the effect of RA on differential patterning of cell proliferation in the hippocampal region of neurogenesis, the subgranular zone. The expression of CYP26A1 and CYP26B1 was also investigated in the adult human brain and colocalization of CYP26A1 and the RA synthetic enzyme RALDH2 indicated a different, autocrine role for RA in human hippocampal neurons. Studies with the SH-SY5Y human neuroblastoma cell line implied that the co-expression of RA synthetic and catabolic enzymes maintains retinoid homeostasis within neurons. This presents a novel view of RA in human neurons as part of an autocrine, intracellular signaling system.


Subject(s)
Autocrine Communication , Brain/enzymology , Homeostasis , Paracrine Communication , Retinoic Acid 4-Hydroxylase/metabolism , Tretinoin/metabolism , Aldehyde Dehydrogenase 1 Family , Animals , Cell Line, Tumor , Cell Proliferation , Cerebral Cortex/enzymology , Corpus Striatum/enzymology , Female , Gene Expression , Hippocampus/metabolism , Hippocampus/physiology , Humans , Male , Middle Aged , Rats , Retinal Dehydrogenase/metabolism
15.
PLoS One ; 10(3): e0119763, 2015.
Article in English | MEDLINE | ID: mdl-25789758

ABSTRACT

In this study the effects of photoperiod and diet, and their interaction, were examined for their effects on growth and body composition in juvenile F344 rats over a 4-week period. On long (16L:8D), relative to short (8L:16D), photoperiod food intake and growth rate were increased, but percentage adiposity remained constant (ca 3-4%). On a high fat diet (HFD), containing 22.8% fat (45% energy as fat), food intake was reduced, but energy intake increased on both photoperiods. This led to a small increase in adiposity (up to 10%) without overt change in body weight. These changes were also reflected in plasma leptin and lipid levels. Importantly while both lean and adipose tissue were strongly regulated by photoperiod on a chow diet, this regulation was lost for adipose, but not lean tissue, on HFD. This implies that a primary effect of photoperiod is the regulation of growth and lean mass accretion. Consistent with this both hypothalamic GHRH gene expression and serum IGF-1 levels were photoperiod dependent. As for other animals and humans, there was evidence of central hyposomatotropism in response to obesity, as GHRH gene expression was suppressed by the HFD. Gene expression of hypothalamic AgRP and CRH, but not NPY nor POMC, accorded with the energy balance status on long and short photoperiod. However, there was a general dissociation between plasma leptin levels and expression of these hypothalamic energy balance genes. Similarly there was no interaction between the HFD and photoperiod at the level of the genes involved in thyroid hormone metabolism (Dio2, Dio3, TSHß or NMU), which are important mediators of the photoperiodic response. These data suggest that photoperiod and HFD influence body weight and body composition through independent mechanisms but in each case the role of the hypothalamic energy balance genes is not predictable based on their known function.


Subject(s)
Energy Metabolism/physiology , Obesity/metabolism , Photoperiod , Adiposity/physiology , Animals , Body Composition/physiology , Diet, High-Fat , Humans , Hypothalamus/metabolism , Insulin-Like Growth Factor I/metabolism , Leptin/blood , Obesity/blood , Obesity/physiopathology , Rats , Receptors, Neuropeptide/blood , Receptors, Pituitary Hormone-Regulating Hormone/blood
16.
J Neurochem ; 129(3): 366-76, 2014 May.
Article in English | MEDLINE | ID: mdl-24266881

ABSTRACT

The retinoids are a family of compounds that in nature are derived from vitamin A or pro-vitamin A carotenoids. An essential part of the diet for mammals, vitamin A has long been known to be essential for many organ systems in the adult. More recently, however, they have been shown to be necessary for function of the brain and new discoveries point to a central role in processes ranging from neuroplasticity to neurogenesis. Acting in several regions of the central nervous system including the eye, hippocampus and hypothalamus, one common factor in its action is control of biological rhythms. This review summarizes the role of vitamin A in the brain; its action through the metabolite retinoic acid via specific nuclear receptors, and the regulation of its concentration through controlled synthesis and catabolism. The action of retinoic acid to regulate several rhythms in the brain and body, from circadian to seasonal, is then discussed to finish with the importance of retinoic acid in the regular pattern of sleep. We review the role of vitamin A and retinoic acid (RA) as mediators of rhythm in the brain. In the suprachiasmatic nucleus and hippocampus they control expression of circadian clock genes while in the cortex retinoic acid is required for delta oscillations of sleep. Retinoic acid is also central to a second rhythm that keeps pace with the seasons, regulating function in the hypothalamus and pineal gland.


Subject(s)
Brain/physiology , Circadian Rhythm/physiology , Neuronal Plasticity/physiology , Retinoids/metabolism , Animals , Humans
17.
PLoS One ; 8(4): e62003, 2013.
Article in English | MEDLINE | ID: mdl-23637944

ABSTRACT

Exposure to short days (SD) induces profound changes in the physiology and behaviour of Siberian hamsters, including gonadal regression and up to 30% loss in body weight. In a continuous SD environment after approximately 20 weeks, Siberian hamsters spontaneously revert to a long day (LD) phenotype, a phenomenon referred to as the photorefractory response. Previously we have identified a number of genes that are regulated by short photoperiod in the neuropil and ventricular ependymal (VE) cells of the hypothalamus, although their importance and contribution to photoperiod induced physiology is unclear. In this refractory model we hypothesised that the return to LD physiology involves reversal of SD expression levels of key hypothalamic genes to their LD values and thereby implicate genes required for LD physiology. Male Siberian hamsters were kept in either LD or SD for up to 39 weeks during which time SD hamster body weight decreased before increasing, after more than 20 weeks, back to LD values. Brain tissue was collected between 14 and 39 weeks for in situ hybridization to determine hypothalamic gene expression. In VE cells lining the third ventricle, expression of nestin, vimentin, Crbp1 and Gpr50 were down-regulated at 18 weeks in SD photoperiod, but expression was not restored to the LD level in photorefractory hamsters. Dio2, Mct8 and Tsh-r expression were altered by SD photoperiod and were fully restored, or even exceeded values found in LD hamsters in the refractory state. In hypothalamic nuclei, expression of Srif and Mc3r mRNAs was altered at 18 weeks in SD, but were similar to LD expression values in photorefractory hamsters. We conclude that in refractory hamsters not all VE cell functions are required to establish LD physiology. However, thyroid hormone signalling from ependymal cells and reversal of neuronal gene expression appear to be essential for the SD refractory response.


Subject(s)
Ependyma/metabolism , Hypothalamic Hormones/biosynthesis , Hypothalamus/metabolism , Iodide Peroxidase/metabolism , Photoperiod , Seasons , Adaptation, Physiological , Animals , Body Weight/physiology , Cricetinae , Iodide Peroxidase/biosynthesis , Male , Monocarboxylic Acid Transporters/biosynthesis , Nestin/biosynthesis , Phodopus , Receptor, Melanocortin, Type 3/biosynthesis , Receptors, G-Protein-Coupled/biosynthesis , Retinol-Binding Proteins, Cellular/biosynthesis , Somatostatin/biosynthesis , Transcriptome , Vimentin/biosynthesis , Iodothyronine Deiodinase Type II
18.
Stem Cells Dev ; 22(18): 2477-86, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23631375

ABSTRACT

Calcium signals affect many developmental processes, including proliferation, migration, survival, and apoptosis, processes that are of particular importance in stem cells intended for cell replacement therapies. The mechanisms underlying Ca(2+) signals, therefore, have a role in determining how stem cells respond to their environment, and how these responses might be controlled in vitro. In this study, we examined the spontaneous Ca(2+) activity in human neural progenitor cells during proliferation and differentiation. Pharmacological characterization indicates that in proliferating cells, most activity is the result of transient receptor potential (TRP) channels that are sensitive to Gd(3+) and La(3+), with the more subtype selective antagonist Ruthenium red also reducing activity, suggesting the involvement of transient receptor potential vanilloid (TRPV) channels. In differentiating cells, Gd(3+) and La(3+)-sensitive TRP channels also appear to underlie the spontaneous activity; however, no sub-type-specific antagonists had any effect. Protein levels of TRPV2 and TRPV3 decreased in differentiated cells, which is demonstrated by western blot. Thus, it appears that TRP channels represent the main route of Ca(2+) entry in human neural progenitor cells (hNPCs), but the responsible channel types are subject to substitution under differentiating conditions. The level of spontaneous activity could be increased and decreased by lowering and raising the extracellular K(+) concentration. Proliferating cells in low K(+) slowed the cell cycle, with a disproportionate increased percentage of cells in G1 phase and a reduction in S phase. Taken together, these results suggest a link between external K(+) concentration, spontaneous Ca(2+) transients, and cell cycle distribution, which is able to influence the fate of stem and progenitor cells.


Subject(s)
Calcium Channels/metabolism , Neural Stem Cells/metabolism , TRPV Cation Channels/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , G1 Phase/drug effects , Gadolinium/chemistry , Humans , Neural Stem Cells/chemistry , Potassium/metabolism , Ruthenium Red/pharmacology , TRPV Cation Channels/biosynthesis
19.
Trends Neurosci ; 35(12): 733-41, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22959670

ABSTRACT

In the central nervous system (CNS) the function of retinoic acid, the active metabolite of vitamin A, is best understood from its action in guiding embryonic development; as development comes to completion, retinoic acid signaling declines. However, it is increasingly recognized that this signaling mechanism does not disappear in the adult brain but becomes more regionally focused and takes on new roles. These functions are often tied to processes of neural plasticity whether in the hippocampus, through homeostatic neural plasticity, the olfactory bulb or the hypothalamus. The role of retinoic acid in the control of plastic processes has led to suggestions of its involvement in neural disorders, both degenerative and psychiatric. This review presents a snapshot of developments in these areas over recent years.


Subject(s)
Brain/physiology , Neuronal Plasticity/physiology , Tretinoin/physiology , Humans
20.
J Neurochem ; 122(4): 789-99, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22681644

ABSTRACT

Retinoic acid (RA) has been found to regulate hypothalamic function, but precisely where it acts is unknown. This study shows expression of retinaldehyde dehydrogenase (RALDH) enzymes in tanycytes that line the third ventricle in an area overlapping with the site of hypothalamic neural stem cells. The influence of RA was examined on the proliferation of progenitors lining the third ventricle using organotypic slice cultures. As has been shown in other regions of neurogenesis, RA was found to inhibit proliferation. Investigations of the dynamics of RALDH1 expression in the rat hypothalamus have shown that this enzyme is in tanycytes under photoperiodic control with highest levels during long versus short days. In parallel to this shift in RA synthesis, cell proliferation in the third ventricle was found to be lowest during long days when RA was highest, implying that RALDH1 synthesized RA may regulate neural stem cell proliferation. A second RA synthesizing enzyme, RALDH2 was also present in tanycytes lining the third ventricle. In contrast to RALDH1, RALDH2 showed little change with photoperiodicity, but surprisingly the protein was present in the apparent absence of mRNA transcript and it is hypothesized that the endocytic tanycytes may take this enzyme up from the cerebrospinal fluid (CSF).


Subject(s)
Cell Proliferation/drug effects , Hypothalamus/cytology , Hypothalamus/enzymology , Photoperiod , Retinal Dehydrogenase/biosynthesis , Tretinoin/pharmacology , Aldehyde Dehydrogenase 1 Family , Animals , Blotting, Western , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , Hypothalamus/drug effects , Immunohistochemistry , In Situ Hybridization , Isoenzymes/biosynthesis , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Organ Culture Techniques , Polymerase Chain Reaction , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Retinal Dehydrogenase/cerebrospinal fluid , Third Ventricle/cytology , Third Ventricle/drug effects , Third Ventricle/metabolism , Tretinoin/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...