Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
FASEB J ; 37(10): e23189, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37713040

RESUMEN

A protein altering variant in the gene encoding zinc finger homeobox-3 (ZFHX3) has recently been associated with lower BMI in a human genome-wide association study. We investigated metabolic parameters in mice harboring a missense mutation in Zfhx3 (Zfhx3Sci/+ ) and looked for altered in situ expression of transcripts that are associated with energy balance in the hypothalamus to understand how ZFHX3 may influence growth and metabolic effects. One-year-old male and female Zfhx3Sci/+ mice weighed less, had shorter body length, lower fat mass, smaller mesenteric fat depots, and lower circulating insulin, leptin, and insulin-like growth factor-1 (IGF1) concentrations than Zfhx3+/+ littermates. In a second cohort of 9-20-week-old males and females, Zfhx3Sci/+ mice ate less than wildtype controls, in proportion to body weight. In a third cohort of female-only Zfhx3Sci/+ and Zfhx3+/+ mice that underwent metabolic phenotyping from 6 to 14 weeks old, Zfhx3Sci/+ mice weighed less and had lower lean mass and energy expenditure, but fat mass did not differ. We detected increased expression of somatostatin and decreased expression of growth hormone-releasing hormone and growth hormone-receptor mRNAs in the arcuate nucleus (ARC). Similarly, ARC expression of orexigenic neuropeptide Y was decreased and ventricular ependymal expression of orphan G protein-coupled receptor Gpr50 was decreased. We demonstrate for the first time an energy balance effect of the Zfhx3Sci mutation, likely by altering expression of key ARC neuropeptides to alter growth, food intake, and energy expenditure.


Asunto(s)
Genes Homeobox , Proteínas de Homeodominio , Hipotálamo , Mutación Missense , Animales , Femenino , Masculino , Ratones , Expresión Génica , Estudio de Asociación del Genoma Completo , Proteínas de Homeodominio/genética , Hipotálamo/metabolismo , Dedos de Zinc
2.
Mol Psychiatry ; 26(6): 2263-2276, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32203157

RESUMEN

Excessive alcohol intake is associated with 5.9% of global deaths. However, this figure is especially acute in men such that 7.6% of deaths can be attributed to alcohol intake. Previous studies identified a significant interaction between genotypes of the galanin (GAL) gene with anxiety and alcohol abuse in different male populations but were unable to define a mechanism. To address these issues the current study analysed the human UK Biobank cohort and identified a significant interaction (n = 115,865; p = 0.0007) between allelic variation (GG or CA genotypes) in the highly conserved human GAL5.1 enhancer, alcohol intake (AUDIT questionnaire scores) and anxiety in men. Critically, disruption of GAL5.1 in mice using CRISPR genome editing significantly reduced GAL expression in the amygdala and hypothalamus whilst producing a corresponding reduction in ethanol intake in KO mice. Intriguingly, we also found the evidence of reduced anxiety-like behaviour in male GAL5.1KO animals mirroring that seen in humans from our UK Biobank studies. Using bioinformatic analysis and co-transfection studies we further identified the EGR1 transcription factor, that is co-expressed with GAL in amygdala and hypothalamus, as being important in the protein kinase C (PKC) supported activity of the GG genotype of GAL5.1 but less so in the CA genotype. Our unique study uses a novel combination of human association analysis, CRISPR genome editing in mice, animal behavioural analysis and cell culture studies to identify a highly conserved regulatory mechanism linking anxiety and alcohol intake that might contribute to increased susceptibility to anxiety and alcohol abuse in men.


Asunto(s)
Bancos de Muestras Biológicas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Consumo de Bebidas Alcohólicas/genética , Animales , Ansiedad/genética , Etanol , Masculino , Ratones , Reino Unido
3.
J Neuroendocrinol ; 32(2): e12819, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31800973

RESUMEN

The present study investigated neuroanatomically localised changes in de novo DNA methyltransferase expression in the female Siberian hamster (Phodopus sungorus). The objectives were to identify the neuroendocrine substrates that exhibit rhythmic Dnmt3a and Dnmt3b expression across the oestrous cycle and also examine the role of ovarian steroids. Hypothalamic Dnmt3a expression was observed to significantly increase during the transition from pro-oestrous to oestrous. A single bolus injection of diethylstilbestrol and progesterone was sufficient to increase Dnmt3a cell numbers and Dnmt3b immunoreactive intensity in the suprachiasmatic nucleus. In vitro analyses using an embryonic rodent cell line revealed that diethylstilbestrol was sufficient to induce Dnmt3b expression. Up-regulating DNA methylation in vitro reduced the expression of vasoactive intestinal polypeptide, Vip, and the circadian clock gene, Bmal1. Together, these data indicate that ovarian steroids drive de novo DNA methyltransferase expression in the mammalian suprachiasmatic nucleus and increased methylation may regulate genes involved in the circadian timing of oestrous: Vip and Bmal1. Overall, epigenetically mediated neuroendocrine reproductive events may reflect an evolutionarily ancient process involved in the timing of female fertility.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Hormonas Gonadales/metabolismo , Ovario/metabolismo , Núcleo Supraquiasmático/metabolismo , Animales , Relojes Circadianos , Metilación de ADN , Ciclo Estral/metabolismo , Femenino , Sistemas Neurosecretores/metabolismo , Phodopus
4.
Psychoneuroendocrinology ; 109: 104407, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31445429

RESUMEN

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


Asunto(s)
Consumo de Bebidas Alcohólicas/genética , Conducta Adictiva/genética , Receptor Cannabinoide CB1/genética , Animales , Ansiedad/genética , Trastornos de Ansiedad/genética , Encéfalo/metabolismo , Cannabinoides/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Genotipo , Hipocampo/metabolismo , Hipotálamo/metabolismo , Intrones/genética , Desequilibrio de Ligamiento/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Polimorfismo de Nucleótido Simple/genética , Receptor Cannabinoide CB1/metabolismo
5.
Proc Natl Acad Sci U S A ; 116(26): 13116-13121, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31189592

RESUMEN

Synthesis of triiodothyronine (T3) in the hypothalamus induces marked seasonal neuromorphology changes across taxa. How species-specific responses to T3 signaling in the CNS drive annual changes in body weight and energy balance remains uncharacterized. These experiments sequenced and annotated the Siberian hamster (Phodopus sungorus) genome, a model organism for seasonal physiology research, to facilitate the dissection of T3-dependent molecular mechanisms that govern predictable, robust, and long-term changes in body weight. Examination of the Phodopus genome, in combination with transcriptome sequencing of the hamster diencephalon under winter and summer conditions, and in vivo-targeted expression analyses confirmed that proopiomelanocortin (pomc) is a primary genomic target for the long-term T3-dependent regulation of body weight. Further in silico analyses of pomc promoter sequences revealed that thyroid hormone receptor 1ß-binding motif insertions have evolved in several genera of the Cricetidae family of rodents. Finally, experimental manipulation of food availability confirmed that hypothalamic pomc mRNA expression is dependent on longer-term photoperiod cues and is unresponsive to acute, short-term food availability. These observations suggest that species-specific responses to hypothalamic T3, driven in part by the receptor-binding motif insertions in some cricetid genomes, contribute critically to the long-term regulation of energy balance and the underlying physiological and behavioral adaptations associated with the seasonal organization of behavior.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Phodopus/fisiología , Fotoperiodo , Proopiomelanocortina/metabolismo , Aclimatación/fisiología , Animales , Peso Corporal/fisiología , Frío/efectos adversos , Biología Computacional , Regulación hacia Abajo , Ingestión de Alimentos/fisiología , Evolución Molecular , Femenino , Privación de Alimentos/fisiología , Perfilación de la Expresión Génica , Masculino , Anotación de Secuencia Molecular , Neuropéptidos/metabolismo , Proopiomelanocortina/genética , Regiones Promotoras Genéticas/genética , Dominios y Motivos de Interacción de Proteínas/genética , Receptores de Hormona Tiroidea/metabolismo , Estaciones del Año , Especificidad de la Especie , Triyodotironina/administración & dosificación , Triyodotironina/metabolismo , Aumento de Peso/efectos de los fármacos , Aumento de Peso/fisiología , Secuenciación Completa del Genoma
6.
Physiol Behav ; 207: 41-47, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31034840

RESUMEN

This study aimed to determine the relative efficacy of the macronutrients, protein, fat and carbohydrate to induce satiation and satiety in rats in relation to macronutrient activation of neurons in the nucleus of the solitary tract (NTS). Male Sprague Dawley rats were schedule-fed twice a day for 2 h, receiving 100% of daily ad-libitum energy intake. On test day 1, 30 min before the first scheduled meal of the day, rats were gavaged with an 8 kcal isocaloric, isovolumetric solution of a glucose, lipid or peptone macronutrient solution or a non-caloric saline solution. To assess satiation, thirty minutes later rats were given access to food for 2 h and food intake determined. A second 2 h food access period 3 h later was used for assessment of satiety. On the second test day, rats were gavaged as before and killed 90 min after food presentation. Blood was collected for measurement of circulating metabolic markers. Brains were removed for analysis of c-Fos expression by in situ hybridization in the NTS. Rats which received saline consumed a similar amount of food compared to pre-gavage intakes. However, rats gavaged with a caloric macronutrient solution all reduced food intake by 18-20 kcal. Interestingly, the reduction in caloric intake was greater than the caloric value of the macronutrient solution gavaged and was sustained following the second scheduled meal. Quantification by in situ hybridization of c-Fos mRNA expression in the NTS 90 min post-gavage, showed a significant increase with each macronutrient, but was 24-29% higher with a lipid or peptone gavage compared to a glucose gavage. In conclusion, when delivered directly to the stomach, all macronutrients can be equally effective in inducing satiation with significant neuronal activation in the NTS of the hindbrain.


Asunto(s)
Carbohidratos de la Dieta/farmacología , Grasas de la Dieta/farmacología , Proteínas en la Dieta/farmacología , Saciedad/efectos de los fármacos , Respuesta de Saciedad/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Química Encefálica/efectos de los fármacos , Química Encefálica/genética , Carbohidratos de la Dieta/administración & dosificación , Grasas de la Dieta/administración & dosificación , Proteínas en la Dieta/administración & dosificación , Ingestión de Alimentos , Regulación de la Expresión Génica , Masculino , Comidas , Proteínas Proto-Oncogénicas c-fos/biosíntesis , Ratas , Ratas Sprague-Dawley , Núcleo Solitario/citología , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/fisiología
8.
J Neuroendocrinol ; 31(3): e12680, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30585661

RESUMEN

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.


Asunto(s)
Adaptación Fisiológica , Peso Corporal/fisiología , Fenómenos Cronobiológicos , Modelos Neurológicos , Hipófisis/metabolismo , Reproducción , Animales , Regulación del Apetito , Metabolismo Energético , Células Ependimogliales/metabolismo , Humanos , Melatonina/metabolismo , Neurogénesis , Fotoperiodo , Estaciones del Año , Hormonas Tiroideas/metabolismo
9.
Sci Rep ; 8(1): 15648, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30353127

RESUMEN

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.


Asunto(s)
Proteínas de Fase Aguda/genética , Dieta Alta en Grasa/efectos adversos , Endotoxemia/complicaciones , Obesidad/etiología , Serpinas/genética , Receptor Toll-Like 4/inmunología , Regulación hacia Arriba , Animales , Endotoxemia/genética , Endotoxemia/inmunología , Endotoxemia/patología , Microbioma Gastrointestinal , Regulación de la Expresión Génica , Genotipo , Hipotálamo/inmunología , Hipotálamo/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/inmunología , Obesidad/patología , Transducción de Señal , Receptor Toll-Like 4/genética
11.
Appetite ; 117: 234-246, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28687371

RESUMEN

Food structure contributes to the induction of satiation and the maintenance of satiety following intake of a meal. There is evidence from human studies that protein-crosslinking of a milk-protein based meal may enhance satiety, but the mechanism underpinning this effect is unknown. We investigated whether a rat model would respond in a similar manner and might provide mechanistic insight into enhanced satiety by structural modification of a food source. Rats were schedule fed a modified AIN-93M based diet in a liquid form or protein-crosslinked to produce a soft-solid form. This was compared to a modified AIN-93M solid diet. Average daily caloric intake was in the order solid > liquid > crosslinked. Body composition was unaltered in the solid group, but there was a loss of fat in the liquid group and a loss of lean and fat tissue in the crosslinked group. Compared to rats fed a solid diet, acute responses in circulating GLP-1, leptin and insulin were eliminated or attenuated in rats fed a liquid or crosslinked diet. Quantification of homeostatic neuropeptide expression in the hypothalamus showed elevated levels of Npy and Agrp in rats fed the liquid diet. Measurement of food intake after a scheduled meal indicated that reduced energy intake of liquid and crosslinked diets is not due to enhancement of satiety. When continuously available ad-libitum, rats fed a liquid diet showed reduced weight gain despite greater 24 h caloric intake. During the dark phase, caloric intake was reduced, but compensated for during the light phase. We conclude that structural modification from a liquid to a solidified state is beneficial for satiation, with less of a detrimental effect on metabolic parameters and homeostatic neuropeptides.


Asunto(s)
Dieta Reductora , Ingestión de Energía , Hipotálamo/metabolismo , Proteínas de la Leche/administración & dosificación , Neuronas/metabolismo , Respuesta de Saciedad , Transglutaminasas/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Manipulación de Alimentos , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/metabolismo , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Leptina/sangre , Leptina/metabolismo , Masculino , Proteínas de la Leche/efectos adversos , Proteínas de la Leche/química , Proteínas de la Leche/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Sobrepeso/sangre , Sobrepeso/dietoterapia , Sobrepeso/metabolismo , Sobrepeso/prevención & control , Ratas Sprague-Dawley , Aumento de Peso , Pérdida de Peso
12.
J Comp Physiol B ; 187(5-6): 857-868, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28365894

RESUMEN

Thyroid hormones play an important role in regulating seasonal adaptations of mammals. Several studies suggested that reduced availability of 3,3',5-triiodothyronine (T3) in the hypothalamus is required for the physiological adaptation to winter in Djungarian hamsters. We have previously shown that T3 is involved in the regulation of daily torpor, but it remains unclear, whether T3 affects torpor by central or peripheral mechanisms. To determine the effect of T3 concentrations within the hypothalamus in regulating daily torpor, we tested the hypothesis that low hypothalamic T3 metabolism would favour torpor and high T3 concentrations would not. In experiment 1 gene expression in torpid hamsters was assessed for transporters carrying thyroid hormones between cerebrospinal fluid and hypothalamic cells and for deiodinases enzymes, activating or inactivating T3 within hypothalamic cells. Gene expression analysis suggests reduced T3 in hypothalamic cells during torpor. In experiment 2, hypothalamic T3 concentrations were altered via microdialysis and torpor behaviour was continuously monitored by implanted body temperature transmitters. Increased T3 concentrations in the hypothalamus reduced expression of torpor as well as torpor bout duration and depth. Subsequent analysis of gene expression in the ependymal layer of the third ventricle showed clear up-regulation of T3 inactivating deiodinase 3 but no changes in several other genes related to photoperiodic adaptations in hamsters. Finally, serum analysis revealed that increased total T3 serum concentrations were not necessary to inhibit torpor expression. Taken together, our results are consistent with the hypothesis that T3 availability within the hypothalamus significantly contributes to the regulation of daily torpor via a central pathway.


Asunto(s)
Hipotálamo/fisiología , Phodopus/genética , Phodopus/fisiología , Letargo/fisiología , Triyodotironina/fisiología , Animales , Regulación de la Expresión Génica , Masculino , Microdiálisis , Tiroxina/sangre , Tiroxina/fisiología , Triyodotironina/sangre
13.
PLoS One ; 12(2): e0172724, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28235047

RESUMEN

VGF (non-acronymic) was first highlighted to have a role in energy homeostasis through experiments involving dietary manipulation in mice. Fasting increased VGF mRNA in the Arc and levels were subsequently reduced upon refeeding. This anabolic role for VGF was supported by observations in a VGF null (VGF-/-) mouse and in the diet-induced and gold-thioglucose obese mice. However, this anabolic role for VGF has not been supported by a number of subsequent studies investigating the physiological effects of VGF-derived peptides. Intracerebroventricular (ICV) infusion of TLQP-21 increased resting energy expenditure and rectal temperature in mice and protected against diet-induced obesity. Similarly, ICV infusion of TLQP-21 into Siberian hamsters significantly reduced body weight, but this was due to a decrease in food intake, with no effect on energy expenditure. Subsequently NERP-2 was shown to increase food intake in rats via the orexin system, suggesting opposing roles for these VGF-derived peptides. Thus to further elucidate the role of hypothalamic VGF in the regulation of energy homeostasis we utilised a recombinant adeno-associated viral vector to over-express VGF in adult male Siberian hamsters, thus avoiding any developmental effects or associated functional compensation. Initially, hypothalamic over-expression of VGF in adult Siberian hamsters produced no effect on metabolic parameters, but by 12 weeks post-infusion hamsters had increased oxygen consumption and a tendency to increased carbon dioxide production; this attenuated body weight gain, reduced interscapular white adipose tissue and resulted in a compensatory increase in food intake. These observed changes in energy expenditure and food intake were associated with an increase in the hypothalamic contents of the VGF-derived peptides AQEE, TLQP and NERP-2. The complex phenotype of the VGF-/- mice is a likely consequence of global ablation of the gene and its derived peptides during development, as well as in the adult.


Asunto(s)
Peso Corporal/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Neuropéptidos/biosíntesis , Obesidad/tratamiento farmacológico , Aumento de Peso/efectos de los fármacos , Animales , Peso Corporal/fisiología , Cricetinae , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Ratones , Ratones Obesos , Proteínas del Tejido Nervioso/administración & dosificación , Neuropéptidos/administración & dosificación , Neuropéptidos/genética , Obesidad/genética , Obesidad/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Phodopus , Ratas , Aumento de Peso/fisiología
14.
Obes Surg ; 27(1): 177-185, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27576578

RESUMEN

BACKGROUND: Recently, the US FDA has approved "vagal blocking therapy or vBLoc® therapy" as a new treatment for obesity. The aim of the present study was to study the mechanism-of-action of "VBLOC" in rat models. METHODS: Rats were implanted with VBLOC, an intra-abdominal electrical device with leads placed around gastric vagal trunks through an abdominal incision and controlled by wireless device. Body weight, food intake, hunger/satiety, and metabolic parameters were monitored by a comprehensive laboratory animal monitoring system. Brain-gut responses were analyzed physiologically. RESULTS: VBLOC reduced body weight and food intake, which was associated with increased satiety but not with decreased hunger. Brain activities in response to VBLOC included increased gene expression of leptin and CCKb receptors, interleukin-1ß, tumor necrosis factor, and transforming growth factor ß1 in the brainstem; increased CCK, somatostatin, and tyrosine hydroxylase in the hippocampus; increased NPY, AgRP, and Foxa2 in the hypothalamus; and reduced CCKb receptor, melanocortin 4 receptor, and insulin receptor in the hypothalamus. Plasma concentrations of CCK, gastrin, glucagon, GLP-1, and PYY and gastric acid secretion were unchanged in response to VBLOC. CONCLUSIONS: Based on the present study, we may suggest that VBLOC induces satiety through vagal signaling, leading to reduced food intake and loss of body weight.

16.
Scand J Gastroenterol ; 52(4): 442-449, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27996337

RESUMEN

OBJECTIVE: We wanted to exam the steady-state energy balance by using high-fat diet-induced obese (DIO) rats and mice as models for positive energy balance, and gastric bypassed (GB) rats and gene knockout of muscarinic acetylcholine M3 receptor (M3KO) mice as models for negative energy balance. METHODS: One hundred and thirty-two rats and mice were used. Energy balance was measured by a comprehensive laboratory animal monitoring system. Gene expression was analysed by in situ hybridisation in M3KO mice. RESULTS: DIO rats reached the plateau of body weight 28 weeks after starting high-fat diet (25% heavier than controls), whereas DIO mice reached the plateau after 6 weeks (23% heavier than controls). At the plateau, DIO rats had higher calorie intake during the light phase but not during the dark phase, while mice had the same calorie intake per day as controls. DIO rats and mice had lower energy expenditure (EE) and respiratory exchange ratio (RER) than controls. GB-rats reached the plateau (15% weight loss) 2 weeks after surgery and had the same calorie intake as sham-operated controls. EE, but not RER, was higher in GB rats than controls during the dark phase. The lean M3KO mice (25% lighter than wild-type (WT) mice at the plateau between 6 and 15 months of age) had the same calorie intake but higher EE, RER and hypothalamic mRNA expression of NPY, AgRP and leptin receptor than WT mice. CONCLUSION: When body weight gain or loss reached a plateau, the steady-state energy balance was mainly maintained by EE and/or RER rather than calorie intake.


Asunto(s)
Metabolismo Energético , Obesidad/metabolismo , Obesidad/cirugía , Pérdida de Peso , Animales , Encéfalo/patología , Grasas de la Dieta , Modelos Animales de Enfermedad , Femenino , Derivación Gástrica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M3/genética
17.
Dis Model Mech ; 9(11): 1245-1255, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27821603

RESUMEN

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.


Asunto(s)
Investigación Biomédica , Obesidad/patología , Animales , Dieta , Modelos Animales de Enfermedad , Humanos , Modelos Genéticos , Obesidad/genética , Investigación Biomédica Traslacional
18.
Sci Rep ; 6: 29689, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27406810

RESUMEN

In nature Siberian hamsters utilize the decrement in day length following the summer solstice to implement physiological adaptations in anticipation of the forthcoming winter, but also exploit an intrinsic interval timer to initiate physiological recrudescence following the winter solstice. However, information is lacking on the temporal dynamics in natural photoperiod of photoperiodically regulated genes and their relationship to physiological adaptations. To address this, male Siberian hamsters born and maintained outdoors were sampled every month over the course of one year. As key elements of the response to photoperiod, thyroid hormone signalling components were assessed in the hypothalamus. From maximum around the summer solstice (late-June), Dio2 expression rapidly declined in advance of physiological adaptations. This was followed by a rapid increase in Mct8 expression (T3/T4 transport), peaking early-September before gradually declining to minimum expression by the following June. Dio3 showed a transient peak of expression beginning late-August. A recrudescence of testes and body mass occurred from mid-February, but Dio2 expression remained low until late-April of the following year, converging with the time of year when responsiveness to short-day length is re-established. Other photoperiodically regulated genes show temporal regulation, but of note is a transient peak in Gpr50 around late-July.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hipotálamo/metabolismo , Fotoperiodo , Estaciones del Año , Hormonas Tiroideas/biosíntesis , Animales , Cricetinae , Femenino , Masculino , Phodopus , Hormonas Tiroideas/genética
19.
J Mol Endocrinol ; 56(2): 123-34, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26643910

RESUMEN

The Siberian hamster (Phodopus sungorus) survives winter by decreasing food intake and catabolizing abdominal fat reserves, resulting in a sustained, profound loss of body weight. Hypothalamic tanycytes are pivotal for this process. In these cells, short-winter photoperiods upregulate deiodinase 3, an enzyme that regulates thyroid hormone availability, and downregulate genes encoding components of retinoic acid (RA) uptake and signaling. The aim of the current studies was to identify mechanisms by which seasonal changes in thyroid hormone and RA signaling from tanycytes might ultimately regulate appetite and energy expenditure. proVGF is one of the most abundant peptides in the mammalian brain, and studies have suggested a role for VGF-derived peptides in the photoperiodic regulation of body weight in the Siberian hamster. In silico studies identified possible thyroid and vitamin D response elements in the VGF promoter. Using the human neuroblastoma SH-SY5Y cell line, we demonstrate that RA increases endogenous VGF expression (P<0.05) and VGF promoter activity (P<0.0001). Similarly, treatment with 1,25-dihydroxyvitamin D3 increased endogenous VGF mRNA expression (P<0.05) and VGF promoter activity (P<0.0001), whereas triiodothyronine (T3) decreased both (P<0.01 and P<0.0001). Finally, intra-hypothalamic administration of T3 blocked the short day-induced increase in VGF expression in the dorsomedial posterior arcuate nucleus of Siberian hamsters. Thus, we conclude that VGF expression is a likely target of photoperiod-induced changes in tanycyte-derived signals and is potentially a regulator of seasonal changes in appetite and energy expenditure.


Asunto(s)
Calcitriol/farmacología , Factores de Crecimiento Nervioso/metabolismo , Activación Transcripcional , Triyodotironina/fisiología , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Cricetinae , Expresión Génica , Humanos , Masculino , Factores de Crecimiento Nervioso/genética , Phodopus , Regiones Promotoras Genéticas , Triyodotironina/farmacología
20.
Curr Biol ; 25(22): 2997-3003, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26549257

RESUMEN

Hypothalamic tanycytes are considered to function as sensors of peripheral metabolism. To facilitate this role, they express a wide range of receptors, including fibroblast growth factor receptor 1 (FGFR1). Using a monoclonal antibody (IMC-H7) that selectively antagonizes the FGFR1c isoform, we investigated possible actions of FGFR1c in a natural animal model of adiposity, the Siberian hamster. Infusion of IMC-H7 into the third ventricle suppressed appetite and increased energy expenditure. Likewise, peripheral treatment with IMC-H7 decreased appetite and body weight and increased energy expenditure and fat oxidation. A greater reduction in body weight and caloric intake was observed in response to IMC-H7 during the long-day fat state as compared to the short-day lean state. This enhanced response to IMC-H7 was also observed in calorically restricted hamsters maintained in long days, suggesting that it is the central photoperiodic state rather than the peripheral adiposity that determines the response to FGFR1c antagonism. Hypothalamic thyroid hormone availability is controlled by deiodinase enzymes (DIO2 and DIO3) expressed in tanycytes and is the key regulator of seasonal cycles of energy balance. Therefore, we determined the effect of IMC-H7 on hypothalamic expression of these deiodinase enzymes. The reductions in food intake and body weight were always associated with decreased expression of DIO2 in the hypothalamic ependymal cell layer containing tanycytes. These data provide further support for the notion the tanycytes are an important component of the mechanism by which the hypothalamus integrates central and peripheral signals to regulate energy intake and expenditure.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Ingestión de Alimentos/efectos de los fármacos , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/antagonistas & inhibidores , Adiposidad/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Ritmo Circadiano/fisiología , Cricetinae , Hipotálamo/metabolismo , Masculino , Modelos Animales , Phodopus , Fotoperiodo , Isoformas de Proteínas/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/inmunología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Delgadez/metabolismo , Hormonas Tiroideas/metabolismo , Pérdida de Peso/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA