Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Appetite ; 174: 106031, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35395362

RESUMEN

Behavioral plasticity refers to changes occurring due to external influences on an organism, including adaptation, learning, memory and enduring influences from early life experience. There are 2 types of behavioral plasticity: "developmental", which refers to gene/environment interactions affecting a phenotype, and "activational" which refers to innate physiology and can involve structural physiological changes of the body. In this review, we focus on feeding behavior, and studies involving neuropeptides that influence behavioral plasticity - primarily opioids, orexin, neuropeptide Y, and oxytocin. In each section of the review, we include examples of behavioral plasticity as it relates to actions of these neuropeptides. It can be concluded from this review that eating behavior is influenced by a number of external factors, including time of day, type of food available, energy balance state, and stressors. The reviewed work underscores that environmental factors play a critical role in feeding behavior and energy balance, but changes in eating behavior also result from a multitude of non-environmental factors, such that there can be no single mechanism or variable that can explain ingestive behavior.


Asunto(s)
Neuropéptidos , Conducta Alimentaria/fisiología , Humanos , Neuropéptido Y/genética , Oxitocina
2.
PLoS Genet ; 15(12): e1008455, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31800589

RESUMEN

SLC18B1 is a sister gene to the vesicular monoamine and acetylcholine transporters, and the only known polyamine transporter, with unknown physiological role. We reveal that Slc18b1 knock out mice has significantly reduced polyamine content in the brain providing the first evidence that Slc18b1 is functionally required for regulating polyamine levels. We found that this mouse has impaired short and long term memory in novel object recognition, radial arm maze and self-administration paradigms. We also show that Slc18b1 KO mice have altered expression of genes involved in Long Term Potentiation, plasticity, calcium signalling and synaptic functions and that expression of components of GABA and glutamate signalling are changed. We further observe a partial resistance to diazepam, manifested as significantly lowered reduction in locomotion after diazepam treatment. We suggest that removal of Slc18b1 leads to reduction of polyamine contents in neurons, resulting in reduced GABA signalling due to long-term reduction in glutamatergic signalling.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Transporte de Catión/genética , Memoria a Largo Plazo , Memoria a Corto Plazo , Poliaminas/metabolismo , Animales , Señalización del Calcio , Técnicas de Inactivación de Genes , Ácido Glutámico/metabolismo , Aprendizaje por Laberinto , Ratones , Plasticidad Neuronal , Ácido gamma-Aminobutírico/metabolismo
3.
BMC Genomics ; 20(1): 281, 2019 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-30971208

RESUMEN

BACKGROUND: Ketamine ester analogs, SN 35210 and SN 35563, demonstrate different pharmacological profiles to ketamine in animal models. Both confer hypnosis with predictably rapid offset yet, paradoxically, SN35563 induces a prolonged anti-nociceptive state. To explore underlying mechanisms, broad transcriptome changes were measured and compared across four relevant target regions of the rat brain. RESULTS: SN 35563 produced large-scale alteration of gene expression in the Basolateral Amygdala (BLA) and Paraventricular Nucleus of the Thalamus (PVT), in excess of 10x that induced by ketamine and SN 35210. A smaller and quantitatively similar number of gene changes were observed in the Insula (INS) and Nucleus Accumbens (ACB) for all three agents. In the BLA and PVT, SN 35563 caused enrichment for gene pathways related to the function and structure of glutamatergic synapses in respect to: release of neurotransmitter, configuration of postsynaptic AMPA receptors, and the underlying cytoskeletal scaffolding and alignment. CONCLUSION: The analgesic ketamine ester analog SN 35563 induces profound large-scale changes in gene expression in key pain-related brain regions reflecting its unique prolonged pharmacodynamic profile.


Asunto(s)
Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ésteres/química , Ketamina/análogos & derivados , Ketamina/farmacología , Transcripción Genética/efectos de los fármacos , Animales , Femenino , Redes Reguladoras de Genes/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
4.
Nat Rev Neurosci ; 15(2): 98-110, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24588018

RESUMEN

The melanocortin system has a well-established role in the regulation of energy homeostasis, but there is growing evidence of its involvement in memory, nociception, mood disorders and addiction. In this Review, we focus on the role of the melanocortin 4 receptor and provide an integrative view of the molecular mechanisms that lead to melanocortin-induced changes in synaptic plasticity within these diverse physiological systems. We also highlight the importance of melanocortin peptides and receptors in chronic pain syndromes, memory impairments, depression and drug abuse, and the possibility of targeting them for therapeutic purposes.


Asunto(s)
Melanocortinas/metabolismo , Receptor de Melanocortina Tipo 4/fisiología , Transducción de Señal/fisiología , Sinapsis/fisiología , Transmisión Sináptica/fisiología , Animales , Moléculas de Adhesión Celular Neuronal , Trastorno Depresivo/tratamiento farmacológico , Trastorno Depresivo/metabolismo , Metabolismo Energético/fisiología , Humanos , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/metabolismo , Dolor/tratamiento farmacológico , Dolor/metabolismo
5.
Appetite ; 125: 278-286, 2018 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-29471071

RESUMEN

Human and laboratory animal studies suggest that dietary supplementation of a free essential amino acid, l-tryptophan (TRP), reduces food intake. It is unclear whether an acute gastric preload of TRP decreases consumption and whether central mechanisms underlie TRP-driven hypophagia. We examined the effect of TRP administered via intragastric gavage on energy- and palatability-induced feeding in mice. We sought to identify central mechanisms through which TRP suppresses appetite. Effects of TRP on consumption of energy-dense and energy-dilute tastants were established in mice stimulated to eat by energy deprivation or palatability. A conditioned taste aversion (CTA) paradigm was used to assess whether hypophagia is unrelated to sickness. c-Fos immunohistochemistry was employed to detect TRP-induced activation of feeding-related brain sites and of oxytocin (OT) neurons, a crucial component of satiety circuits. Also, expression of OT mRNA was assessed with real-time PCR. The functional importance of OT in mediating TRP-driven hypophagia was substantiated by showing the ability of OT receptor blockade to abolish TRP-induced decrease in feeding. TRP reduced intake of energy-dense standard chow in deprived animals and energy-dense palatable chow in sated mice. Anorexigenic doses of TRP did not cause a CTA. TRP failed to affect intake of palatable yet calorie-dilute or noncaloric solutions (10% sucrose, 4.1% Intralipid or 0.1% saccharin) even for TRP doses that decreased water intake in thirsty mice. Fos analysis revealed that TRP increases activation of several key feeding-related brain areas, especially in the brain stem and hypothalamus. TRP activated hypothalamic OT neurons and increased OT mRNA levels, whereas pretreatment with an OT antagonist abolished TRP-driven hypophagia. We conclude that intragastric TRP decreases food and water intake, and TRP-induced hypophagia is partially mediated via central circuits that encompass OT.


Asunto(s)
Apetito/efectos de los fármacos , Encéfalo/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Oxitocina/agonistas , Triptófano/farmacología , Animales , Encéfalo/citología , Grasas de la Dieta/administración & dosificación , Sacarosa en la Dieta/administración & dosificación , Suplementos Dietéticos , Ingestión de Líquidos/efectos de los fármacos , Privación de Alimentos , Lípidos/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Receptores de Oxitocina/metabolismo , Sacarina/administración & dosificación , Respuesta de Saciedad/efectos de los fármacos , Edulcorantes/administración & dosificación , Gusto , Sed , Agua
6.
BMC Neurosci ; 16: 13, 2015 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-25887538

RESUMEN

BACKGROUND: Obesity is a growing global concern with strong associations with cardiovascular disease, cancer and type-2 diabetes. Although various genome-wide association studies have identified more than 40 genes associated with obesity, these genes cannot fully explain the heritability of obesity, suggesting there may be other contributing factors, including epigenetic effects. RESULTS: We performed genome wide DNA methylation profiling comparing normal-weight and obese 9-13 year old children to investigate possible epigenetic changes correlated with obesity. Of note, obese children had significantly lower methylation levels at a CpG site located near coronin 7 (CORO7), which encodes a tryptophan-aspartic acid dipeptide (WD)-repeat containing protein most likely involved in Golgi complex morphology and function. Anatomical profiling of coronin 7 (Coro7) mRNA expression in mice revealed that it is highly expressed in appetite and energy balance regulating regions, including the hypothalamus, striatum and locus coeruleus, the main noradrenergic brain site. Interestingly, we found that food deprivation in mice downregulates hypothalamic Coro7 mRNA levels, and injecting ethanol, an appetite stimulant, increased the number of Coro7 expressing cells in the locus coeruleus. Finally, by employing the genetically-tractable Drosophila melanogaster model we were able to demonstrate an evolutionarily conserved metabolic function for the CORO7 homologue pod1. Knocking down the pod1 in the Drosophila adult nervous system increased their resistance to starvation. Furthermore, feeding flies a high-calorie diet significantly increased pod1 expression. CONCLUSION: We conclude that coronin 7 is involved in the regulation of energy homeostasis and this role stems, to some degree, from the effect on feeding for calories and reward.


Asunto(s)
Peso Corporal/fisiología , Proteínas de Drosophila/metabolismo , Proteínas de Microfilamentos/metabolismo , Adolescente , Animales , Estimulantes del Apetito/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Niño , Estudios de Cohortes , Dieta Alta en Grasa , Proteínas de Drosophila/genética , Drosophila melanogaster , Etanol/farmacología , Femenino , Privación de Alimentos/fisiología , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones Endogámicos C57BL , Proteínas de Microfilamentos/genética , Obesidad/genética , ARN Mensajero/metabolismo , Inanición/metabolismo
7.
Appetite ; 86: 81-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25453587

RESUMEN

Research on oxytocin (OT) has yielded two seemingly unrelated sets of discoveries: OT has prosocial effects, and it elicits termination of feeding, especially of food rich in carbohydrates. Here we investigated whether OT's involvement in food intake is affected by the social context in mice, with particular focus on the role of dominance. We used two approaches: injections and gene expression analysis. We housed two males per cage and determined a dominant one. Then we injected a blood-brain barrier penetrant OT receptor antagonist L-368,899 in either dominant or subordinate animals and gave them 10-min access to a sucrose solution in the apparatus in which social exposure was modified and it ranged from none to unrestricted contact. L-368,899 increased the amount of consumed sugar in dominant mice regardless of whether these animals had access to sucrose in the non-social or social contexts (olfactory-derived or partial social exposure). The antagonist also increased the proportion of time that dominant mice spent drinking the sweet solution in the paradigm in which both mice had to share a single source of sucrose. L-368,899-treated subordinate mice consumed more sucrose solution than saline controls only when the environment in which sugar was presented was devoid of social cues related to the dominant animal. Finally, we investigated whether hypothalamic OT gene expression differs between dominant and subordinate mice consuming sugar and found OT mRNA levels to be higher in dominant mice. We conclude that social context and dominance affect OT's effect on appetite for sucrose.


Asunto(s)
Apetito/efectos de los fármacos , Canfanos/administración & dosificación , Sacarosa en la Dieta , Preferencias Alimentarias/efectos de los fármacos , Piperazinas/administración & dosificación , Receptores de Oxitocina/antagonistas & inhibidores , Conducta Social , Animales , Conducta Animal , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/psicología , Preferencias Alimentarias/psicología , Expresión Génica , Hipotálamo/química , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oxitocina/genética , Oxitocina/fisiología , ARN Mensajero/análisis , Predominio Social , Soluciones , Sacarosa
8.
PLoS Genet ; 8(3): e1002568, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22438821

RESUMEN

Neurobeachin (Nbea) regulates neuronal membrane protein trafficking and is required for the development and functioning of central and neuromuscular synapses. In homozygous knockout (KO) mice, Nbea deficiency causes perinatal death. Here, we report that heterozygous KO mice haploinsufficient for Nbea have higher body weight due to increased adipose tissue mass. In several feeding paradigms, heterozygous KO mice consumed more food than wild-type (WT) controls, and this consumption was primarily driven by calories rather than palatability. Expression analysis of feeding-related genes in the hypothalamus and brainstem with real-time PCR showed differential expression of a subset of neuropeptide or neuropeptide receptor mRNAs between WT and Nbea+/- mice in the sated state and in response to food deprivation, but not to feeding reward. In humans, we identified two intronic NBEA single-nucleotide polymorphisms (SNPs) that are significantly associated with body-mass index (BMI) in adult and juvenile cohorts. Overall, data obtained in mice and humans suggest that variation of Nbea abundance or activity critically affects body weight, presumably by influencing the activity of feeding-related neural circuits. Our study emphasizes the importance of neural mechanisms in body weight control and points out NBEA as a potential risk gene in human obesity.


Asunto(s)
Índice de Masa Corporal , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Conducta Alimentaria , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Obesidad/genética , Tejido Adiposo/metabolismo , Adolescente , Animales , Tronco Encefálico/metabolismo , Niño , Privación de Alimentos , Regulación de la Expresión Génica/genética , Estudios de Asociación Genética , Humanos , Hipotálamo/metabolismo , Masculino , Proteínas de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple
9.
Front Neuroendocrinol ; 33(2): 127-39, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22305720

RESUMEN

Food reward, not hunger, is the main driving force behind eating in the modern obesogenic environment. Palatable foods, generally calorie-dense and rich in sugar/fat, are thus readily overconsumed despite the resulting health consequences. Important advances have been made to explain mechanisms underlying excessive consumption as an immediate response to presentation of rewarding tastants. However, our understanding of long-term neural adaptations to food reward that oftentimes persist during even a prolonged absence of palatable food and contribute to the reinstatement of compulsive overeating of high-fat high-sugar diets, is much more limited. Here we discuss the evidence from animal and human studies for neural and molecular adaptations in both homeostatic and non-homeostatic appetite regulation that may underlie the formation of a "feed-forward" system, sensitive to palatable food and propelling the individual from a basic preference for palatable diets to food craving and compulsive, addiction-like eating behavior.


Asunto(s)
Adaptación Fisiológica/genética , Conducta Adictiva , Retroalimentación Fisiológica/fisiología , Hiperfagia/genética , Hiperfagia/fisiopatología , Animales , Conducta Adictiva/etiología , Conducta Adictiva/genética , Conducta Adictiva/fisiopatología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Conducta Compulsiva/etiología , Conducta Compulsiva/genética , Conducta Compulsiva/fisiopatología , Humanos , Hiperfagia/psicología , Modelos Cardiovasculares , Recompensa , Transducción de Señal/fisiología , Trastornos Relacionados con Sustancias/genética , Trastornos Relacionados con Sustancias/fisiopatología
10.
Physiol Behav ; 271: 114318, 2023 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-37543105

RESUMEN

Peripheral and central injections of oxytocin (OT) in laboratory animals decrease eating for energy and palatability, but the hypophagic response is dependent on the administration route. Human studies rely on intranasal (IN) administration of the peptide, the route underutilized in OT animal feeding studies thus far. Therefore, we examined the effect of IN OT on various aspects of food consumption in rats: (a) overnight deprivation-induced standard chow intake, (b) episodic (2-h) consumption of calorie-dense and palatable high-fat high-sugar (HFHS) chow, (c) 2-h episodic intake of palatable and calorie-dilute sucrose and Intralipid solutions, and (d) 2-h sucrose solution intake in rats habituated to ingesting this solution daily for several weeks. Finally, we assessed c-Fos changes in response to the acute IN OT administration in rats habituated to daily sugar consumption. We found that IN 20µg OT decreased deprivation-induced intake of standard chow and HFHS chow in nondeprived rats without affecting water consumption. IN OT also reduced 2-hour episodic fluid consumption of sucrose, but not Intralipid. In the habitual sugar consumption paradigm, acute IN OT diminished sucrose solution intake in animals accustomed to the 2-hour/day sucrose meal regimen. In rats habitually consuming sucrose, IN OT altered c-Fos immunoreactivity in brain areas related to energy homeostasis and reward, including the central nucleus of the amygdala, the hypothalamic paraventricular and the arcuate nuclei. We conclude that IN OT is an effective appetite suppressant for carbohydrate/sugar diets in rats and its effects involve feeding-related brain circuits.

11.
BMC Gastroenterol ; 12: 134, 2012 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-23009096

RESUMEN

BACKGROUND: G protein-coupled receptors (GPCRs) represent one of the largest families of transmembrane receptors and the most common drug target. The Adhesion subfamily is the second largest one of GPCRs and its several members are known to mediate neural development and immune system functioning through cell-cell and cell-matrix interactions. The distribution of these receptors has not been characterized in detail in the gastrointestinal (GI) tract. Here we present the first comprehensive anatomical profiling of mRNA expression of all 30 Adhesion GPCRs in the rat GI tract divided into twelve subsegments. METHODS: Using RT-qPCR, we studied the expression of Adhesion GPCRs in the esophagus, the corpus and antrum of the stomach, the proximal and distal parts of the duodenum, ileum, jejunum and colon, and the cecum. RESULTS: We found that twenty-one Adhesion GPCRs (70%) had a widespread (expressed in five or more segments) or ubiquitous (expressed in eleven or more segments) distribution, seven (23%) were restricted to a few segments of the GI tract and two were not expressed in any segment. Most notably, almost all Group III members were ubiquitously expressed, while the restricted expression was characteristic for the majority of group VII members, hinting at more specific/localized roles for some of these receptors. CONCLUSIONS: Overall, the distribution of Adhesion GPCRs points to their important role in GI tract functioning and defines them as a potentially crucial target for pharmacological interventions.


Asunto(s)
Tracto Gastrointestinal/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Animales , Masculino , Filogenia , Ratas , Receptores Acoplados a Proteínas G/genética
12.
Genes (Basel) ; 13(2)2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35205303

RESUMEN

A core yet understudied symptom of autism is aberrant eating behaviour, including extremely narrow food preferences. Autistic individuals often refuse to eat despite hunger unless preferred food is given. We hypothesised that, apart from aberrant preference, underfeeding stems from abnormal hunger processing. Utilising an adult male VPA rat, a model of autism, we examined intake of 'bland' chow in animals maintained on this diet continuously, eating this food after fasting and after both food and water deprivation. We assessed body weight in adulthood to determine whether lower feeding led to slower growth. Since food intake is highly regulated by brain processes, we looked into the activation (c-Fos immunoreactivity) of central sites controlling appetite in animals subjected to food deprivation vs. fed ad libitum. Expression of genes involved in food intake in the hypothalamus and brain stem, regions responsible for energy balance, was measured in deprived vs. sated animals. We performed our analyses on VPAs and age-matched healthy controls. We found that VPAs ate less of the 'bland' chow when fed ad libitum and after deprivation than controls did. Their body weight increased more slowly than that of controls when maintained on the 'bland' food. While hungry controls had lower c-Fos IR in key feeding-related areas than their ad libitum-fed counterparts, in hungry VPAs c-Fos was unchanged or elevated compared to the fed ones. The lack of changes in expression of feeding-related genes upon deprivation in VPAs was in contrast to several transcripts affected by fasting in healthy controls. We conclude that hunger processing is dysregulated in the VPA rat.


Asunto(s)
Trastorno Autístico , Ingestión de Alimentos , Animales , Trastorno Autístico/inducido químicamente , Trastorno Autístico/genética , Peso Corporal , Ingestión de Alimentos/genética , Expresión Génica , Masculino , Proteínas Proto-Oncogénicas c-fos/genética , Ratas , Ácido Valproico/efectos adversos
13.
J Neuroendocrinol ; 34(9): e13106, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35192207

RESUMEN

The neuropeptide oxytocin (OT) has emerged as an important anorexigen in the regulation of food intake and energy balance. It has been shown that the release of OT and activation of hypothalamic OT neurons coincide with food ingestion. Its effects on feeding have largely been attributed to limiting meal size through interactions in key regulatory brain regions governing the homeostatic control of food intake such as the hypothalamus and hindbrain in addition to key feeding reward areas such as the nucleus accumbens and ventral tegmental area. Furthermore, the magnitude of an anorexigenic response to OT and feeding-related activation of the brain OT circuit are modified by the composition and flavor of a diet, as well as by a social context in which a meal is consumed. OT is particularly effective in reducing consumption of carbohydrates and sweet tastants. Pharmacologic, genetic, and pair-feeding studies indicate that OT-elicited weight loss cannot be fully explained by reductions of food intake and that the overall impact of OT on energy balance is also partly a result of OT-elicited changes in lipolysis, energy expenditure, and glucose regulation. Peripheral administration of OT mimics many of its effects when it is given into the central nervous system, raising the questions of whether and to what extent circulating OT acts through peripheral OT receptors to regulate energy balance. Although OT has been found to elicit weight loss in female mice, recent studies have indicated that sex and estrous cycle may impact oxytocinergic modulation of food intake. Despite the overall promising basic research data, attempts to use OT in the clinical setting to combat obesity and overeating have generated somewhat mixed results. The focus of this mini-review is to briefly summarize the role of OT in feeding and metabolism, address gaps and inconsistencies in our knowledge, and discuss some of the limitations to the potential use of chronic OT that should help guide future research on OT as a tailor-made anti-obesity therapeutic.


Asunto(s)
Ingestión de Alimentos , Oxitocina , Animales , Carbohidratos/farmacología , Carbohidratos/uso terapéutico , Femenino , Glucosa/farmacología , Ratones , Obesidad/tratamiento farmacológico , Oxitocina/fisiología , Receptores de Oxitocina/metabolismo , Pérdida de Peso
14.
Foods ; 11(2)2022 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-35053873

RESUMEN

The natural 20:80 whey:casein ratio in cow's milk (CM) for adults and infants is adjusted to reflect the 60:40 ratio of human milk, but the feeding and metabolic consequences of this adjustment have been understudied. In adult human subjects, the 60:40 CM differently affects glucose metabolism and hormone release than the 20:80 CM. In laboratory animals, whey-adapted goat's milk is consumed in larger quantities. It is unknown whether whey enhancement of CM would have similar consequences on appetite and whether it would affect feeding-relevant brain regulatory mechanisms. In this set of studies utilizing laboratory mice, we found that the 60:40 CM was consumed more avidly than the 20:80 control formulation by animals motivated to eat by energy deprivation and by palatability (in the absence of hunger) and that this hyperphagia stemmed from prolongation of the meal. Furthermore, in two-bottle choice paradigms, whey-adapted CM was preferred against the natural 20:80 milk. The intake of the whey-adapted CM induced neuronal activation (assessed through analysis of c-Fos expression in neurons) in brain sites promoting satiation, but importantly, this activation was less pronounced than after ingestion of the natural 20:80 whey:casein CM. Activation of hypothalamic neurons synthesizing anorexigenic neuropeptide oxytocin (OT) was also less robust after the 60:40 CM intake than after the 20:80 CM. Pharmacological blockade of the OT receptor in mice led to an increase in the consumption only of the 20:80 CM, thus, of the milk that induced greater activation of OT neurons. We conclude that the whey-adapted CM is overconsumed compared to the natural 20:80 CM and that this overconsumption is associated with weakened responsiveness of central networks involved in satiety signalling, including OT.

15.
Nutrients ; 14(5)2022 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-35267900

RESUMEN

The opioid antagonist naltrexone (NTX) decreases intake of preferred diets in rats at very low doses relative to doses needed to decrease intake of "bland" laboratory chow. In the absence of an opioid agonist, NTX is not discriminable using operant techniques. In the current study, we found that rats given intermittent access to a 25% sucrose solution learned to discriminate between various naltrexone doses and saline. None of the rats given only water learned to discriminate between naltrexone and saline. When access to the sucrose solution was discontinued for 14 days, the rats lost the ability to discriminate between NTX and saline. We also studied the changes of c-Fos IR in selected brain regions in rats treated with saline versus NTX that were drinking water or 25% sucrose. An injection of NTX or saline resulted in a significant drug, diet, and interaction effect in various brain regions associated with feeding behavior, particularly the amygdala, accumbens, and hypothalamic sites. Thus, we found that ingestion of a sucrose solution results in the ability of rats to reliably discriminate naltrexone administration. In addition, sucrose and naltrexone altered c-Fos IR in an interactive fashion in brain regions known to be involved in ingestion behavior.


Asunto(s)
Naltrexona , Receptores Opioides , Animales , Conducta Alimentaria , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Ratas , Sacarosa/farmacología
16.
Cells ; 11(6)2022 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-35326421

RESUMEN

The statin drug target, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), is strongly linked to body mass index (BMI), yet how HMGCR influences BMI is not understood. In mammals, studies of peripheral HMGCR have not clearly identified a role in BMI maintenance and, despite considerable central nervous system expression, a function for central HMGCR has not been determined. Similar to mammals, Hmgcr is highly expressed in the Drosophila melanogaster brain. Therefore, genetic and pharmacological studies were performed to identify how central Hmgcr regulates Drosophila energy metabolism and feeding behavior. We found that inhibiting Hmgcr, in insulin-producing cells of the Drosophila pars intercerebralis (PI), the fly hypothalamic equivalent, significantly reduces the expression of insulin-like peptides, severely decreasing insulin signaling. In fact, reducing Hmgcr expression throughout development causes decreased body size, increased lipid storage, hyperglycemia, and hyperphagia. Furthermore, the Hmgcr induced hyperphagia phenotype requires a conserved insulin-regulated α-glucosidase, target of brain insulin (tobi). In rats and mice, acute inhibition of hypothalamic Hmgcr activity stimulates food intake. This study presents evidence of how central Hmgcr regulation of metabolism and food intake could influence BMI.


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Animales , Drosophila melanogaster/metabolismo , Ingestión de Alimentos , Metabolismo Energético , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Hiperfagia , Insulina/metabolismo , Mamíferos/metabolismo , Ratones , Ratas
17.
Biochem Biophys Res Commun ; 411(4): 702-7, 2011 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-21781957

RESUMEN

Solute carriers (SLCs), the second largest super-family of membrane proteins in the human genome, transport amino acids, sugars, fatty acids, inorganic ions, essential metals and drugs over membranes. To date no study has provided a comprehensive analysis of SLC localization along the entire GI tract. The aim of the present study was to provide a comprehensive, segment-specific description of the localization of SLC genes along the rat GI tract by employing bioinformatics and molecular biology methods. The Unigene database was screened for rat SLC entries in the intestinal tissue. Using qPCR we measured expression of the annotated genes in the GI tract divided into the following segments: the esophagus, the corpus and the antrum of the stomach, the proximal and distal parts of the duodenum, ileum, jejunum and colon, and the cecum. Our Unigene-derived gene pool was expanded with data from in-house tissue panels and a literature search. We found 44 out of 78 (56%) of gut SLC transcripts to be expressed in all GI tract segments, whereas the majority of remaining SLCs were detected in more than five segments. SLCs are predominantly expressed in gut regions with absorptive functions although expression was also found in segments unrelated to absorption. The proximal jejunum had the highest number of differentially expressed SLCs. In conclusion, SLCs are a crucial molecular component of the GI tract, with many of them expressed along the entire GI tract. This work presents the first overall road map of localization of transporter genes in the GI tract.


Asunto(s)
Proteínas de Transporte de Catión/genética , Tracto Gastrointestinal/metabolismo , Perfilación de la Expresión Génica , Animales , Masculino , Ratas , Ratas Endogámicas
18.
Biochem Biophys Res Commun ; 408(3): 422-6, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21514276

RESUMEN

Single nucleotide polymorphisms in the fat mass and obesity-associated (FTO) gene have been associated with obesity in humans. Alterations in Fto expression in transgenic animals affect body weight, energy expenditure and food intake. Fto, a nuclear protein and proposed transcription co-factor, has been speculated to affect energy balance through a functional relationship with specific genes encoding feeding-related peptides. Herein, we employed double immunohistochemistry and showed that the majority of neurons synthesizing a satiety mediator, oxytocin, coexpress Fto in the brain of male and female mice. We then overexpressed Fto in a murine hypothalamic cell line and, using qPCR, detected a 50% increase in the level of oxytocin mRNA. Expression levels of several other feeding-related genes, including neuropeptide Y (NPY) and Agouti-related protein (AgRP), were unaffected by the FTO transfection. Addition of 10 and 100 nmol oxytocin to the cell culture medium did not affect Fto expression in hypothalamic cells. We conclude that Fto, a proposed transcription co-factor, influences expression of the gene encoding a satiety mediator, oxytocin.


Asunto(s)
Encéfalo/metabolismo , Regulación de la Expresión Génica , Oxo-Ácido-Liasas/metabolismo , Oxitocina/genética , Oxitocina/metabolismo , Respuesta de Saciedad , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Oxigenasas de Función Mixta , Neuronas/metabolismo
19.
BMC Neurosci ; 12: 117, 2011 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-22087873

RESUMEN

BACKGROUND: The Fat mass and obesity gene (FTO) has been identified through genome wide association studies as an important genetic factor contributing to a higher body mass index (BMI). However, the molecular context in which this effect is mediated has yet to be determined. We investigated the potential molecular network for FTO by analyzing co-expression and protein-protein interaction databases, Coxpresdb and IntAct, as well as the functional coupling predicting multi-source database, FunCoup. Hypothalamic expression of FTO-linked genes defined with this bioinformatics approach was subsequently studied using quantitative real time-PCR in mouse feeding models known to affect FTO expression. RESULTS: We identified several candidate genes for functional coupling to FTO through database studies and selected nine for further study in animal models. We observed hypothalamic expression of Profilin 2 (Pfn2), cAMP-dependent protein kinase catalytic subunit beta (Prkacb), Brain derived neurotrophic factor (Bdnf), neurotrophic tyrosine kinase, receptor, type 2 (Ntrk2), Signal transducer and activator of transcription 3 (Stat3), and Btbd12 to be co-regulated in concert with Fto. Pfn2 and Prkacb have previously not been linked to feeding regulation. CONCLUSIONS: Gene expression studies validate several candidates generated through database studies of possible FTO-interactors. We speculate about a wider functional role for FTO in the context of current and recent findings, such as in extracellular ligand-induced neuronal plasticity via NTRK2/BDNF, possibly via interaction with the transcription factor CCAAT/enhancer binding protein ß (C/EBPß).


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Hipotálamo/metabolismo , Glicoproteínas de Membrana/genética , Oxigenasas de Función Mixta/genética , Obesidad/genética , Oxo-Ácido-Liasas/genética , Proteínas Tirosina Quinasas/genética , Transducción de Señal/genética , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato , Animales , Biología Computacional/métodos , Estudio de Asociación del Genoma Completo/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo
20.
Curr Nutr Rep ; 10(4): 391-398, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34417997

RESUMEN

PURPOSE OF REVIEW: In research on autism spectrum disorder (ASD), cognitive, speech- and anxiety-related impairments have been the focus of the majority of studies. One consistently reported ASD symptom that has rarely attracted attention is disordered appetite. The goal of this paper is to assess whether ASD-related dysregulation of food intake impacts consumption of palatable foods, including sugar. RECENT FINDINGS: Aberrant neural processing at the reward system level is at least partially responsible for excessive intake of palatable tastants, including sugar. Impaired oxytocin (OT) signaling likely contributes to the magnitude of this overconsumption. Since intake for reward is generally elevated in individuals with ASD, one strategy to curb sugar overconsumption might utilize presentation of alternative palatable food choices that are more nutritionally adequate than sucrose. Furthermore, OT, which is clinically tested to alleviate other ASD symptoms, might be an effective tool to curb overconsumption of sugar, as well as - likely - of other excessively ingested palatable foods, especially those that have sweet taste.


Asunto(s)
Trastorno del Espectro Autista , Apetito , Ingestión de Alimentos , Humanos , Oxitocina
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA