Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
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
2.
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
3.
PLoS Genet ; 12(6): e1006104, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27280443

RESUMEN

Several reports suggest obesity and bipolar disorder (BD) share some physiological and behavioural similarities. For instance, obese individuals are more impulsive and have heightened reward responsiveness, phenotypes associated with BD, while bipolar patients become obese at a higher rate and earlier age than people without BD; however, the molecular mechanisms of such an association remain obscure. Here we demonstrate, using whole transcriptome analysis, that Drosophila Ets96B, homologue of obesity-linked gene ETV5, regulates cellular systems associated with obesity and BD. Consistent with a role in obesity and BD, loss of nervous system Ets96B during development increases triacylglyceride concentration, while inducing a heightened startle-response, as well as increasing hyperactivity and reducing sleep. Of notable interest, mouse Etv5 and Drosophila Ets96B are expressed in dopaminergic-rich regions, and loss of Ets96B specifically in dopaminergic neurons recapitulates the metabolic and behavioural phenotypes. Moreover, our data indicate Ets96B inhibits dopaminergic-specific neuroprotective systems. Additionally, we reveal that multiple SNPs in human ETV5 link to body mass index (BMI) and BD, providing further evidence for ETV5 as an important and novel molecular intermediate between obesity and BD. We identify a novel molecular link between obesity and bipolar disorder. The Drosophila ETV5 homologue Ets96B regulates the expression of cellular systems with links to obesity and behaviour, including the expression of a conserved endoplasmic reticulum molecular chaperone complex known to be neuroprotective. Finally, a connection between the obesity-linked gene ETV5 and bipolar disorder emphasizes a functional relationship between obesity and BD at the molecular level.


Asunto(s)
Trastorno Bipolar/genética , Proteínas de Unión al ADN/genética , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Obesidad/genética , Factores de Transcripción/fisiología , Animales , Índice de Masa Corporal , Cromatina/metabolismo , Cruzamientos Genéticos , ADN Complementario/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Proteínas de Drosophila/genética , Regulación de la Expresión Génica , Biblioteca de Genes , Silenciador del Gen , Humanos , Masculino , Oxidación-Reducción , Fosforilación Oxidativa , Fenotipo , Filogenia , Polimorfismo de Nucleótido Simple , Interferencia de ARN , Factores de Transcripción/genética
4.
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
5.
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
6.
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
7.
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.

8.
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
9.
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
10.
J Clin Med ; 11(1)2021 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-35011797

RESUMEN

A recent case report has shown that an adjunctive oxytocin + naltrexone (OT + NTX) treatment promoted more robust hypophagia and body weight reduction than OT alone in an adolescent male with hypothalamic obesity after craniopharyngioma resection. Thus far, there has been no basic research in adolescent laboratory animals that would examine whether the benefit of OT + NTX on appetite extends onto adolescent individuals without surgically induced overeating. Thus, here we examined whether low doses of combined OT + NTX acutely affect post-deprivation intake of energy-dense, standard chow; intake of energy-dense and palatable high-fat high-sugar (HFHS) diet; or calorie-dilute, palaTable 10% sucrose solution without deprivation in adolescent male rats. We assessed whether OT + NTX decreases water intake after water deprivation or produces a conditioned taste aversion (CTA). Finally, by using c-Fos immunoreactivity, we determined changes in activity of feeding-related brain areas after OT + NTX. We found that individual subthreshold doses of OT and NTX decreased feeding induced by energy and by palatability. Significant c-Fos changes were noted in the arcuate and dorsomedial hypothalamic nuclei. The hypophagic doses of OT + NTX did not suppress water intake in thirsty rats and did not cause a CTA, which suggests that feeding reduction is not a secondary effect of gastrointestinal discomfort or changes in thirst processing. We conclude that OT + NTX is an effective drug combination to reduce appetite in adolescent male rats.

11.
Physiol Behav ; 238: 113464, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34022256

RESUMEN

In a recent case report involving a male with hypothalamic obesity, concurrent administration of oxytocin (OT) and an opioid receptor antagonist, naltrexone (NTX), synergistically affected energy balance. Here, by using laboratory rats, we examined whether the reported synergy between OT and NTX in the context of food intake extends beyond that one unique case. We found that intravenous OT+NTX combination, at doses subthreshold for each of the drugs individually, decreased episodic consumption of a 10% sucrose solution in non-deprived animals. Daily administration of OT and NTX just before a scheduled, 2-hour, high-fat high-sugar (HFHS) meal over 24 days, decreased cumulative HFHS diet intake, but without a change in body weight due to compensatory standard chow intake during the remainder of the day. The NTX-OT treatment affected expression of several feeding-related genes in the hypothalamus, brain stem and nucleus accumbens, brain regions essential for the regulation of energy- and reward-driven consumption. We conclude that OT and NTX act synergistically to decrease food consumption in rats and that this transient effect is accompanied by changes in brain processes relevant to feeding.


Asunto(s)
Naltrexona , Oxitocina , Animales , Peso Corporal , Encéfalo , Ingestión de Alimentos , Expresión Génica , Humanos , Masculino , Naltrexona/farmacología , Ratas
12.
Compr Physiol ; 11(2): 1425-1447, 2021 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-33577129

RESUMEN

Ingestion of food activates a cascade of endocrine responses (thereby reflecting a contemporaneous feeding status) that include the release of hormones from the gastrointestinal (GI) tract, such as cholecystokinin (CCK), glucagonlike peptide YY (PYY), peptide PP, and oleoylethanolamide, as well as suppression of ghrelin secretion. The pancreas and adipose tissue, on the other hand, release hormones that serve as a measure of the current metabolic state or the long-term energy stores, that is, insulin, leptin, and adiponectin. It is well known and intuitively understandable that these hormones target either directly (by crossing the blood-brain barrier) or indirectly (e.g., via vagal input) the "homeostatic" brainstem-hypothalamic pathways involved in the regulation of appetite. The current article focuses on yet another target of the metabolic and GI hormones that is critical in inducing changes in food intake, namely, the reward system. We discuss the physiological basis of this functional interaction, its importance in the control of appetite, and the impact that disruption of this crosstalk has on energy intake in select physiological and pathophysiological states. We conclude that metabolic and GI hormones have a capacity to strengthen or weaken a response of the reward system to a given food, and thus, they are fundamental in ensuring that feeding reward is plastic and dependent on the energy status of the organism. © 2021 American Physiological Society. Compr Physiol 11:1425-1447, 2021.


Asunto(s)
Hormonas Gastrointestinales , Péptido YY , Apetito , Colecistoquinina , Humanos , Recompensa
13.
Foods ; 10(3)2021 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-33808819

RESUMEN

Adjustment of protein content in milk formulations modifies protein and energy levels, ensures amino acid intake and affects satiety. The shift from the natural whey:casein ratio of ~20:80 in animal milk is oftentimes done to reflect the 60:40 ratio of human milk. Studies show that 20:80 versus 60:40 whey:casein milks differently affect glucose metabolism and hormone release; these data parallel animal model findings. It is unknown whether the adjustment from the 20:80 to 60:40 ratio affects appetite and brain processes related to food intake. In this set of studies, we focused on the impact of the 20:80 vs. 60:40 whey:casein content in milk on food intake and feeding-related brain processes in the adult organism. By utilising laboratory mice, we found that the 20:80 whey:casein milk formulation was consumed less avidly and was less preferred than the 60:40 formulation in short-term choice and no-choice feeding paradigms. The relative PCR analyses in the hypothalamus and brain stem revealed that the 20:80 whey:casein milk intake upregulated genes involved in early termination of feeding and in an interplay between reward and satiety, such as melanocortin 3 receptor (MC3R), oxytocin (OXT), proopiomelanocortin (POMC) and glucagon-like peptide-1 receptor (GLP1R). The 20:80 versus 60:40 whey:casein formulation intake differently affected brain neuronal activation (assessed through c-Fos, an immediate-early gene product) in the nucleus of the solitary tract, area postrema, ventromedial hypothalamic nucleus and supraoptic nucleus. We conclude that the shift from the 20:80 to 60:40 whey:casein ratio in milk affects short-term feeding and relevant brain processes.

14.
Am J Physiol Regul Integr Comp Physiol ; 299(2): R655-63, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20427724

RESUMEN

Nociceptin/orphanin FQ (N/OFQ), the nociceptin opioid peptide (NOP) receptor ligand, increases feeding when injected centrally. Initial data suggest that N/OFQ blocks the development of a conditioned taste aversion (CTA). The current project further characterized the involvement of N/OFQ in the regulation of hunger vs. aversive responses in rats by employing behavioral, immunohistochemical, and real-time PCR methodology. We determined that the same low dose of the NOP antagonist [Nphe(1)]N/OFQ(1-13)NH(2) delivered via the lateral ventricle diminishes both N/OFQ- and deprivation-induced feeding. This anorexigenic effect did not stem from aversive consequences, as the antagonist did not cause the development of a CTA. When [Nphe(1)]N/OFQ(1-13)NH(2) was administered with LiCl, it moderately delayed extinction of the LiCl-induced CTA. Injection of LiCl + antagonist compared with LiCl alone generated an increase in c-Fos immunoreactivity in the central nucleus of the amygdala. The antagonist alone elevated Fos immunoreactivity in the paraventricular nucleus of the hypothalamus, nucleus of the solitary tract, and central nucleus of the amygdala. Hypothalamic NOP mRNA levels were decreased during energy intake restriction induced by aversion, as well as in non-CTA rats food-restricted to match CTA-reduced consumption. Brain stem NOP was upregulated only in aversion. Prepro-N/OFQ mRNA showed a trend toward upregulation in restricted rats (P = 0.068). We conclude that the N/OFQ system promotes feeding by affecting the need to replenish lacking calories and by reducing aversive responsiveness. It may belong to mechanisms that shift a balance between the drive to ingest energy and avoidance of potentially tainted food.


Asunto(s)
Conducta Animal , Encéfalo/metabolismo , Condicionamiento Psicológico , Ingestión de Alimentos , Ingestión de Energía , Hambre , Péptidos Opioides/metabolismo , Transducción de Señal , Amígdala del Cerebelo/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Tronco Encefálico/metabolismo , Condicionamiento Psicológico/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Energía/efectos de los fármacos , Extinción Psicológica , Regulación de la Expresión Génica , Hambre/efectos de los fármacos , Hipotálamo/metabolismo , Inmunohistoquímica , Inyecciones Intraventriculares , Cloruro de Litio/administración & dosificación , Masculino , Antagonistas de Narcóticos , Péptidos Opioides/genética , Fragmentos de Péptidos/administración & dosificación , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Receptor de Nociceptina , Nociceptina
15.
Behav Brain Res ; 380: 112369, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-31743731

RESUMEN

It is well accepted that opioids promote feeding for reward. Some studies suggest a potential involvement in hunger-driven intake, but they suffer from the scarcity of methodologies differentiating between factors that intersect eating for pleasure versus energy. Here, we used a unique food deprivation discrimination paradigm to test a hypothesis that, since opioids appear to control feeding reward, injection of opioid agonists would not produce effects akin to 22 h of food deprivation. We trained rats to discriminate between 22 h and 2 h food deprivation in a two-lever, operant discrimination procedure. We tested whether opioid agonists at orexigenic doses produce discriminative stimulus effects similar to 22 h deprivation. We injected DAMGO, DSLET, or orphanin FQ in the paraventricular hypothalamic nucleus (PVN), a site regulating hunger/satiety, and butorphanol subcutaneously (to produce maximum consumption). We assessed the ability of the opioid antagonist, naltrexone, to reduce the discriminative stimulus effects of 22 h deprivation and of the 22 h deprivation-like discriminative stimulus effects of PVN-injected hunger mediator, neuropeptide Y (NPY). In contrast to PVN NPY, centrally or peripherally injected opioid agonists failed to induce discriminative stimuli similar to those of 22 h deprivation. In line with that, naltrexone did not reduce the hunger discriminative stimuli induced by either 22 h deprivation or NPY administration in 2 h food-restricted subjects, even though doses used therein were sufficient to decrease deprivation-induced feeding in a non-operant setting in animals familiar with consequences of 2 h and 22 h deprivation. We conclude that opioids promote feeding for reward rather than in order to replenish lacking energy.


Asunto(s)
Analgésicos Opioides/farmacología , Conducta Animal/efectos de los fármacos , Aprendizaje Discriminativo/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Privación de Alimentos , Hambre/efectos de los fármacos , Antagonistas de Narcóticos/farmacología , Neuropéptido Y/efectos de los fármacos , Saciedad/efectos de los fármacos , Percepción del Tiempo/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Animales , Condicionamiento Operante/efectos de los fármacos , Masculino , Naltrexona/farmacología , Antagonistas de Narcóticos/administración & dosificación , Ratas , Ratas Sprague-Dawley , Recompensa
16.
Curr Top Behav Neurosci ; 43: 239-269, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-29886517

RESUMEN

Hypothalamic integration of gastrointestinal and adipose tissue-derived hormones serves as a key element of neuroendocrine control of food intake. Leptin, adiponectin, oleoylethanolamide, cholecystokinin, and ghrelin, to name a few, are in a constant "cross talk" with the feeding-related brain circuits that encompass hypothalamic populations synthesizing anorexigens (melanocortins, CART, oxytocin) and orexigens (Agouti-related protein, neuropeptide Y, orexins). While this integrated neuroendocrine circuit successfully ensures that enough energy is acquired, it does not seem to be equally efficient in preventing excessive energy intake, especially in the obesogenic environment in which highly caloric and palatable food is constantly available. The current review presents an overview of intricate mechanisms underlying hypothalamic integration of energy balance-related peripheral endocrine input. We discuss vulnerabilities and maladaptive neuroregulatory processes, including changes in hypothalamic neuronal plasticity that propel overeating despite negative consequences.


Asunto(s)
Hipotálamo , Ingestión de Alimentos , Metabolismo Energético , Humanos , Hiperfagia , Leptina , Obesidad
17.
Neurosci Lett ; 711: 134409, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31374323

RESUMEN

Administration of the mixed opioid agonist-antagonist butorphanol tartrate (BT) has been shown to robustly increase food intake in rodent models utilizing adult and young animals. BT at orexigenic doses increases c-Fos-immunoreactivity (IR) in brain areas associated with feeding for energy as well as for reward, including the paraventricular nucleus of the hypothalamus, central nucleus of the amygdala and nucleus of the solitary tract. Interestingly, aged rats given standard chow show a diminished feeding response to BT. It is not known, however, whether this weakened orexigenic response in aged animals extends to palatable tastants and whether it is accompanied by changes in brain activation. In the current study, we injected adult (11-12 months) and aged (26-27 months) rats with BT and studied the effect on intake of chow and palatable ingestants (liquid and solid). We found that BT produced only a moderate increase in consumption of bland or palatable chow as well as sweet solutions (both caloric and non-caloric) in aged rats, and that higher BT doses are required to generate such eating in old animals compared to adults. This blunted hyperphagia after BT is accompanied by diminished c-Fos IR in the central and basolateral amygdala, regions that process emotional aspects of behaviors, including food intake. Thus, aged rats exhibit diminished responsiveness to the feeding effects of BT, independent of the type of diet; and it appears to be due, in part, to diminished neural activity in central circuits involved in emotional behavior.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/efectos de los fármacos , Butorfanol/farmacología , Conducta Alimentaria/efectos de los fármacos , Antagonistas de Narcóticos/farmacología , Animales , Encéfalo/metabolismo , Hiperfagia/inducido químicamente , Masculino , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Sprague-Dawley
18.
Curr Nutr Rep ; 8(2): 120-128, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30945139

RESUMEN

PURPOSE OF REVIEW: Eating behavior provides energy to ensure proper functioning of the organism. Reward aids in seeking foods that bring energy and pleasant taste, whose consumption is safe. As evidenced by the obesity "epidemic" which largely stems from overeating, reward becomes a detriment when palatable tastants are available in unlimited quantities. This review presents recent evidence on mechanisms underlying palatability-driven excessive consumption of sugar. RECENT FINDINGS: Appetite for sugar is propelled by changes in the morphology and activity of the reward system reminiscent of addiction. Sugar intake also shifts the hunger-satiety continuum, facilitating initiation of consumption in the absence of energy needs and maintenance of feeding despite ingestion of large food loads that endanger homeostasis. Ingestion of excessive amounts of sugar relies on triggering mechanisms that promote addictive-like behaviors, and on overriding neuroendocrine signals that protect internal milieu.


Asunto(s)
Conducta Adictiva , Conducta Alimentaria/fisiología , Recompensa , Azúcares/administración & dosificación , Animales , Apetito , Encéfalo/efectos de los fármacos , Ingestión de Alimentos , Humanos , Hiperfagia , Obesidad , Azúcares/efectos adversos , Gusto/fisiología
19.
Artículo en Inglés | MEDLINE | ID: mdl-31156549

RESUMEN

Centrally and peripherally administered oxytocin (OT) decreases food intake and activation of the endogenous OT systems, which is associated with termination of feeding. Evidence gathered thus far points to OT as a facilitator of early satiation, a peptide that reduces the need for a meal that has already begun. It is not known, however, whether OT can diminish a feeling of hunger, thereby decreasing a perceived need to seek calories. Therefore, in the current project, we first confirmed that intraperitoneal (i.p.) OT at 0.3-1 mg/kg reduces food intake in deprived and non-deprived rats. We then used those OT doses in a unique hunger discrimination protocol. First, rats were trained to discriminate between 22- and 2-h food deprivation (hungry vs. sated state) in a two-lever operant procedure. After rats acquired the discrimination, they were food-restricted for 22 h and given i.p. OT before a generalization test session. OT did not decrease 22-h deprivation-appropriate responding to match that following 2-h food deprivation, thus, it did not reduce the perceived level of hunger. In order to better understand the mechanisms behind this ineffectiveness of OT, we used c-Fos immunohistochemistry to determine whether i.p. OT activates a different subset of feeding-related brain sites under 22- vs. 2-h deprivation. We found that in sated animals, OT induces c-Fos changes in a broader network of hypothalamic and brain stem sites compared to those affected in the hungry state. Finally, by employing qPCR analysis, we asked whether food deprivation vs. sated state have an impact on OT receptor expression in the brain stem, a CNS "entry" region for peripheral OT. Fasted animals had significantly lower OT receptor mRNA levels than their ad libitum-fed counterparts. We conclude that OT does not diminish a feeling of hunger before a start of a meal. Instead OT's anorexigenic properties are manifested once consumption has already begun which is-at least to some extent-driven by changes in brain responsiveness to OT treatment in the hungry vs. fed state. OT should be viewed as a mediator of early satiation rather than as a molecule that diminishes perceived hunger.

20.
Nutrients ; 11(4)2019 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-30925727

RESUMEN

Goat's (GM) and cow's milk (CM) are dietary alternatives with select health benefits shown in human and animal studies. Surprisingly, no systematic analysis of palatability or preference for GM vs. CM has been performed to date. Here, we present a comprehensive investigation of short-term intake and palatability profiles of GM and CM in laboratory mice and rats. We studied consumption in no-choice and choice scenarios, including meal microstructure, and by using isocaloric milks and milk-enriched solid diets. Feeding results are accompanied by qPCR data of relevant genes in the energy balance-related hypothalamus and brain stem, and in the nucleus accumbens, which regulates eating for palatability. We found that GM and CM are palatable to juvenile, adult, and aged rodents. Given a choice, animals prefer GM- to CM-based diets. Analysis of meal microstructure using licking patterns points to enhanced palatability of and, possibly, greater motivation toward GM over CM. Most profound changes in gene expression after GM vs. CM were associated with the brain systems driving consumption for reward. We conclude that, while both GM and CM are palatable, GM is preferred over CM by laboratory animals, and this preference is driven by central mechanisms controlling eating for pleasure.


Asunto(s)
Encéfalo/metabolismo , Bovinos , Conducta Alimentaria , Regulación de la Expresión Génica/efectos de los fármacos , Cabras , Leche , Envejecimiento/fisiología , Animales , Encéfalo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Especificidad de la Especie , Gusto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA