Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Eur J Neurosci ; 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38057909

RESUMO

Food deprivation drives eating through multiple signals and circuits. Decreased glucose availability (i.e., cytoglucopenia) drives eating and also increases the value of sucrose. Ventral tegmental area (VTA) dopamine neurons (DANs) contribute to the evaluation of taste stimuli, but their role in integrating glucoprivic signals remains unknown. We monitored VTA DAN activity via Cre-dependent expression of a calcium indicator with in vivo fibre photometry. In ad libitum fed rats, intraoral sucrose evoked a phasic increase in DAN activity. To manipulate glucose availability, we administered (intraperitoneal, lateral or fourth ventricular) the antiglycolytic agent 5-thio-D-glucose (5TG), which significantly augmented the phasic DAN activity to sucrose. 5TG failed to alter DAN activity to water or saccharin, suggesting the response was selective for caloric stimuli. 5TG enhancement of sucrose-evoked DAN activity was stronger after fourth ventricular administration, suggesting a critical node of action within the hindbrain. As 5TG also increases blood glucose, in a separate study, we used peripheral insulin, which stimulates eating, to decrease blood glucose-which was associated with increased DAN activity to intraoral sucrose. DAN activity developed to a cue predictive of intraoral sucrose. While 5TG augmented cue-evoked DAN activity, its action was most potent when delivered to the lateral ventricle. Together, the studies point to central glucose availability as a key modulator of phasic DAN activity to food and food-cues. As glucose sensing neurons are known to populate the hypothalamus and brainstem, results suggest differential modulation of cue-evoked and sucrose-evoked DAN activity.

2.
Proc Natl Acad Sci U S A ; 117(48): 30744-30754, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33199591

RESUMO

Thirst is a highly potent drive that motivates organisms to seek out and consume balance-restoring stimuli. The detection of dehydration is well understood and involves signals of peripheral origin and the sampling of internal milieu by first order homeostatic neurons within the lamina terminalis-particularly glutamatergic neurons of the subfornical organ expressing CaMKIIa (SFOCaMKIIa). However, it remains unknown whether mesolimbic dopamine pathways that are critical for motivation and reinforcement integrate information from these "early" dehydration signals. We used in vivo fiber photometry in the ventral tegmental area and measured phasic dopamine responses to a water-predictive cue. Thirst, but not hunger, potentiated the phasic dopamine response to the water cue. In euvolemic rats, the dipsogenic hormone angiotensin II, but not the orexigenic hormone ghrelin, potentiated the dopamine response similarly to that observed in water-deprived rats. Chemogenetic manipulations of SFOCaMKIIa revealed bidirectional control of phasic dopamine signaling during cued water reward. Taking advantage of within-subject designs, we found predictive relationships between changes in cue-evoked dopamine response and changes in behavioral responses-supporting a role for dopamine in motivation induced by homeostatic need. Collectively, we reveal a putative mechanism for the invigoration of goal-directed behavior: internal milieu communicates to first order, need state-selective circuits to potentiate the mesolimbic dopamine system's response to cues predictive of restorative stimuli.


Assuntos
Dopamina/metabolismo , Neurônios Dopaminérgicos/metabolismo , Transdução de Sinais , Órgão Subfornical/metabolismo , Animais , Comportamento Animal , Biomarcadores , Sinais (Psicologia) , Fenômenos Eletrofisiológicos , Potenciais Evocados , Feminino , Imuno-Histoquímica , Masculino , Motivação , Ratos , Reforço Psicológico
3.
Horm Behav ; 126: 104855, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32991888

RESUMO

Oxytocin potently reduces food intake and is a potential target system for obesity treatment. A better understanding of the behavioral and neurobiological mechanisms mediating oxytocin's anorexigenic effects may guide more effective obesity pharmacotherapy development. The present study examined the effects of central (lateral intracerebroventricular [ICV]) administration of oxytocin in rats on motivated responding for palatable food. Various conditioning procedures were employed to measure distinct appetitive behavioral domains, including food seeking in the absence of consumption (conditioned place preference expression), impulsive responding for food (differential reinforcement of low rates of responding), effort-based appetitive decision making (high-effort palatable vs. low-effort bland food), and sucrose reward value encoding following a motivational shift (incentive learning). Results reveal that ICV oxytocin potently reduces food-seeking behavior, impulsivity, and effort-based palatable food choice, yet does not influence encoding of sucrose reward value in the incentive learning task. To investigate a potential neurobiological mechanism mediating these behavioral outcomes, we utilized in vivo fiber photometry in ventral tegmental area (VTA) dopamine neurons to examine oxytocin's effect on phasic dopamine neuron responses to sucrose-predictive Pavlovian cues. Results reveal that ICV oxytocin significantly reduced food cue-evoked dopamine neuron activity. Collectively, these data reveal that central oxytocin signaling inhibits various obesity-relevant conditioned appetitive behaviors, potentially via reductions in food cue-driven phasic dopamine neural responses in the VTA.


Assuntos
Sinais (Psicologia) , Comportamento Alimentar/efeitos dos fármacos , Motivação/efeitos dos fármacos , Ocitocina/administração & dosagem , Recompensa , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Comportamento Apetitivo/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Alimentos , Infusões Intraventriculares , Aprendizagem/efeitos dos fármacos , Masculino , Ocitocina/metabolismo , Ocitocina/farmacologia , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Ratos Transgênicos , Reforço Psicológico , Transdução de Sinais/efeitos dos fármacos , Área Tegmentar Ventral/metabolismo
4.
Nutr Neurosci ; 22(4): 273-283, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28944721

RESUMO

OBJECTIVES: Added dietary sugars contribute substantially to the diet of children and adolescents in the USA, and recent evidence suggests that consuming sugar-sweetened beverages (SSBs) during early life has deleterious effects on hippocampal-dependent memory function. Here, we test whether the effects of early-life sugar consumption on hippocampal function persist into adulthood when access to sugar is restricted to the juvenile/adolescent phase of development. METHODS: Male rats were given ad libitum access to an 11% weight-by-volume sugar solution (made with high fructose corn syrup-55) throughout the adolescent phase of development (post-natal day (PN) 26-56). The control group received a second bottle of water instead, and both groups received ad libitum standard laboratory chow and water access throughout the study. At PN 56 sugar solutions were removed and at PN 175 rats were subjected to behavioral testing for hippocampal-dependent episodic contextual memory in the novel object in context (NOIC) task, for anxiety-like behavior in the Zero maze, and were given an intraperitoneal glucose tolerance test. RESULTS: Early-life exposure to SSBs conferred long-lasting impairments in hippocampal-dependent memory function later in life- yet had no effect on body weight, anxiety-like behavior, or glucose tolerance. A second experiment demonstrated that NOIC performance was impaired at PN 175 even when SSB access was limited to 2 hours daily from PN 26-56. DISCUSSION: Our data suggest that even modest SSB consumption throughout early life may have long-term negative consequences on memory function during adulthood.


Assuntos
Xarope de Milho Rico em Frutose/administração & dosagem , Memória , Animais , Ansiedade/etiologia , Ingestão de Energia , Teste de Tolerância a Glucose , Hipocampo/fisiologia , Masculino , Ratos Sprague-Dawley
5.
J Nutr ; 147(1): 20-28, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27903830

RESUMO

BACKGROUND: The gut microbiome has been implicated in various metabolic and neurocognitive disorders and is heavily influenced by dietary factors, but there is a paucity of research on the effects of added sugars on the gut microbiome. OBJECTIVE: With the use of a rodent model, our goal was to determine how added-sugar consumption during the juvenile and adolescent phase of development affects the gut microbiome. METHODS: Forty-two juvenile male Sprague-Dawley rats [postnatal day (PND) 26; 50-70 g] were given access to 1 of 3 different 11%-carbohydrate solutions designed to model a range of monosaccharide ratios commonly consumed in sugar-sweetened beverages: 1) 35% fructose:65% glucose, 2) 50% fructose:50% glucose, 3) 65% fructose:35% glucose, and 4) control (no sugar). After ad libitum access to the respective solutions for the juvenile and adolescent period (PND 26-80), fecal samples were collected for next-generation 16S ribosomal RNA sequencing and multivariate microbial composition analyses. Energy intake, weight change, and adiposity index were analyzed in relation to sugar consumption and the microbiota. RESULTS: Body weight, adiposity index, and total caloric intake did not differ as a result of sugar consumption. However, sugar consumption altered the gut microbiome independently of anthropometric measures and caloric intake. At the genus level, Prevotella [linear discriminant analysis (LDA) score = -4.62; P < 0.001] and Lachnospiraceae incertae sedis (LDA score = -3.01; P = 0.03) were reduced, whereas Bacteroides (LDA score = 4.19; P < 0.001), Alistipes (LDA score = 3.88; P < 0.001), Lactobacillus (LDA score = 3.78; P < 0.001), Clostridium sensu stricto (LDA score = 3.77; P < 0.001), Bifidobacteriaceae (LDA score = 3.59; P = 0.001), and Parasutterella (LDA score = 3.79; P = 0.004) were elevated by sugar consumption. No overall pattern could be attributable to monosaccharide ratio. CONCLUSIONS: Early-life sugar consumption affects the gut microbiome in rats independently of caloric intake, body weight, or adiposity index; these effects are robust across a range of fructose-to-glucose ratios.


Assuntos
Frutose/administração & dosagem , Glucose/administração & dosagem , Microbiota/efeitos dos fármacos , Obesidade/microbiologia , Animais , Fezes/microbiologia , Masculino , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Ratos , Ratos Sprague-Dawley
6.
Hippocampus ; 25(2): 227-39, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25242636

RESUMO

Excessive consumption of added sugars negatively impacts metabolic systems; however, effects on cognitive function are poorly understood. Also unknown is whether negative outcomes associated with consumption of different sugars are exacerbated during critical periods of development (e.g., adolescence). Here we examined the effects of sucrose and high fructose corn syrup-55 (HFCS-55) intake during adolescence or adulthood on cognitive and metabolic outcomes. Adolescent or adult male rats were given 30-day access to chow, water, and either (1) 11% sucrose solution, (2) 11% HFCS-55 solution, or (3) an extra bottle of water (control). In adolescent rats, HFCS-55 intake impaired hippocampal-dependent spatial learning and memory in a Barne's maze, with moderate learning impairment also observed for the sucrose group. The learning and memory impairment is unlikely based on nonspecific behavioral effects as adolescent HFCS-55 consumption did not impact anxiety in the zero maze or performance in a non-spatial response learning task using the same mildly aversive stimuli as the Barne's maze. Protein expression of pro-inflammatory cytokines (interleukin 6, interleukin 1ß) was increased in the dorsal hippocampus for the adolescent HFCS-55 group relative to controls with no significant effect in the sucrose group, whereas liver interleukin 1ß and plasma insulin levels were elevated for both adolescent-exposed sugar groups. In contrast, intake of HFCS-55 or sucrose in adults did not impact spatial learning, glucose tolerance, anxiety, or neuroinflammatory markers. These data show that consumption of added sugars, particularly HFCS-55, negatively impacts hippocampal function, metabolic outcomes, and neuroinflammation when consumed in excess during the adolescent period of development.


Assuntos
Sacarose Alimentar/administração & dosagem , Ingestão de Alimentos/imunologia , Ingestão de Alimentos/fisiologia , Xarope de Milho Rico em Frutose/administração & dosagem , Hipocampo/imunologia , Memória Espacial/fisiologia , Animais , Ansiedade/fisiopatologia , Peso Corporal , Ensaio de Imunoadsorção Enzimática , Teste de Tolerância a Glucose , Hipocampo/crescimento & desenvolvimento , Immunoblotting , Insulina/sangue , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Fígado/metabolismo , Masculino , Aprendizagem em Labirinto/fisiologia , Neuroimunomodulação/fisiologia , Ratos Sprague-Dawley
7.
Physiol Behav ; 215: 112771, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31821815

RESUMO

Phasic dopamine activity is evoked by reliable predictors of food reward and plays a role in cue-triggered, goal-directed behavior. While this important signal is modulated by physiological state (e.g. hunger, satiety), the mechanisms by which physiological state is integrated by dopamine neurons is only beginning to be elucidated. Activation of central receptors for glucagon-like peptide-1 (GLP-1R) via long-acting agonists (e.g., Exendin-4) suppresses food intake and food-directed motivated behavior, in part, through action in regions with dopamine cell bodies, terminals, and/or neural populations that directly target the mesolimbic dopamine system. However, the effects of GLP-1R activation on cue-evoked, phasic dopamine signaling remain unknown. Here, in vivo fiber photometry was used to capture real-time signaling dynamics selectively from dopamine neurons in the ventral tegmental area of male and female transgenic (tyrosine hydroxylase-Cre; TH:Cre+) rats trained to associate an audio cue with the brief availability of a sucrose solution. Cue presentation evoked a brief spike in dopamine activity. Administration of Exendin-4 (Ex4; 0, 0.05, 0.1 µg) to the lateral ventricle both dose-dependently suppressed sucrose-directed behaviors and the magnitude of cue-evoked dopamine activity. Moreover, the amplitude of cue evoked dopamine activity was significantly correlated with subsequent sucrose-directed behaviors. While female rats exhibited overall reduced dopamine responses to the sucrose-paired cue relative to males, there was no significant interaction with Ex4. Together, these findings support a role for central GLP-1Rs in modulating a form of dopamine signaling that influences approach behavior and provide a potential mechanism whereby GLP-1 suppresses food-directed behaviors.


Assuntos
Sinais (Psicologia) , Dopamina/fisiologia , Exenatida/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Alimentos , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Estimulação Acústica , Animais , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ingestão de Alimentos/efeitos dos fármacos , Exenatida/administração & dosagem , Feminino , Injeções Intraventriculares , Masculino , Motivação , Ratos , Ratos Long-Evans , Caracteres Sexuais , Sacarose/farmacologia , Área Tegmentar Ventral/citologia , Área Tegmentar Ventral/efeitos dos fármacos
8.
Nat Commun ; 10(1): 4923, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31664021

RESUMO

Behavioral impulsivity is common in various psychiatric and metabolic disorders. Here we identify a hypothalamus to telencephalon neural pathway for regulating impulsivity involving communication from melanin-concentrating hormone (MCH)-expressing lateral hypothalamic neurons to the ventral hippocampus subregion (vHP). Results show that both site-specific upregulation (pharmacological or chemogenetic) and chronic downregulation (RNA interference) of MCH communication to the vHP increases impulsive responding in rats, indicating that perturbing this system in either direction elevates impulsivity. Furthermore, these effects are not secondary to either impaired timing accuracy, altered activity, or increased food motivation, consistent with a specific role for vHP MCH signaling in the regulation of impulse control. Results from additional functional connectivity and neural pathway tracing analyses implicate the nucleus accumbens as a putative downstream target of vHP MCH1 receptor-expressing neurons. Collectively, these data reveal a specific neural circuit that regulates impulsivity and provide evidence of a novel function for MCH on behavior.


Assuntos
Hipocampo/metabolismo , Região Hipotalâmica Lateral/metabolismo , Hormônios Hipotalâmicos/metabolismo , Comportamento Impulsivo , Melaninas/metabolismo , Hormônios Hipofisários/metabolismo , Animais , Hormônios Hipotalâmicos/genética , Masculino , Melaninas/genética , Vias Neurais , Neurônios/metabolismo , Núcleo Accumbens/metabolismo , Hormônios Hipofisários/genética , Ratos , Ratos Sprague-Dawley , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo
9.
Front Psychiatry ; 9: 410, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30233430

RESUMO

Motivated behaviors are often initiated in response to perturbations of homeostasis. Indeed, animals and humans have fundamental drives to procure (appetitive behaviors) and eventually ingest (consummatory behaviors) substances based on deficits in body fluid (e.g., thirst) and energy balance (e.g., hunger). Consumption, in turn, reinforces motivated behavior and is therefore considered rewarding. Over the years, the constructs of homeostatic (within the purview of the hypothalamus) and reward (within the purview of mesolimbic circuitry) have been used to describe need-based vs. need-free consumption. However, many experiments have demonstrated that mesolimbic circuits and "higher-order" brain regions are also profoundly influenced by changes to physiological state, which in turn generate behaviors that are poised to maintain homeostasis. Mesolimbic pathways, particularly dopamine neurons of the ventral tegmental area (VTA) and their projections to nucleus accumbens (NAc), can be robustly modulated by a variety of energy balance signals, including post-ingestive feedback relaying nutrient content and hormonal signals reflecting hunger and satiety. Moreover, physiological states can also impact VTA-NAc responses to non-nutritive rewards, such as drugs of abuse. Coupled with recent evidence showing hypothalamic structures are modulated in anticipation of replenished need, classic boundaries between circuits that convey perturbations in homeostasis and those that drive motivated behavior are being questioned. In the current review, we examine data that have revealed the importance of mesolimbic dopamine neurons and their downstream pathways as a dynamic neurobiological mechanism that provides an interface between physiological state, perturbations to homeostasis, and reward-seeking behaviors.

10.
Neuropharmacology ; 131: 487-496, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29191751

RESUMO

Social cues are potent regulators of feeding behavior, yet the neurobiological mechanisms through which social cues influence food intake are poorly understood. Here we investigate the hypothesis that the appetite-promoting gut-derived hormone, ghrelin, signals in the hippocampus to promote learned social aspects of feeding behavior. We utilized a procedure known as 'social transmission of food preference' (STFP) in which rats ('Observers') experience a social interaction with another rat ('Demonstrators') that recently consumed flavored/scented chow. STFP learning in Observer rats is indicated by a significant preference for the Demonstrator paired flavor of chow vs. a novel unpaired flavor of chow in a subsequent consumption choice test. Our results show that relative to vehicle treatment, ghrelin targeted to the ventral CA1 subregion of the hippocampus (vHP) enhanced STFP learning in rats. Additionally, STFP was impaired following peripheral injections of l-cysteine that reduce circulating ghrelin levels, suggesting that vHP ghrelin-mediated effects on STFP require peripheral ghrelin release. Finally, the endogenous relevance of vHP ghrelin receptor (GHSR-1A) signaling in STFP is supported by our data showing that STFP learning was eliminated following targeted viral vector RNA interference-mediated knockdown of vHP GHSR-1A mRNA. Control experiments indicate that vHP ghrelin-mediated STFP effects are not secondary to altered social exploration and food intake, nor to altered food preference learning based on nonsocial olfactory cues. Overall these data reveal a novel neurobiological system that promotes conditioned, social aspects of feeding behavior.


Assuntos
Comportamento Alimentar/fisiologia , Comportamento Alimentar/psicologia , Hipocampo/metabolismo , Aprendizagem/fisiologia , Receptores de Grelina/metabolismo , Comportamento Social , Animais , Cisteína , Técnicas de Silenciamento de Genes , Grelina/metabolismo , Masculino , Percepção Olfatória/fisiologia , Interferência de RNA , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Receptores de Grelina/genética
11.
Nat Commun ; 9(1): 2181, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29872139

RESUMO

The vagus nerve is the primary means of neural communication between the gastrointestinal (GI) tract and the brain. Vagally mediated GI signals activate the hippocampus (HPC), a brain region classically linked with memory function. However, the endogenous relevance of GI-derived vagal HPC communication is unknown. Here we utilize a saporin (SAP)-based lesioning procedure to reveal that selective GI vagal sensory/afferent ablation in rats impairs HPC-dependent episodic and spatial memory, effects associated with reduced HPC neurotrophic and neurogenesis markers. To determine the neural pathways connecting the gut to the HPC, we utilize monosynaptic and multisynaptic virus-based tracing methods to identify the medial septum as a relay connecting the medial nucleus tractus solitarius (where GI vagal afferents synapse) to dorsal HPC glutamatergic neurons. We conclude that endogenous GI-derived vagal sensory signaling promotes HPC-dependent memory function via a multi-order brainstem-septal pathway, thereby identifying a previously unknown role for the gut-brain axis in memory control.


Assuntos
Trato Gastrointestinal/inervação , Hipocampo/fisiologia , Vias Neurais/fisiologia , Células Receptoras Sensoriais/fisiologia , Nervo Vago/fisiologia , Animais , Córtex Cerebral/fisiologia , Masculino , Memória/fisiologia , Ratos Sprague-Dawley , Sinapses/fisiologia , Telencéfalo/fisiologia
12.
Cell Metab ; 28(1): 55-68.e7, 2018 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-29861386

RESUMO

Classical mechanisms through which brain-derived molecules influence behavior include neuronal synaptic communication and neuroendocrine signaling. Here we provide evidence for an alternative neural communication mechanism that is relevant for food intake control involving cerebroventricular volume transmission of the neuropeptide melanin-concentrating hormone (MCH). Results reveal that the cerebral ventricles receive input from approximately one-third of MCH-producing neurons. Moreover, MCH cerebrospinal fluid (CSF) levels increase prior to nocturnal feeding and following chemogenetic activation of MCH-producing neurons. Utilizing a dual viral vector approach, additional results reveal that selective activation of putative CSF-projecting MCH neurons increases food intake. In contrast, food intake was reduced following immunosequestration of MCH endogenously present in CSF, indicating that neuropeptide transmission through the cerebral ventricles is a physiologically relevant signaling pathway for energy balance control. Collectively these results suggest that neural-CSF volume transmission signaling may be a common neurobiological mechanism for the control of fundamental behaviors.


Assuntos
Ventrículos Cerebrais/metabolismo , Ingestão de Alimentos/psicologia , Comportamento Alimentar/fisiologia , Hormônios Hipotalâmicos/líquido cefalorraquidiano , Melaninas/líquido cefalorraquidiano , Neurônios/metabolismo , Hormônios Hipofisários/líquido cefalorraquidiano , Animais , Masculino , Neuropeptídeos/metabolismo , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica
13.
Front Behav Neurosci ; 11: 9, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28194099

RESUMO

Consumption of a Western Diet (WD) that is high in saturated fat and added sugars negatively impacts cognitive function, particularly mnemonic processes that rely on the integrity of the hippocampus. Emerging evidence suggests that the gut microbiome influences cognitive function via the gut-brain axis, and that WD factors significantly alter the proportions of commensal bacteria in the gastrointestinal tract. Here we review mechanisms through which consuming a WD negatively impacts neurocognitive function, with a particular focus on recent evidence linking the gut microbiome with dietary- and metabolic-associated hippocampal impairment. We highlight evidence linking gut bacteria to altered intestinal permeability and blood brain barrier integrity, thus making the brain more vulnerable to the influx of deleterious substances from the circulation. WD consumption also increases production of endotoxin by commensal bacteria, which may promote neuroinflammation and cognitive dysfunction. Recent findings also show that diet-induced alterations in gut microbiota impair peripheral insulin sensitivity, which is associated with hippocampal neuronal derrangements and associated mnemonic deficits. In some cases treatment with specific probiotics or prebiotics can prevent or reverse some of the deleterious impact of WD consumption on neuropsychological outcomes, indicating that targeting the microbiome may be a successful strategy for combating dietary- and metabolic-associated cognitive impairment.

14.
Physiol Behav ; 162: 10-7, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27072509

RESUMO

Feeding is a highly complex behavior that is influenced by learned associations between external and internal cues. The type of excessive feeding behavior contributing to obesity onset and metabolic deficit may be based, in part, on conditioned appetitive and ingestive behaviors that occur in response to environmental and/or interoceptive cues associated with palatable food. Therefore, there is a critical need to understand the neurobiology underlying learned aspects of feeding behavior. The stomach-derived "hunger" hormone, ghrelin, stimulates appetite and food intake and may function as an important biological substrate linking mnemonic processes with feeding control. The current review highlights data supporting a role for ghrelin in mediating the cognitive and neurobiological mechanisms that underlie conditioned feeding behavior. We discuss the role of learning and memory on food intake control (with a particular focus on hippocampal-dependent memory processes) and provide an overview of conditioned cephalic endocrine responses. A neurobiological framework is provided through which conditioned cephalic ghrelin secretion signals in neurons in the hippocampus, which then engage orexigenic neural circuitry in the lateral hypothalamus to express learned feeding behavior.


Assuntos
Comportamento Alimentar/fisiologia , Grelina/metabolismo , Memória/fisiologia , Animais , Sistema Nervoso Central/fisiologia , Ingestão de Alimentos/fisiologia , Sistema Endócrino/metabolismo , Humanos , Obesidade/metabolismo , Obesidade/fisiopatologia
15.
Neuropsychopharmacology ; 40(2): 327-37, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25035078

RESUMO

Glucagon-like peptide-1 (GLP-1) is produced in the small intestines and in nucleus tractus solitarius (NTS) neurons. Activation of central GLP-1 receptors (GLP-1Rs) reduces feeding and body weight. The neural circuits mediating these effects are only partially understood. Here we investigate the inhibition of food intake and motivated responding for food in rats following GLP-1R activation in the ventral hippocampal formation (HPFv), a region only recently highlighted in food intake control. Increased HPFv GLP-1R activity following exendin-4 administration potently reduced food intake (both chow and Western diet) and body weight, whereas HPFv GLP-1R blockade increased food intake. These hypophagic effects were based on reduced meal size, and likely do not involve nausea as HPFv exendin-4 did not induce a conditioned flavor avoidance. HPFv GLP-1R activation also reduced effort-based responding for food under an operant progressive ratio reinforcement schedule, but did not affect food conditioned place preference expression. To investigate possible routes of HPFv GLP-1 signaling, immunohistochemical analysis revealed the absence of GLP-1 axon terminals in the HPFv, suggesting volume transmission as a mechanism of action. Consistent with this, the presence of active GLP-1 was detected in both the cerebrospinal fluid (CSF) and the HPFv. The source of CSF GLP-1 may be NTS GLP-1-producing neurons, as, (1) ∼30% of NTS GLP-1 neurons colocalized with the retrograde tracer fluorogold (FG) following lateral ventricle FG injection, and (2) GLP-1-immunoreactive axon terminals were observed adjacent to the ventricular ependymal layer. Collectively these findings illuminate novel neuronal and behavioral mechanisms mediating food intake reduction by GLP-1.


Assuntos
Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Hipocampo/fisiologia , Motivação/fisiologia , Ração Animal , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Axônios/efeitos dos fármacos , Axônios/fisiologia , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Exenatida , Comportamento Alimentar/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1/antagonistas & inibidores , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Masculino , Motivação/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Peptídeos/farmacologia , Ratos Sprague-Dawley , Esquema de Reforço , Rombencéfalo/citologia , Rombencéfalo/efeitos dos fármacos , Rombencéfalo/fisiologia , Comportamento Espacial/efeitos dos fármacos , Comportamento Espacial/fisiologia , Percepção Gustatória/efeitos dos fármacos , Percepção Gustatória/fisiologia , Peçonhas/farmacologia
16.
Elife ; 42015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26745307

RESUMO

Feeding behavior rarely occurs in direct response to metabolic deficit, yet the overwhelming majority of research on the biology of food intake control has focused on basic metabolic and homeostatic neurobiological substrates. Most animals, including humans, have habitual feeding patterns in which meals are consumed based on learned and/or environmental factors. Here we illuminate a novel neural system regulating higher-order aspects of feeding through which the gut-derived hormone ghrelin communicates with ventral hippocampus (vHP) neurons to stimulate meal-entrained conditioned appetite. Additional results show that the lateral hypothalamus (LHA) is a critical downstream substrate for vHP ghrelin-mediated hyperphagia and that vHP ghrelin activated neurons communicate directly with neurons in the LHA that express the neuropeptide, orexin. Furthermore, activation of downstream orexin-1 receptors is required for vHP ghrelin-mediated hyperphagia. These findings reveal novel neurobiological circuitry regulating appetite through which ghrelin signaling in hippocampal neurons engages LHA orexin signaling.


Assuntos
Apetite , Grelina/metabolismo , Hipocampo/fisiologia , Hipotálamo/fisiologia , Vias Neurais/fisiologia , Orexinas/metabolismo , Transdução de Sinais , Animais , Masculino , Ratos Sprague-Dawley
17.
Front Aging Neurosci ; 6: 88, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24847262

RESUMO

Both obesity and Alzheimer's disease (AD) are major health burdens in Western societies. While commonly viewed as having separate etiologies, this review highlights data suggesting that intake of "Western diets", diets high in saturated fatty acids (SFA) and simple carbohydrates, may pose a common environmental risk factor contributing to the development of both of these adverse pathologies. We discuss the effects of Western Diet intake on learning and memory processes that are dependent on the hippocampus, as well as the importance of this brain region in both obesity development and the onset of Alzheimer's and other dementias. A putative mechanism is discussed that mechanistically links Western diet consumption, blood brain barrier (BBB) degradation, and subsequent hippocampal damage and dementia pathology.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA