Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Appetite ; 194: 107172, 2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-38135183

RESUMO

Avoidant/restrictive food intake disorder (ARFID) is diagnosed when food avoidance leads to clinically significant nutritional, weight/growth, or psychosocial impairment. As many as 81.5% of children and adolescents diagnosed with ARFID have a history of a medical condition associated with pain, fatigue, or malaise. ARFID is diagnosed and treatment begins after the medical condition is resolved but food avoidance remains. Effective treatment involves repeated exposure to eating food and related stimuli aimed at creating inhibitory learning to counteract learned fears and aversions. Treatment usually involves positive reinforcement of food approach behavior and escape extinction/response prevention to eliminate food avoidant behavior. To shed light on the neural mechanisms that may maintain ARFID and to identify candidate pharmacological treatments for adjuncts to behavioral interventions, this paper systematically reviews research on drug treatments that successfully reduce conditioned taste aversions (CTA) in animal models by disrupting reconsolidation or promoting extinction. The mechanism of action of these treatments, brain areas involved, and whether these CTA findings have been used to understand human eating behavior are assessed. Collectively, the results provide insight into possible neural mechanisms associated with resuming oral intake following CTA akin to the therapeutic goals of ARFID treatment and suggest that CTA animal models hold promise to facilitate the development of interventions to prevent feeding problems. The findings also reveal the need to investigate CTA reduction in juvenile and female animals and show that CTA is rarely studied to understand disordered human feeding even though CTA has been observed in humans and parallels many of the characteristics of rodent CTA.


Assuntos
Transtorno Alimentar Restritivo Evitativo , Transtornos da Alimentação e da Ingestão de Alimentos , Criança , Adolescente , Humanos , Feminino , Paladar , Comportamento Alimentar/psicologia , Terapia Comportamental , Estudos Retrospectivos , Ingestão de Alimentos
2.
Nutrients ; 15(23)2023 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-38068808

RESUMO

Dietary restriction of the essential amino acid, methionine, has been shown to induce unique metabolic protection. The peripheral benefits of methionine restriction (MR) are well established and include improvements in metabolic, energy, inflammatory, and lifespan parameters in preclinical models. These benefits all occur despite MR increasing energy intake, making MR an attractive dietary intervention for the prevention or reversal of many metabolic and chronic conditions. New and emerging evidence suggests that MR also benefits the brain and promotes cognitive health. Despite widespread interest in MR over the past few decades, many findings are limited in scope, and gaps remain in our understanding of its comprehensive effects on the brain and cognition. This review details the current literature investigating the impact of MR on cognition in various mouse models, highlights some of the key mechanisms responsible for its cognitive benefits, and identifies gaps that should be addressed in MR research moving forward. Overall findings indicate that in animal models, MR is associated with protection against obesity-, age-, and Alzheimer's disease-induced impairments in learning and memory that depend on different brain regions, including the prefrontal cortex, amygdala, and hippocampus. These benefits are likely mediated by increases in fibroblast growth factor 21, alterations in methionine metabolism pathways, reductions in neuroinflammation and central oxidative stress, and potentially alterations in the gut microbiome, mitochondrial function, and synaptic plasticity.


Assuntos
Metionina , Obesidade , Camundongos , Animais , Metionina/metabolismo , Obesidade/metabolismo , Racemetionina , Ingestão de Energia , Cognição
3.
J Neurochem ; 2023 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-37391269

RESUMO

Alzheimer's disease (AD) is the most common form of dementia. Obesity in middle age increases AD risk and severity, which is alarming given that obesity prevalence peaks at middle age and obesity rates are accelerating worldwide. Midlife, but not late-life obesity increases AD risk, suggesting that this interaction is specific to preclinical AD. AD pathology begins in middle age, with accumulation of amyloid beta (Aß), hyperphosphorylated tau, metabolic decline, and neuroinflammation occurring decades before cognitive symptoms appear. We used a transcriptomic discovery approach in young adult (6.5 months old) male and female TgF344-AD rats that overexpress mutant human amyloid precursor protein and presenilin-1 and wild-type (WT) controls to determine whether inducing obesity with a high-fat/high-sugar "Western" diet during preclinical AD increases brain metabolic dysfunction in dorsal hippocampus (dHC), a brain region vulnerable to the effects of obesity and early AD. Analyses of dHC gene expression data showed dysregulated mitochondrial and neurotransmission pathways, and up-regulated genes involved in cholesterol synthesis. Western diet amplified the number of genes that were different between AD and WT rats and added pathways involved in noradrenergic signaling, dysregulated inhibition of cholesterol synthesis, and decreased intracellular lipid transporters. Importantly, the Western diet impaired dHC-dependent spatial working memory in AD but not WT rats, confirming that the dietary intervention accelerated cognitive decline. To examine later consequences of early transcriptional dysregulation, we measured dHC monoamine levels in older (13 months old) AD and WT rats of both sexes after long-term chow or Western diet consumption. Norepinephrine (NE) abundance was significantly decreased in AD rats, NE turnover was increased, and the Western diet attenuated the AD-induced increases in turnover. Collectively, these findings indicate obesity during prodromal AD impairs memory, potentiates AD-induced metabolic decline likely leading to an overproduction of cholesterol, and interferes with compensatory increases in NE transmission.

4.
Hypertension ; 80(6): 1258-1273, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37035922

RESUMO

BACKGROUND: Heart failure (HF) is a debilitating disease affecting >64 million people worldwide. In addition to impaired cardiovascular performance and associated systemic complications, most patients with HF suffer from depression and substantial cognitive decline. Although neuroinflammation and brain hypoperfusion occur in humans and rodents with HF, the underlying neuronal substrates, mechanisms, and their relative contribution to cognitive deficits in HF remains unknown. METHODS: To address this critical gap in our knowledge, we used a well-established HF rat model that mimics clinical outcomes observed in the human population, along with a multidisciplinary approach combining behavioral, electrophysiological, neuroanatomical, molecular and systemic physiological approaches. RESULTS: Our studies support neuroinflammation, hypoperfusion/hypoxia, and neuronal deficits in the hippocampus of HF rats, which correlated with the progression and severity of the disease. An increased expression of AT1aRs (Ang II [angiotensin II] receptor type 1a) in hippocampal microglia preceded the onset of neuroinflammation. Importantly, blockade of AT1Rs with a clinically used therapeutic drug (Losartan), and delivered in a clinically relevant manner, efficiently reversed neuroinflammatory end points (but not hypoxia ones), resulting in turn in improved cognitive performance in HF rats. Finally, we show than circulating Ang II can leak and access the hippocampal parenchyma in HF rats, constituting a possible source of Ang II initiating the neuroinflammatory signaling cascade in HF. CONCLUSIONS: In this study, we identified a neuronal substrate (hippocampus), a mechanism (Ang II-driven neuroinflammation) and a potential neuroprotective therapeutic target (AT1aRs) for the treatment of cognitive deficits in HF.


Assuntos
Disfunção Cognitiva , Insuficiência Cardíaca , Ratos , Humanos , Animais , Angiotensina II/farmacologia , Doenças Neuroinflamatórias , Insuficiência Cardíaca/metabolismo , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Hipocampo
5.
Neurosci Biobehav Rev ; 132: 110-129, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34813827

RESUMO

This paper reviews evidence demonstrating a bidirectional relationship between memory and eating in humans and rodents. In humans, amnesia is associated with impaired processing of hunger and satiety cues, disrupted memory of recent meals, and overconsumption. In healthy participants, meal-related memory limits subsequent ingestive behavior and obesity is associated with impaired memory and disturbances in the hippocampus. Evidence from rodents suggests that dorsal hippocampal neural activity contributes to the ability of meal-related memory to control future intake, that endocrine and neuropeptide systems act in the ventral hippocampus to provide cues regarding energy status and regulate learned aspects of eating, and that consumption of hypercaloric diets and obesity disrupt these processes. Collectively, this evidence indicates that diet-induced obesity may be caused and/or maintained, at least in part, by a vicious cycle wherein excess intake disrupts hippocampal functioning, which further increases intake. This perspective may advance our understanding of how the brain controls eating, the neural mechanisms that contribute to eating-related disorders, and identify how to treat diet-induced obesity.


Assuntos
Ingestão de Alimentos , Comportamento Alimentar , Comportamento Alimentar/fisiologia , Hipocampo/fisiologia , Humanos , Obesidade , Saciação
6.
Behav Brain Res ; 414: 113452, 2021 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-34274373

RESUMO

Chronic heart failure (HF) is a serious disorder that afflicts more than 26 million patients worldwide. HF is comorbid with depression, anxiety and memory deficits that have serious implications for quality of life and self-care in patients who have HF. Still, there are few studies that have assessed the effects of severely reduced ejection fraction (≤40 %) on cognition in non-human animal models. Moreover, limited information is available regarding the effects of HF on genetic markers of synaptic plasticity in brain areas critical for memory and mood regulation. We induced HF in male rats and tested mood and anxiety (sucrose preference and elevated plus maze) and memory (spontaneous alternation and inhibitory avoidance) and measured the simultaneous expression of 84 synaptic plasticity-associated genes in dorsal (DH) and ventral hippocampus (VH), basolateral (BLA) and central amygdala (CeA) and prefrontal cortex (PFC). We also included the hypothalamic paraventricular nucleus (PVN), which is implicated in neurohumoral activation in HF. Our results show that rats with severely reduced ejection fraction recapitulate behavioral symptoms seen in patients with chronic HF including, increased anxiety and impaired memory in both tasks. HF also downregulated several synaptic-plasticity genes in PFC and PVN, moderate decreases in DH and CeA and minimal effects in BLA and VH. Collectively, these findings identify candidate brain areas and molecular mechanisms underlying HF-induced disturbances in mood and memory.


Assuntos
Tonsila do Cerebelo/metabolismo , Sintomas Comportamentais/etiologia , Expressão Gênica , Insuficiência Cardíaca/complicações , Hipocampo/metabolismo , Transtornos da Memória/etiologia , Plasticidade Neuronal/genética , Núcleo Hipotalâmico Paraventricular/metabolismo , Animais , Ansiedade/etiologia , Comportamento Animal/fisiologia , Modelos Animais de Doenças , Regulação para Baixo , Masculino , Ratos , Ratos Wistar
7.
Neurobiol Learn Mem ; 184: 107490, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34302951

RESUMO

Memories of emotionally arousing events tend to endure longer than other memories. This review compiles findings from several decades of research investigating the role of the amygdala in modulating memories of emotional experiences. Episodic memory is a kind of declarative memory that depends upon the hippocampus, and studies suggest that the basolateral complex of the amygdala (BLA) modulates episodic memory consolidation through interactions with the hippocampus. Although many studies in rodents and imaging studies in humans indicate that the amygdala modulates memory consolidation and plasticity processes in the hippocampus, the anatomical pathways through which the amygdala affects hippocampal regions that are important for episodic memories were unresolved until recent optogenetic advances made it possible to visualize and manipulate specific BLA efferent pathways during memory consolidation. Findings indicate that the BLA influences hippocampal-dependent memories, as well as synaptic plasticity, histone modifications, gene expression, and translation of synaptic plasticity associated proteins in the hippocampus. More recent findings from optogenetic studies suggest that the BLA modulates spatial memory via projections to the medial entorhinal cortex, and that the frequency of activity in this pathway is a critical element of this modulation.


Assuntos
Tonsila do Cerebelo/fisiologia , Hipocampo/fisiologia , Consolidação da Memória/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Córtex Entorrinal/fisiologia , Humanos , Vias Neurais/fisiologia
8.
Learn Mem ; 28(6): 187-194, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34011515

RESUMO

Research into the neural mechanisms that underlie higher-order cognitive control of eating behavior suggests that ventral hippocampal (vHC) neurons, which are critical for emotional memory, also inhibit energy intake. We showed previously that optogenetically inhibiting vHC glutamatergic neurons during the early postprandial period, when the memory of the meal would be undergoing consolidation, caused rats to eat their next meal sooner and to eat more during that next meal when the neurons were no longer inhibited. The present research determined whether manipulations known to interfere with synaptic plasticity and memory when given pretraining would increase energy intake when given prior to ingestion. Specifically, we tested the effects of blocking vHC glutamatergic N-methyl-D-aspartate receptors (NMDARs) and activity-regulated cytoskeleton-associated protein (Arc) on sucrose ingestion. The results showed that male rats consumed a larger sucrose meal on days when they were given vHC infusions of the NMDAR antagonist APV or Arc antisense oligodeoxynucleotides than on days when they were given control infusions. The rats did not accommodate for that increase by delaying the onset of their next sucrose meal (i.e., decreased satiety ratio) or by eating less during the next meal. These data suggest that vHC NMDARs and Arc limit meal size and inhibit meal initiation.


Assuntos
Hipocampo , Receptores de N-Metil-D-Aspartato , Animais , Ingestão de Energia , Comportamento Alimentar , Masculino , Ratos , Ratos Sprague-Dawley
9.
eNeuro ; 6(1)2019.
Artigo em Inglês | MEDLINE | ID: mdl-30693314

RESUMO

Memory of a recently eaten meal can serve as a powerful mechanism for controlling future eating behavior because it provides a record of intake that likely outlasts most physiological signals generated by the meal. In support, impairing the encoding of a meal in humans increases the amount ingested at the next eating episode. However, the brain regions that mediate the inhibitory effects of memory on future intake are unknown. In the present study, we tested the hypothesis that dorsal hippocampal (dHC) and ventral hippocampal (vHC) glutamatergic pyramidal neurons play a critical role in the inhibition of energy intake during the postprandial period by optogenetically inhibiting these neurons at specific times relative to a meal. Male Sprague Dawley rats were given viral vectors containing CaMKIIα-eArchT3.0-eYFP or CaMKIIα-GFP and fiber optic probes into dHC of one hemisphere and vHC of the other. Compared to intake on a day in which illumination was not given, inhibition of dHC or vHC glutamatergic neurons after the end of a chow, sucrose, or saccharin meal accelerated the onset of the next meal and increased the amount consumed during that next meal when the neurons were no longer inhibited. Inhibition given during a meal did not affect the amount consumed during that meal or the next one but did hasten meal initiation. These data show that dHC and vHC glutamatergic neuronal activity during the postprandial period is critical for limiting subsequent ingestion and suggest that these neurons inhibit future intake by consolidating the memory of the preceding meal.


Assuntos
Ingestão de Alimentos/fisiologia , Hipocampo/fisiologia , Período Pós-Prandial/fisiologia , Células Piramidais/fisiologia , Animais , Sacarose Alimentar , Comportamento Alimentar/fisiologia , Ácido Glutâmico/metabolismo , Memória/fisiologia , Inibição Neural , Optogenética , Ratos Sprague-Dawley , Sacarina , Técnicas de Cultura de Tecidos
10.
Behav Brain Res ; 359: 792-798, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30076854

RESUMO

Evidence suggests that meal-related memory influences later eating behavior. Memory can serve as a powerful mechanism for controlling eating behavior because it provides a record of recent intake that likely outlasts most physiological signals generated by ingestion. Dorsal (dHC) and ventral hippocampal (vHC) neurons are critical for memory, and we demonstrated previously that they limit energy intake during the postprandial period. If dHC or vHC neurons control intake through a process that requires memory, then ingestion should increase events necessary for synaptic plasticity in dHC and vHC during the postprandial period. To test this, we determined whether ingesting a sucrose solution induced posttranslational events critical for hippocampal synaptic plasticity: phosphorylation of AMPAR GluA1 subunits at 1) serine 831 (pSer831) and 2) serine 845 (pSer845). We also examined whether increasing the amount of previous experience with the sucrose solution, which would be expected to decrease the mnemonic demand involved in an ingestion bout, would also attenuate sucrose-induced phosphorylation. Quantitative immunoblotting of dHC and vHC membrane fractions demonstrated that sucrose ingestion increased postprandial pSer831 in dHC but not vHC. Increased previous sucrose experience prevented sucrose-induced dHC pSer831. Sucrose ingestion did not affect pSer845 in either dHC or vHC. Thus, the present findings show that ingestion activates a postranslational event necessary for synaptic plasticity in an experience-dependent manner, which is consistent with the hypothesis that dHC neurons form a memory of a meal during the postprandial period.


Assuntos
Comportamento Alimentar/efeitos dos fármacos , Hipocampo/citologia , Neurônios/efeitos dos fármacos , Receptores de AMPA/metabolismo , Sacarose/metabolismo , Edulcorantes/metabolismo , Animais , Comportamento Alimentar/fisiologia , Ácido Glutâmico/farmacologia , Masculino , Fosforilação/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Serina/metabolismo , Sacarose/administração & dosagem , Edulcorantes/administração & dosagem , Fatores de Tempo
11.
Hippocampus ; 27(3): 274-284, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28121049

RESUMO

Evidence suggests that the memory of a recently ingested meal limits subsequent intake. Given that ventral hippocampal (vHC) neurons are involved in memory and energy intake, the present experiment tested the hypothesis that vHC neurons contribute to the formation of a memory of a meal and inhibit energy intake during the postprandial period. We tested (1) whether pharmacological inactivation of vHC neurons during the period following a sucrose meal, when the memory of the meal would be undergoing consolidation, accelerates the onset of the next sucrose meal and increases intake and (2) whether sucrose intake increases vHC expression of the synaptic plasticity marker activity-regulated cytoskeletal-associated protein (Arc). Adult male Sprague-Dawley rats were trained to consume a 32% sucrose solution daily at the same time and location. On the experimental day, the rats were given intra-vHC infusions of the GABAA receptor agonist muscimol or vehicle after they finished their first sucrose meal. Compared to vehicle infusions, postmeal intra-vHC muscimol infusions decreased the latency to the next sucrose meal, increased the amount of sucrose consumed during that meal, increased the total number of sucrose meals and the total amount of sucrose ingested. In addition, rats that consumed sucrose had higher levels of Arc expression in both vHC CA1 and CA3 subfields than cage control rats. Collectively, these findings are the first to show that vHC neurons inhibit energy intake during the postprandial period and support the hypothesis that vHC neurons form a memory of a meal and inhibit subsequent intake. © 2016 Wiley Periodicals, Inc.


Assuntos
Ingestão de Energia/fisiologia , Comportamento Alimentar/fisiologia , Hipocampo/fisiologia , Memória/fisiologia , Neurônios/fisiologia , Período Pós-Prandial/fisiologia , Animais , Cateteres de Demora , Proteínas do Citoesqueleto/metabolismo , Sacarose Alimentar , Agonistas de Receptores de GABA-A/farmacologia , Hipocampo/efeitos dos fármacos , Masculino , Muscimol/farmacologia , Proteínas do Tecido Nervoso/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/efeitos dos fármacos , Ratos Sprague-Dawley
12.
Physiol Behav ; 173: 1-8, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28108332

RESUMO

We hypothesize that dorsal hippocampal (dHC) neurons, which are critical for episodic memory, form a memory of a meal and inhibit the initiation of the next meal and the amount ingested during that meal. In support, we showed previously that (1) consuming a sucrose meal induces expression of the synaptic plasticity marker activity-regulated cytoskeleton-associated protein (Arc) in dHC neurons and (2) reversible inactivation of these neurons immediately following a sucrose meal accelerates the onset of the next meal and increases the size of that meal. These data suggest that hippocampal-dependent memory inhibits intake; therefore, the following experiments were conducted to determine whether hippocampal-dependent memory impairments are associated with increased intake. We reported recently that one episode of early life inflammatory pain impairs dHC-dependent memory in adult rats. The present study determined whether neonatal inflammatory pain also increases sucrose intake and attenuates sucrose-associated Arc expression. Male and female Sprague-Dawley rats were given an intraplantar injection of the inflammatory agent carrageenan (1%) on the day of birth and sucrose intake and sucrose-associated dHC Arc expression were measured in adulthood. Neonatal inflammatory pain increased sucrose intake in adult female and male rats, decreased sucrose-associated dHC Arc expression in female rats, and tended to have a similar effect on Arc expression in male rats. Neonatal inflammatory pain significantly decreased the interval between two sucrose meals in female but not in male rats. Morphine administration at the time of insult attenuated the effects of injury on sucrose intake. Collectively, these findings indicate that one brief episode of inflammatory pain on the day of birth has a long long-lasting, sex-dependent impact on intake of a palatable food in adulthood.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Ingestão de Alimentos/fisiologia , Hipocampo/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Dor/patologia , Caracteres Sexuais , Sacarose/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Carragenina/toxicidade , Proteínas do Citoesqueleto/genética , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Hipocampo/patologia , Inflamação/induzido quimicamente , Inflamação/complicações , Masculino , Proteínas do Tecido Nervoso/genética , Dor/etiologia , Gravidez , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley
13.
Physiol Behav ; 162: 112-9, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27083124

RESUMO

There is a large gap in our understanding of how top-down cognitive processes, such as memory, influence energy intake. Similarly, there is limited knowledge regarding how the brain controls the timing of meals and meal frequency. Understanding how cognition influences ingestive behavior and how the brain controls meal frequency will provide a more complete explanation of the neural mechanisms that regulate energy intake and may also increase our knowledge of the factors that contribute to diet-induced obesity. We hypothesize that dorsal hippocampal neurons, which are critical for memory of personal experiences (i.e., episodic memory), form a memory of a meal, inhibit meal onset during the period following a meal, and limit the amount ingested at the next meal. In support, we describe evidence from human research suggesting that episodic memory of a meal inhibits intake and review data from human and non-human animals showing that impaired hippocampal function is associated with increased intake. We then describe evidence from our laboratory showing that inactivation of dorsal hippocampal neurons decreases the interval between sucrose meals and increases intake at the next meal. We also describe our evidence suggesting that sweet orosensation is sufficient to induce synaptic plasticity in dorsal hippocampal neurons and raise the possibility that impaired dorsal hippocampal function and episodic memory deficits contribute to the development and/or maintenance of diet-induced obesity. Finally, we raise some critical questions that need to be addressed in future research.


Assuntos
Cognição/fisiologia , Hipocampo/fisiologia , Memória Episódica , Tamanho da Porção , Animais , Ingestão de Alimentos/fisiologia , Ingestão de Alimentos/psicologia , Hipocampo/citologia , Humanos , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia
14.
Behav Brain Res ; 307: 46-53, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-27025446

RESUMO

Hippocampal gastrin-releasing peptide receptors (GRPR) regulate memory formation and extinction, and disturbances in GRPR signaling may contribute to cognitive impairment associated with neurodevelopmental disorders. Histone acetylation is an important epigenetic mechanism that regulates gene expression involved in memory formation, and histone deacetylase inhibitors (HDACis) rescue memory deficits in several models. The present study determined whether inhibiting histone deacetylation would prevent memory impairments produced by GRPR blockade in the hippocampus. Male Wistar rats were given an intrahippocampal infusion of saline (SAL) or the HDACi sodium butyrate (NaB) shortly before inhibitory avoidance (IA) training, followed by an infusion of either SAL or the selective GRPR antagonist RC-3095 immediately after training. In a second experiment, the infusions were administered before and after a retention test trial that served as extinction training. As expected, RC-3095 significantly impaired consolidation and extinction of IA memory. More importantly, pretraining administration of NaB, at a dose that had no effect when given alone, prevented the effects of RC-3095. In addition, the combination of NaB and RC-3095 increased hippocampal levels of the brain-derived neurotrophic factor (BDNF). These findings indicate that HDAC inhibition can protect against memory impairment caused by GRPR blockade.


Assuntos
Ácido Butírico/administração & dosagem , Extinção Psicológica/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Histona Desacetilases/metabolismo , Consolidação da Memória/efeitos dos fármacos , Transtornos da Memória/prevenção & controle , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Bombesina/análogos & derivados , Bombesina/toxicidade , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Modelos Animais de Doenças , Esquema de Medicação , Hipocampo/metabolismo , Masculino , Transtornos da Memória/induzido quimicamente , Fragmentos de Peptídeos/toxicidade , Ratos , Ratos Wistar , Receptores da Bombesina/antagonistas & inibidores , Estatísticas não Paramétricas
15.
Hippocampus ; 26(3): 405-13, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26386270

RESUMO

There is limited knowledge regarding how the brain controls the timing of meals. Similarly, there is a large gap in our understanding of how top-down cognitive processes, such as memory influence energy intake. We hypothesize that dorsal hippocampal (dHC) neurons, which are critical for episodic memory, form a memory of a meal and inhibit meal onset during the postprandial period. In support, we showed previously that reversible inactivation of these neurons during the period following a sucrose meal accelerates the onset of the next meal. If dHC neurons form a memory of a meal, then consumption should induce synaptic plasticity in dHC neurons. To test this, we determined (1) whether a sucrose meal increases the expression of the synaptic plasticity marker activity-regulated cytoskeleton-associated protein (Arc) in dHC CA1 neurons, (2) whether previous experience with sucrose influences sucrose-induced Arc expression, and (3) whether the orosensory stimulation produced by the noncaloric sweetener saccharin is sufficient to induce Arc expression. Male Sprague-Dawley rats were trained to consume a sweetened solution at a scheduled time daily. On the experimental day, they were given a solution for 7 min, euthanized, and then fluorescence in situ hybridization procedures were used to measure meal-induced Arc mRNA. Compared to caged control rats, Arc expression was significantly higher in rats that consumed sucrose or saccharin. Interestingly, rats given additional experience with sucrose had less Arc expression than rats with less sucrose experience, even though both groups consumed similar amounts on the experimental day. Thus, this study is the first to suggest that orosensory stimulation produced by consuming a sweetened solution and possibly the hedonic value of that sweet stimulation induces synaptic plasticity in dHC CA1 neurons in an experience-dependent manner. Collectively, these findings are consistent with our hypothesis that dHC neurons form a memory of a meal.


Assuntos
Complexo Relacionado com a AIDS/metabolismo , Região CA1 Hipocampal/citologia , Neurônios/efeitos dos fármacos , Edulcorantes/farmacologia , Paladar/fisiologia , Complexo Relacionado com a AIDS/genética , Vias Aferentes/efeitos dos fármacos , Vias Aferentes/fisiologia , Análise de Variância , Animais , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Masculino , Neurônios/fisiologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Sacarina/farmacologia , Sacarose/farmacologia , Paladar/efeitos dos fármacos , Ensino , Fatores de Tempo
16.
Physiol Behav ; 152(Pt A): 56-61, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26367452

RESUMO

Social defeat occurs when an animal is attacked and subjugated by an aggressive conspecific. Following social defeat, male Syrian hamsters fail to display species-typical territorial aggression and instead exhibit submissive or defensive behaviors even when in the presence of a non-aggressive intruder. We have termed this phenomenon conditioned defeat (CD). The mechanisms underlying CD are not fully understood, but data from our lab suggest that at least some of the mechanisms are similar to those that mediate classical fear conditioning. The goal of the present experiment was to test the hypothesis that noradrenergic signaling promotes the consolidation of CD, as in classical fear conditioning, by determining whether CD is disrupted by post-training blockade of noradrenergic activity. In Experiment 1, we determined whether systemic infusions of the noradrenergic receptor antagonist propranolol (0, 1.0, 10, or 20mg/kg) given immediately after a 15 min defeat by a resident aggressor would impair CD tested 48 h later. Hamsters that were given immediate post-training infusions of propranolol (1.0, but not 10 or 20mg/kg) showed significantly less submissive behavior than did those given vehicle infusions supporting the hypothesis that there is noradrenergic modulation of the consolidation of a social defeat experience. In Experiment 2, we demonstrated that propranolol (1.0mg/kg) given immediately, but not 4 or 24h, after defeat impaired CD tested 48 h after defeat indicating that the window within which the memory for social defeat is susceptible to beta-adrenergic modulation is temporary. In Experiment 3, we examined whether central blockade of noradrenergic receptors could recapitulate the effect of systemic injections by giving an intracerebroventricular infusion of propranolol immediately after defeat and examining the effect on CD 24h later. Centrally administered propranolol (20 µg/3 µl but not 2 µg/3 µl) was also effective in dose-dependently reducing consolidation of CD. Collectively, the present results indicate that noradrenergic activity promotes the consolidation of CD and suggest that CD is a valuable model to study the processes by which emotion and stress modulate memory in an ethologically relevant context. These data also suggest that the popular conception in the clinical literature that the anxiolytic effect of propranolol is primarily due to the drug's peripheral effects may need to be reconsidered.


Assuntos
Condicionamento Psicológico/efeitos dos fármacos , Dominação-Subordinação , Consolidação da Memória/efeitos dos fármacos , Mesocricetus/psicologia , Propranolol/administração & dosagem , Psicotrópicos/administração & dosagem , Antagonistas Adrenérgicos beta/administração & dosagem , Animais , Condicionamento Psicológico/fisiologia , Relação Dose-Resposta a Droga , Desamparo Aprendido , Masculino , Consolidação da Memória/fisiologia , Mesocricetus/fisiologia , Distribuição Aleatória , Fatores de Tempo
17.
J Diabetes Complications ; 29(3): 329-37, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25716573

RESUMO

AIMS: Over the past five decades, per capita caloric intake has increased significantly, and diet- and stress-related diseases are more prevalent. The stress hormone epinephrine stimulates hepatic glucose release during a stress response. The present experiment tested the hypothesis that excess caloric intake alters this ability of epinephrine to increase blood glucose. METHODS: Sprague-Dawley rats were fed a high-energy cafeteria-style diet (HED). Weight gain during the first 5 days on the diet was used to divide the rats into an HED-lean group and HED-obese group. After 9 weeks, the rats were injected with epinephrine, and blood glucose was measured. RESULTS: HED-obese rats gained body and fat mass, and developed insulin resistance (IR) and hepatic steatosis. HED-lean and control rats did not differ. Epinephrine produced larger increases in blood glucose in the HED-obese rats than in the HED-lean and control rats. Removing the high-energy components of the diet for 4 weeks reversed the potentiated effects of epinephrine on glucose and corrected the IR but not the steatosis or obesity. CONCLUSIONS: Consumption of a high-energy cafeteria diet potentiates epinephrine-induced hyperglycemia. This effect is associated with insulin resistance but not adiposity or steatosis and is reversed by 4 weeks of standard chow.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Carboidratos da Dieta/efeitos adversos , Epinefrina/efeitos adversos , Hiperglicemia/induzido quimicamente , Animais , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Progressão da Doença , Ingestão de Alimentos/fisiologia , Hiperglicemia/patologia , Resistência à Insulina , Masculino , Obesidade/sangue , Obesidade/patologia , Ratos , Ratos Sprague-Dawley
18.
Neurobiol Learn Mem ; 118: 30-41, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25451312

RESUMO

The present experiment tested the hypothesis that neonatal injury disrupts adult hippocampal functioning and that normal aging or chronic stress during adulthood, which are known to have a negative impact on hippocampal function, exacerbate these effects. Male and female Sprague-Dawley rats were given an intraplantar injection of the inflammatory agent carrageenan (1%) on the day of birth and their memory was tested in the hippocampal-dependent spatial water maze in adulthood and again in middle age. We found that neonatal injury impaired hippocampal-dependent memory in adulthood, that the effects of injury on memory were more pronounced in middle-aged male rats, and that chronic stress accelerated the onset of these memory deficits. Neonatal injury also decreased glucocorticoid receptor mRNA in the dorsal CA1 area of middle-aged rats, a brain region critical for spatial memory. Morphine administration at the time of injury completely reversed injury-induced memory deficits, but neonatal morphine treatments in the absence of injury produced significant memory impairments in adulthood. Collectively, these findings are consistent with our hypothesis that neonatal injury produces long-lasting disruption in adult hippocampal functioning.


Assuntos
Hipocampo/fisiopatologia , Inflamação/complicações , Transtornos da Memória/etiologia , Dor/fisiopatologia , Memória Espacial/fisiologia , Estresse Psicológico/complicações , Fatores Etários , Animais , Animais Recém-Nascidos , Feminino , Hipocampo/metabolismo , Masculino , Morfina/administração & dosagem , Dor/etiologia , Dor/psicologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/metabolismo , Fatores Sexuais , Memória Espacial/efeitos dos fármacos
19.
Am J Physiol Regul Integr Comp Physiol ; 306(10): R701-13, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24573183

RESUMO

A wide variety of species, including vertebrate and invertebrates, consume food in bouts (i.e., meals). Decades of research suggest that different mechanisms regulate meal initiation (when to start eating) versus meal termination (how much to eat in a meal, also known as satiety). There is a very limited understanding of the mechanisms that regulate meal onset and the duration of the postprandial intermeal interval (ppIMI). In the present review, we examine issues involved in measuring meal onset and some of the limited available evidence regarding how it is regulated. Then, we describe our recent work indicating that dorsal hippocampal neurons inhibit meal onset during the ppIMI and describe the processes that may be involved in this. We also synthesize recent evidence, including evidence from our laboratory, suggesting that overeating impairs hippocampal functioning and that impaired hippocampal functioning, in turn, contributes to the development and/or maintenance of diet-induced obesity. Finally, we identify critical questions and challenges for future research investigating neural controls of meal onset.


Assuntos
Ingestão de Alimentos/fisiologia , Ingestão de Alimentos/psicologia , Hipocampo/fisiologia , Memória/fisiologia , Metabolismo Energético/fisiologia , Humanos , Hiperfagia/fisiopatologia , Período Pós-Prandial/fisiologia , Resposta de Saciedade/fisiologia , Fatores de Tempo
20.
Behav Neurosci ; 127(5): 771-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24128364

RESUMO

Over the past five decades, per capita caloric intake has increased by approximately 28% in the United States. Excessive intake of calories from fats and sugars (high energy diets; HEDs) negatively impacts hippocampal-dependent memory. These deleterious effects of HEDs on hippocampal function involve HED-induced decreases in neuronal growth factors, neurogenesis, and synaptic plasticity. Given that HEDs also alter responses to emotional arousal, the present experiment determined whether the effects of HEDs on memory depend on the emotional arousal produced by the memory task during encoding. Rats were fed a high fat/sugar cafeteria-style diet for 4 weeks and then tested in a low or high emotional arousal version of a spatial object place recognition task. The results demonstrated that the HED prevented the memory-enhancing effects of emotional arousal. Thus, altered responses to emotional arousal likely contribute to HED-induced memory impairments, particularly in stressful memory tasks such as the spatial water maze.


Assuntos
Nível de Alerta/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Emoções/efeitos dos fármacos , Memória/efeitos dos fármacos , Animais , Nível de Alerta/fisiologia , Gorduras na Dieta/toxicidade , Emoções/fisiologia , Masculino , Memória/fisiologia , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...