Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
eNeuro ; 11(3)2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38346902

RESUMO

The control of ingestive behavior is complex and involves input from many different sources, including the gustatory system. Signals transmitted via the taste nerves trigger responses that promote or discourage ingestion. The lingual taste nerves innervate 70% of taste buds, yet their role in the control of food selection and intake remarkably remains relatively underinvestigated. Here we used our custom five-item Food Choice Monitor to compare postsurgical behavioral responses to chow and a five-choice cafeteria diet (CAF) between male rats that had sham surgery (SHAM) or histologically verified transection of the chorda tympani and glossopharyngeal nerves (2NX). Compared with SHAM rats, 2NX rats ate significantly more of the high-fat CAF foods. The altered food choices led to dramatically increased fat intake and substantially reduced carbohydrate intake by 2NX vs SHAM rats. Furthermore, whether offered chow or CAF, 2NX rats ate fewer, larger meals each day. Eating rates implied that, compared with SHAM, 2NX rats were equally motivated to consume CAF but less motivated to eat chow. Even with these differences, energy intake and weight gain trajectories remained similar between SHAM and 2NX rats. Although some rats experienced CAF before surgery, contrary to our expectations, the effects of prior CAF experience on postsurgical eating were minimal. In conclusion, although total energy intake was unaffected, our results clearly indicate that information from one or both lingual taste nerves has a critical role in food selection, regulation of macronutrient intake, and meal termination but not long-term energy balance.


Assuntos
Preferências Alimentares , Paladar , Ratos , Masculino , Animais , Preferências Alimentares/fisiologia , Paladar/fisiologia , Dieta , Ingestão de Energia , Ingestão de Alimentos , Comportamento Alimentar/fisiologia
2.
Nutrients ; 15(24)2023 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-38140351

RESUMO

Humans choose which foods they will eat from multiple options. The use of cafeteria-style diets with rodent models has increased our understanding of how a multichoice food environment affects eating and health. However, the wide variances in energy density, texture, and the content of micronutrients, fiber, and protein can be interpretatively problematic when human foodstuffs are used to create rodent cafeteria diets. We minimized these differences with a custom rodent cafeteria diet (ROD) that varied similarly to a previously used human-foods cafeteria diet (HUM) in fat and sugar content. Here, we used our custom Five-Item Food Choice Monitor to compare the intake and meal patterns of rats offered ROD and HUM in a crossover design. Compared with chow, rats consumed more calories, sugar, and fat and less protein and carbohydrate while on either of the choice diets (p < 0.05). While energy intake was similar between HUM and ROD, there were differences in the responses. Rats consumed more of the low-fat, low-sugar choice on the ROD compared with the nutritionally similar choice on the HUM leading to differences in fat and carbohydrate intake between the diets (p < 0.05). The stability of macronutrient intake while on either choice diet suggests macronutrient intake is determined by the available foods and is strongly regulated. Therefore, interpretative consideration must be given to the nature of food choices in the context of available options when interpreting cafeteria-diet intake.


Assuntos
Dieta , Gorduras na Dieta , Humanos , Ratos , Animais , Comportamento Alimentar/fisiologia , Ingestão de Alimentos , Ingestão de Energia , Carboidratos , Açúcares
3.
J Endocr Soc ; 7(7): bvad074, 2023 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-37388574

RESUMO

Context: The glucagon-like peptide-1 receptor (GLP-1R) agonist semaglutide (SEMA) produces 15% weight loss when chronically administered to humans with obesity. Methods: In 2 separate experiments, rats received daily injections of either vehicle (VEH) or SEMA starting at 7 µg/kg body weight (BW) and increasing over 10 days to the maintenance dose (70 µg/kg-BW), emulating clinical dose escalation strategies. Results: During dose escalation and maintenance, SEMA rats reduced chow intake and bodyweight. Experiment 2 meal pattern analysis revealed that meal size, not number, mediated these SEMA-induced changes in chow intake. This suggests SEMA affects neural processes controlling meal termination and not meal initiation. Two-bottle preference tests (vs water) began after 10 to 16 days of maintenance dosing. Rats received either an ascending sucrose concentration series (0.03-1.0 M) and 1 fat solution (Experiment 1) or a 4% and 24% sucrose solution in a crossover design (Experiment 2). At lower sucrose concentrations, SEMA-treated rats in both experiments drank sometimes >2× the volume consumed by VEH controls; at higher sucrose concentrations (and 10% fat), intake was similar between treatment groups. Energy intake of SEMA rats became similar to VEH rats. This was unexpected because GLP-1R agonism is thought to decrease the reward and/or increase the satiating potency of palatable foods. Despite sucrose-driven increases in both groups, a significant bodyweight difference between SEMA- and VEH-treated rats remained. Conclusion: The basis of the SEMA-induced overconsumption of sucrose at lower concentrations relative to VEH controls remains unclear, but the effects of chronic SEMA treatment on energy intake and BW appear to depend on the caloric sources available.

4.
Mol Metab ; 75: 101764, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37380023

RESUMO

OBJECTIVE: Our goal is to investigate if microbiota composition modulates reward signaling and assess the role of the vagus in mediating microbiota to brain communication. METHODS: Male germ-free Fisher rats were colonized with gastrointestinal contents from chow (low fat (LF) ConvLF) or HF (ConvHF) fed rats. RESULTS: Following colonization, ConvHF rats consumed significantly more food than ConvLF animals. ConvHF rats displayed lower feeding-induced extracellular DOPAC levels (a metabolite of dopamine) in the Nucleus Accumbens (NAc) as well as reduced motivation for HF foods compared to ConvLF rats. Dopamine receptor 2 (DDR2) expression levels in the NAc were also significantly lower in ConvHF animals. Similar deficits were observed in conventionally raised HF fed rats, showing that diet-driven alteration in reward can be initiated via microbiota. Selective gut to brain deafferentation restored DOPAC levels, DRD2 expression, and motivational drive in ConvHF rats. CONCLUSIONS: We concluded from these data that a HF-type microbiota is sufficient to alter appetitive feeding behavior and that bacteria to reward communication is mediated by the vagus nerve.


Assuntos
Eixo Encéfalo-Intestino , Comportamento Alimentar , Ratos , Masculino , Animais , Ácido 3,4-Di-Hidroxifenilacético , Comportamento Alimentar/fisiologia , Recompensa , Bactérias
5.
Peptides ; 138: 170492, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33422646

RESUMO

In 1973, Gibbs, Young, and Smith showed that exogenous cholecystokinin (CCK) administration reduces food intake in rats. This initial report has led to thousands of studies investigating the physiological role of CCK in regulating feeding behavior. CCK is released from enteroendocrine I cells present along the gastrointestinal (GI) tract. CCK binding to its receptor CCK1R leads to vagal afferent activation providing post-ingestive feedback to the hindbrain. Vagal afferent neurons' (VAN) sensitivity to CCK is modulated by energy status while CCK signaling regulates gene expression of other feeding related signals and receptors expressed by VAN. In addition to its satiation effects, CCK acts all along the GI tract to optimize digestion and nutrient absorption. Diet-induced obesity (DIO) is characterized by reduced sensitivity to CCK and every part of the CCK system is negatively affected by chronic intake of energy-dense foods. EEC have recently been shown to adapt to diet, CCK1R is affected by dietary fats consumption, and the VAN phenotypic flexibility is lost in DIO. Altered endocannabinoid tone, changes in gut microbiota composition, and chronic inflammation are currently being explored as potential mechanisms for diet driven loss in CCK signaling. This review discusses our current understanding of how CCK controls food intake in conditions of leanness and how control is lost in chronic energy excess and obesity, potentially perpetuating excessive intake.


Assuntos
Colecistocinina/genética , Ingestão de Alimentos/genética , Obesidade/genética , Receptor de Colecistocinina A/genética , Animais , Colecistocinina/biossíntese , Colecistocinina/farmacologia , Dieta Hiperlipídica/efeitos adversos , Gorduras na Dieta/efeitos adversos , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Trato Gastrointestinal/metabolismo , Humanos , Neurônios Aferentes/metabolismo , Neurônios Aferentes/patologia , Obesidade/etiologia , Obesidade/fisiopatologia , Obesidade/terapia , Ratos
6.
Physiol Behav ; 226: 113124, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32763334

RESUMO

Vagal afferent neurons (VAN) projecting to the lamina propria of the digestive tract are the primary source of gut-originating signals to the central nervous system (CNS). VAN cell bodies are found in the nodose ganglia (NG). Responsiveness of VAN to gut-originating signals is altered by feeding status with sensitivity to satiety signals such as cholecystokinin (CCK) increasing in the fed state. Chronic high-fat (HF) feeding results in inflammation at the level of the NG associated with a loss of VAN ability to switch phenotype from the fasted to the fed state. HF feeding also leads to compositional changes in the gut microbiota. HF diet consumption notably drives increased Firmicutes to Bacteroidetes phyla ratio and increased members of the Actinobacteria phylum. Firmicutes and Actinobacteria are largely gram positive (GP). In this study, we aimed to determine if byproducts from GP bacteria can induce an inflammatory response in cultured NG and to characterize the mechanism and cell types involved in the response. NG were collected from male Wistar rats and cultured for a total of 72 hours. At 48-68 hours after plating, cultures were treated with neuronal culture media in which Serinicoccus chungangensis had been grown and removed (SUP), lipoteichoic acid (LTA), or meso-diaminopimelic acid (meso-DAP). Some treatments included the glial inhibitors minocycline (MINO) and/or fluorocitrate (FC). The responses were evaluated using immunocytochemistry, qPCR, and electrochemiluminescence. We found that SUP induced an inflammatory response characterized by increased interleukin (IL)-6 staining and increased expression of genes for IL-6, interferon (IFN)γ, and tumor necrosis factor (TNF)α along with genes associated with cell-to-cell communication such as C-C motif chemokine ligand-2 (CCL2). Inclusion of inhibitors attenuated some responses but failed to completely normalize all indications of response, highlighting the role of immunocompetent cellular crosstalk in regulating the inflammatory response. LTA and meso-DAP produced responses that shared characteristics with SUP but were not identical. Our results support a role for HF associated GP bacterial byproducts' ability to contribute to vagal inflammation and to engage signaling from nonneuronal cells.


Assuntos
Microbioma Gastrointestinal , Gânglio Nodoso , Actinobacteria , Animais , Bactérias , Masculino , Neurônios , Ratos , Ratos Wistar
7.
Nutrients ; 11(11)2019 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-31717368

RESUMO

(1) High-fat (HF) diet leads to gut microbiota dysbiosis which is associated with systemic inflammation. Bacterial-driven inflammation is sufficient to alter vagally mediated satiety and induce hyperphagia. Promoting bacterial fermentation improves gastrointestinal (GI) epithelial barrier function and reduces inflammation. Resistant starch escape digestion and can be fermented by bacteria in the distal gut. Therefore, we hypothesized that potato RS supplementation in HF-fed rats would lead to compositional changes in microbiota composition associated with improved inflammatory status and vagal signaling. (2) Male Wistar rats (n = 8/group) were fed a low-fat chow (LF, 13% fat), HF (45% fat), or an isocaloric HF supplemented with 12% potato RS (HFRS) diet. (3) The HFRS-fed rats consumed significantly less energy than HF animals throughout the experiment. Systemic inflammation and glucose homeostasis were improved in the HFRS compared to HF rats. Cholecystokinin-induced satiety was abolished in HF-fed rats and restored in HFRS rats. HF feeding led to a significant decrease in positive c fiber staining in the brainstem which was averted by RS supplementation. (4) The RS supplementation prevented dysbiosis and systemic inflammation. Additionally, microbiota manipulation via dietary potato RS prevented HF-diet-induced reorganization of vagal afferent fibers, loss in CCK-induced satiety, and hyperphagia.


Assuntos
Bactérias/crescimento & desenvolvimento , Encéfalo/fisiopatologia , Suplementos Nutricionais , Disbiose , Microbioma Gastrointestinal , Inflamação/prevenção & controle , Intestinos/inervação , Intestinos/microbiologia , Obesidade/prevenção & controle , Solanum tuberosum , Amido/administração & dosagem , Nervo Vago/fisiopatologia , Ração Animal , Animais , Bactérias/metabolismo , Encéfalo/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Comportamento Alimentar , Fermentação , Hiperfagia/metabolismo , Hiperfagia/microbiologia , Hiperfagia/fisiopatologia , Hiperfagia/prevenção & controle , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/fisiopatologia , Masculino , Obesidade/metabolismo , Obesidade/microbiologia , Obesidade/fisiopatologia , Raízes de Plantas , Ratos Wistar , Resposta de Saciedade , Amido/metabolismo , Nervo Vago/metabolismo , Aumento de Peso
8.
Brain Res ; 1693(Pt B): 134-139, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29360469

RESUMO

Contemporary techniques including the use of germ-free models and next generation sequencing have deepened our understanding of the gut microbiota dynamics and its influence on host physiology. There is accumulating evidence that the gut microbiota can communicate to the CNS and is involved in the development of metabolic and behavioral disorders. Vagal afferent terminals are positioned beneath the gut epithelium where they can receive, directly or indirectly, signals produced by the gut microbiota, to affect host behavior, including feeding behavior. Supplementation with L. Rhamnosus in mice notably causes a decrease in anxiety and these effects are abolished by vagotomy. Additionally, chronic treatment with bacterial byproduct lipopolysaccharide (LPS) blunts vagally-mediated post-ingestive feedback and is associated with increased food intake. Inflammation in the nodose ganglion (NG), the location of vagal afferent neurons' cell bodies, may be a key triggering factor of microbiota-driven vagal alteration. Interestingly, several models show that vagal damage leads to an increase in immune cell (microglia) activation in the NG and remodeling of the vagal pathway. Similarly, diet-driven microbiota dysbiosis is associated with NG microglia activation and decreased vagal outputs to the CNS. Crucially, preventing dysbiosis and microglia activation in high-fat diet fed rodents normalizes vagal innervation and energy intake, highlighting the importance of microbiota/vagal communication in controlling feeding behavior. As of today, new consideration of potential roles for glial influence on vagal communication and new methods of vagal afferent ablation open opportunities to increase our understanding of how the gut microbiota influence its host's health and behavior.


Assuntos
Vias Aferentes/fisiologia , Microbioma Gastrointestinal/fisiologia , Nervo Vago/fisiologia , Animais , Encéfalo/fisiologia , Humanos
9.
Physiol Behav ; 173: 305-317, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28249783

RESUMO

Obesity is one of the major health issues in the United States. Consumption of diets rich in energy, notably from fats and sugars (high-fat/high-sugar diet: HF/HSD) is linked to the development of obesity and a popular dietary approach for weight loss is to reduce fat intake. Obesity research traditionally uses low and high fat diets and there has been limited investigation of the potential detrimental effects of a low-fat/high-sugar diet (LF/HSD) on body fat accumulation and health. Therefore, in the present study, we investigated the effects of HF/HSD and LF/HSD on microbiota composition, gut inflammation, gut-brain vagal communication and body fat accumulation. Specifically, we tested the hypothesis that LF/HSD changes the gut microbiota, induces gut inflammation and alters vagal gut-brain communication, associated with increased body fat accumulation. Sprague-Dawley rats were fed an HF/HSD, LF/HSD or control low-fat/low-sugar diet (LF/LSD) for 4weeks. Body weight, caloric intake, and body composition were monitored daily and fecal samples were collected at baseline, 1, 6 and 27days after the dietary switch. After four weeks, blood and tissues (gut, brain, liver and nodose ganglia) were sampled. Both HF/HSD and LF/HSD-fed rats displayed significant increases in body weight and body fat compared to LF/LSD-fed rats. 16S rRNA sequencing showed that both HF/HSD and LF/HSD-fed animals exhibited gut microbiota dysbiosis characterized by an overall decrease in bacterial diversity and an increase in Firmicutes/Bacteriodetes ratio. Dysbiosis was typified by a bloom in Clostridia and Bacilli and a marked decrease in Lactobacillus spp. LF/HSD-fed animals showed a specific increase in Sutterella and Bilophila, both Proteobacteria, abundances of which have been associated with liver damage. Expression of pro-inflammatory cytokines, such as IL-6, IL-1ß and TNFα, was upregulated in the cecum while levels of tight junction protein occludin were downregulated in both HF/HSD and LF/HSD fed rats. HF/HSD and LF/HSD-fed rats also exhibited an increase in cecum and serum levels of lipopolysaccharide (LPS), a pro-inflammatory bacterial product. Immunofluorescence revealed the withdrawal of vagal afferents from the gut and at their site of termination the nucleus of the solitary tract (NTS) in both the HF/HSD and LF/HSD rats. Moreover, there was significant microglia activation in the nodose ganglia, which contain the vagal afferent neuron cell bodies, of HF/HSD and LF/HSD rats. Taken together, these data indicate that, similar to HF/HSD, consumption of an LF/HSD induces dysbiosis of gut microbiota, increases gut inflammation and alters vagal gut-brain communication. These changes are associated with an increase in body fat accumulation.


Assuntos
Disbiose/induzido quimicamente , Microbiota/efeitos dos fármacos , Obesidade/metabolismo , Obesidade/fisiopatologia , Animais , Composição Corporal , Peso Corporal , Proteínas de Ligação ao Cálcio/metabolismo , Citocinas/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Ingestão de Alimentos , Ingestão de Energia/fisiologia , Glicoproteínas/metabolismo , Lectinas/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Proteínas dos Microfilamentos/metabolismo , RNA Ribossômico 16S/metabolismo , Ratos , Ratos Sprague-Dawley , Rombencéfalo/metabolismo , Estatísticas não Paramétricas , Fatores de Tempo , Versicanas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...