Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Cell ; 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39121857

RESUMO

Negative psychological states impact immunity by altering the gut microbiome. However, the relationship between brain states and microbiome composition remains unclear. We show that Brunner's glands in the duodenum couple stress-sensitive brain circuits to bacterial homeostasis. Brunner's glands mediated the enrichment of gut Lactobacillus species in response to vagus nerve stimulation. Cell-specific ablation of the glands markedly suppressed Lactobacilli counts and heightened vulnerability to infection. In the forebrain, we mapped a vagally mediated, polysynaptic circuit connecting the central nucleus of the amygdala to Brunner's glands. Chronic stress suppressed central amygdala activity and phenocopied the effects of gland lesions. Conversely, excitation of either the central amygdala or parasympathetic vagal neurons activated Brunner's glands and reversed the effects of stress on the gut microbiome and immunity. The findings revealed a tractable brain-body mechanism linking psychological states to host defense.

2.
Cell ; 185(14): 2478-2494.e28, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35662413

RESUMO

Glucagon-like peptide-1 (GLP-1) is a signal peptide released from enteroendocrine cells of the lower intestine. GLP-1 exerts anorectic and antimotility actions that protect the body against nutrient malabsorption. However, little is known about how intestinal GLP-1 affects distant organs despite rapid enzymatic inactivation. We show that intestinal GLP-1 inhibits gastric emptying and eating via intestinofugal neurons, a subclass of myenteric neurons that project to abdominal sympathetic ganglia. Remarkably, cell-specific ablation of intestinofugal neurons eliminated intestinal GLP-1 effects, and their chemical activation functioned as a GLP-1 mimetic. GLP-1 sensing by intestinofugal neurons then engaged a sympatho-gastro-spinal-reticular-hypothalamic pathway that links abnormal stomach distension to craniofacial programs for food rejection. Within this pathway, cell-specific activation of discrete neuronal populations caused systemic GLP-1-like effects. These molecularly identified, delimited enteric circuits may be targeted to ameliorate the abdominal bloating and loss of appetite typical of gastric motility disorders.


Assuntos
Apetite , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Íleo , Neurônios , Estômago , Abdome , Animais , Comunicação Celular , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Íleo/inervação , Íleo/metabolismo , Masculino , Camundongos , Neurônios/metabolismo , Óxido Nítrico/metabolismo , Transdução de Sinais , Estômago/inervação , Estômago/metabolismo
3.
Cell ; 175(3): 665-678.e23, 2018 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-30245012

RESUMO

The gut is now recognized as a major regulator of motivational and emotional states. However, the relevant gut-brain neuronal circuitry remains unknown. We show that optical activation of gut-innervating vagal sensory neurons recapitulates the hallmark effects of stimulating brain reward neurons. Specifically, right, but not left, vagal sensory ganglion activation sustained self-stimulation behavior, conditioned both flavor and place preferences, and induced dopamine release from Substantia nigra. Cell-specific transneuronal tracing revealed asymmetric ascending pathways of vagal origin throughout the CNS. In particular, transneuronal labeling identified the glutamatergic neurons of the dorsolateral parabrachial region as the obligatory relay linking the right vagal sensory ganglion to dopamine cells in Substantia nigra. Consistently, optical activation of parabrachio-nigral projections replicated the rewarding effects of right vagus excitation. Our findings establish the vagal gut-to-brain axis as an integral component of the neuronal reward pathway. They also suggest novel vagal stimulation approaches to affective disorders.


Assuntos
Intestinos/fisiologia , Recompensa , Substância Negra/fisiologia , Nervo Vago/fisiologia , Vias Aferentes/metabolismo , Vias Aferentes/fisiologia , Animais , Dopamina/metabolismo , Neurônios Dopaminérgicos/fisiologia , Ácido Glutâmico/metabolismo , Intestinos/inervação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Optogenética
4.
Cell ; 168(1-2): 311-324.e18, 2017 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-28086095

RESUMO

Superior predatory skills led to the evolutionary triumph of jawed vertebrates. However, the mechanisms by which the vertebrate brain controls predation remain largely unknown. Here, we reveal a critical role for the central nucleus of the amygdala in predatory hunting. Both optogenetic and chemogenetic stimulation of central amygdala of mice elicited predatory-like attacks upon both insect and artificial prey. Coordinated control of cervical and mandibular musculatures, which is necessary for accurately positioning lethal bites on prey, was mediated by a central amygdala projection to the reticular formation in the brainstem. In contrast, prey pursuit was mediated by projections to the midbrain periaqueductal gray matter. Targeted lesions to these two pathways separately disrupted biting attacks upon prey versus the initiation of prey pursuit. Our findings delineate a neural network that integrates distinct behavioral modules and suggest that central amygdala neurons instruct predatory hunting across jawed vertebrates.


Assuntos
Núcleo Central da Amígdala/fisiologia , Comportamento Predatório , Animais , Ansiedade/metabolismo , Núcleo Central da Amígdala/anatomia & histologia , Eletromiografia , Interneurônios/metabolismo , Mandíbula/anatomia & histologia , Mandíbula/inervação , Mandíbula/fisiologia , Camundongos , Pescoço/anatomia & histologia , Pescoço/inervação , Pescoço/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Substância Cinzenta Periaquedutal/fisiologia
6.
Annu Rev Psychol ; 71: 139-164, 2020 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-31561741

RESUMO

The conscious perception of the hedonic sensory properties of caloric foods is commonly believed to guide our dietary choices. Current and traditional models implicate the consciously perceived hedonic qualities of food as driving overeating, whereas subliminal signals arising from the gut would curb our uncontrolled desire for calories. Here we review recent animal and human studies that support a markedly different model for food reward. These findings reveal in particular the existence of subcortical body-to-brain neural pathways linking gastrointestinal nutrient sensors to the brain's reward regions. Unexpectedly, consciously perceptible hedonic qualities appear to play a less relevant, and mostly transient, role in food reinforcement. In this model, gut-brain reward pathways bypass cranial taste and aroma sensory receptors and the cortical networks that give rise to flavor perception. They instead reinforce behaviors independently of the cognitive processes that support overt insights into the nature of our dietary decisions.


Assuntos
Encéfalo , Comportamento Alimentar , Alimentos , Trato Gastrointestinal , Recompensa , Animais , Humanos
7.
Appetite ; 139: 145-151, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31029689

RESUMO

BACKGROUND AND AIM: In most species, including humans, food preference is primarily controlled by nutrient value. However, the gut-brain pathways involved in preference learning remain elusive. The aim of the present study, performed in C57BL6/J mice, was to characterize the roles in nutrient preference of two critical elements of gut-brain pathways, i.e. the duodenum and vagal gut innervation. METHODS: Adult wild-type C57BL6/J mice from a normal-weight cohort sustained one of the following three procedures: duodenal-jejunal bypass intestinal rerouting (DJB), total subdiaphragmatic vagotomy (SDV), or sham surgery. Mice were assessed in short-term two-bottle preference tests before and after 24 h s exposures to solutions containing one of glutamate, lipids, sodium, or glucose. RESULTS: DJB and SDV interfered in preference formation in a nutrient-specific manner: whereas normal preference learning for lipids and glutamate was disrupted by both DJB and SDV, these interventions did not alter the formation of preferences for glucose. Interestingly, sodium preferences were abrogated by DJB but not by SDV. CONCLUSIONS: Different macronutrients make use of distinct gut-brain pathways to influence food preferences, thereby mirroring nutrient-specific processes of food digestion. Specifically, whereas both vagal innervation and duodenal sensing appear critical for generating responses to fats and protein, glucose preferences recruit post-duodenal, vagal-independent pathways in pair with the control of glucose homeostasis. Overall, our data suggest that the physiological processes involved in digesting and absorbing fats, amino acids, and glucose overlap with those mediating learned preferences for each of these nutrients.


Assuntos
Encéfalo/fisiologia , Duodeno/inervação , Preferências Alimentares/fisiologia , Nutrientes/fisiologia , Nervo Vago/fisiologia , Animais , Digestão/fisiologia , Duodeno/cirurgia , Derivação Gástrica , Aprendizagem/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nervo Vago/cirurgia
8.
J Neurosci ; 35(20): 7964-76, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25995480

RESUMO

In rodents, food-predictive cues elicit eating in the absence of hunger (Weingarten, 1983). This behavior is disrupted by the disconnection of amygdala pathways to the lateral hypothalamus (Petrovich et al., 2002). Whether this circuit contributes to long-term weight gain is unknown. Using fMRI in 32 healthy individuals, we demonstrate here that the amygdala response to the taste of a milkshake when sated but not hungry positively predicts weight change. This effect is independent of sex, initial BMI, and total circulating ghrelin levels, but it is only present in individuals who do not carry a copy of the A1 allele of the Taq1A polymorphism. In contrast, A1 allele carriers, who have decreased D2 receptor density (Blum et al., 1996), show a positive association between caudate response and weight change. Regardless of genotype, however, dynamic causal modeling supports unidirectional gustatory input from basolateral amygdala (BLA) to hypothalamus in sated subjects. This finding suggests that, as in rodents, external cues gain access to the homeostatic control circuits of the human hypothalamus via the amygdala. In contrast, during hunger, gustatory inputs enter the hypothalamus and drive bidirectional connectivity with the amygdala. These findings implicate the BLA-hypothalamic circuit in long-term weight change related to nonhomeostatic eating and provide compelling evidence that distinct brain mechanisms confer susceptibility to weight gain depending upon individual differences in dopamine signaling.


Assuntos
Tonsila do Cerebelo/fisiologia , Sinais (Psicologia) , Fome , Saciação , Aumento de Peso/fisiologia , Adolescente , Adulto , Alelos , Feminino , Humanos , Hipotálamo/fisiologia , Masculino , Polimorfismo Genético , Receptores de Dopamina D2/genética , Aumento de Peso/genética
9.
Neuroimage ; 128: 273-283, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26724781

RESUMO

Variations in brain responses to sensory stimuli are typically considered to lack information content and treated as "noise". Alternatively, variable response patterns may reflect the adjustment of biological parameters to external factors. We used functional magnetic resonance imaging in healthy non-dieting individuals to test whether intra-individual variation in brain response to the receipt of milkshake is associated with a range of behavioral and metabolic parameters. We found that, following a meal, high variability in nucleus accumbens (NAcc) response to milkshake is associated with higher body mass index, greater dietary disinhibition, more variable ad libitum food consumption, faster increases in plasma insulin, faster decreases in plasma glucose, and greater weight loss over 1year. Our results thus uncover a series of physiological parameters encrypted as variable responses in NAcc to food stimuli. They also suggest that variations in striatal activity regulate the activation of behavioral and metabolic responses to food availability.


Assuntos
Comportamento Alimentar/fisiologia , Núcleo Accumbens/fisiologia , Adolescente , Adulto , Índice de Massa Corporal , Feminino , Humanos , Interpretação de Imagem Assistida por Computador , Processamento de Imagem Assistida por Computador , Imageamento por Ressonância Magnética , Masculino , Adulto Jovem
10.
J Physiol ; 593(8): 2097-109, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25639597

RESUMO

Sensing of dietary triacylglycerol in the proximal small intestine results in physiological, hormonal and behavioural responses. However, the exact physiological pathways linking intestinal fat sensing to food intake and the activation of brain circuits remain to be identified. In this study we examined the role of triacylglycerol digestion for intestinal fat sensing, and compared the effects of the triacylglycerol digestion products, fatty acids and 2-monoacylglycerol, on behavioural, hormonal and dopaminergic responses in behaving mice. Using an operant task in which mice are trained to self-administer lipid emulsions directly into the stomach, we show that inhibiting triacylglycerol digestion disrupts normal behaviour of self-administration in mice, indicating that fat sensing is conditional to digestion. When administered separately, both digestion products, 2-monoacylglycerol and fatty acids, were sensed by the mice, and self-administration patterns of fatty acids were affected by the fatty acid chain length. Peripheral plasma concentrations of the gut hormones GLP-1, GIP, PYY, CCK and insulin did not offer an explanation of the differing behavioural effects produced by 2-monoacylglycerol and fatty acids. However, combined with behavioural responses, striatal dopamine effluxes induced by gut infusions of oleic acid were significantly greater than those produced by equivalent infusions of 2-oleoylglycerol. Our data demonstrate recruitment of different signalling pathways by fatty acids and 2-monoacylglycerol, and suggest that the structural properties of fat rather than total caloric value determine intestinal sensing and the assignment of reward value to lipids.


Assuntos
Comportamento Animal/efeitos dos fármacos , Condicionamento Operante/efeitos dos fármacos , Ácidos Graxos/metabolismo , Intestino Delgado/metabolismo , Monoglicerídeos/metabolismo , Animais , Comportamento de Escolha/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Ácidos Graxos/farmacologia , Intestino Delgado/efeitos dos fármacos , Lactonas/farmacologia , Lipase/antagonistas & inibidores , Masculino , Camundongos , Monoglicerídeos/farmacologia , Orlistate , Autoadministração , Triglicerídeos/metabolismo
11.
bioRxiv ; 2024 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-38853855

RESUMO

Psychological states can regulate intestinal mucosal immunity by altering the gut microbiome. However, the link between the brain and microbiome composition remains elusive. We show that Brunner's glands in the duodenal submucosa couple brain activity to intestinal bacterial homeostasis. Brunner's glands mediated the enrichment of gut probiotic species in response to stimulation of abdominal vagal fibers. Cell-specific ablation of the glands triggered transmissible dysbiosis associated with an immunodeficiency syndrome that led to mortality upon gut infection with pathogens. The syndrome could be largely prevented by oral or intra-intestinal administration of probiotics. In the forebrain, we identified a vagally-mediated, polysynaptic circuit connecting the glands of Brunner to the central nucleus of the amygdala. Intra-vital imaging revealed that excitation of central amygdala neurons activated Brunner's glands and promoted the growth of probiotic populations. Our findings unveil a vagal-glandular neuroimmune circuitry that may be targeted for the modulation of the gut microbiome. The glands of Brunner may be the critical cells that regulate the levels of Lactobacilli species in the intestine.

12.
J Physiol ; 591(22): 5727-44, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24060992

RESUMO

It is well established that animals including humans attribute greater reinforcing value to glucose-containing sugars compared to their non-caloric counterparts, generally termed 'artificial sweeteners'. However, much remains to be determined regarding the physiological signals and brain systems mediating the attribution of greater reinforcing value to sweet solutions that contain glucose. Here we show that disruption of glucose utilization in mice produces an enduring inhibitory effect on artificial sweetener intake, an effect that did not depend on sweetness perception or aversion. Indeed, such an effect was not observed in mice presented with a less palatable, yet caloric, glucose solution. Consistently, hungry mice shifted their preferences away from artificial sweeteners and in favour of glucose after experiencing glucose in a hungry state. Glucose intake was found to produce significantly greater levels of dopamine efflux compared to artificial sweetener in dorsal striatum, whereas disrupting glucose oxidation suppressed dorsal striatum dopamine efflux. Conversely, inhibiting striatal dopamine receptor signalling during glucose intake in sweet-naïve animals resulted in reduced, artificial sweetener-like intake of glucose during subsequent gluco-deprivation. Our results demonstrate that glucose oxidation controls intake levels of sweet tastants by modulating extracellular dopamine levels in dorsal striatum, and suggest that glucose utilization is one critical physiological signal involved in the control of goal-directed sweetener intake.


Assuntos
Ingestão de Alimentos/fisiologia , Glucose/metabolismo , Edulcorantes/metabolismo , Animais , Corpo Estriado/metabolismo , Corpo Estriado/fisiologia , Dopamina/metabolismo , Fome/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Receptores Dopaminérgicos/metabolismo , Paladar/fisiologia
13.
Amino Acids ; 45(6): 1353-64, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24072505

RESUMO

Dietary intake of L-amino acids impacts on several physiological functions, including the control of gastrointestinal motility, pancreatic secretion, and appetite. However, the biological mechanisms regulating behavioral predilections for certain amino acid types remain poorly understood. We tested the hypothesis that, in mice, the potency with which a given glucogenic amino acid increases glucose utilization reflects its rewarding properties. We have found that: (1) during long-, but not short-, term preference tests, L-alanine and L-serine were preferred over their D-enantiomer counterparts, while no such effect was observed for L-threonine vs. D-threonine; (2) these behavioral patterns were closely associated with the ability of L-amino acids to promote increases in respiratory exchange ratios such that those, and only those, L-amino acids able to promote increases in respiratory exchange ratios were preferred over their D-isomers; (3) these behavioral preferences were independent of gustatory influences, since taste-deficient Trpm5 knockout mice displayed ingestive responses very similar to those of their wild-type counterparts. We conclude that the ability to promote increases in respiratory exchange ratios enhances the reward value of nutritionally relevant amino acids and suggest a mechanistic link between substrate utilization and amino acid preferences.


Assuntos
Aminoácidos/química , Aminoácidos/farmacologia , Aminoácidos/administração & dosagem , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mecânica Respiratória/efeitos dos fármacos , Estereoisomerismo
14.
Cell Rep ; 42(3): 112190, 2023 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-36857179

RESUMO

Although the consumption of carbohydrates is needed for survival, their potent reinforcing properties drive obesity worldwide. In turn, sugar overconsumption reveals a major role for brain reward systems in regulating sugar intake. However, it remains elusive how different cell types within the reward circuitries control the initiation and termination of sugary meals. Here, we identified the distinct nucleus accumbens cell types that mediate the chemosensory versus postprandial properties of sweet sugars. Specifically, D1 neurons enhance sugar intake via specialized connections to taste ganglia, whereas D2 neurons mediate the termination of sugary meals via anatomical connections to circuits involved in appetite suppression. Consistently, D2, but not D1, neurons partially mediate the satiating effects of glucagon-like peptide 1 (GLP-1) agonists. Thus, these nucleus accumbens cell types function as a behavioral switch, enabling positive versus negative control over sugar intake. Our study contributes to unveiling the cellular and circuit substrates of sugar overconsumption.


Assuntos
Neurônios , Núcleo Accumbens , Camundongos , Animais , Núcleo Accumbens/metabolismo , Neurônios/metabolismo , Encéfalo/metabolismo , Açúcares/metabolismo , Receptores de Dopamina D1/metabolismo
15.
Am J Clin Nutr ; 118(1): 314-328, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37149092

RESUMO

Obesity is increasing at an alarming rate. The effectiveness of currently available strategies for the treatment of obesity (including pharmacologic, surgical, and behavioral interventions) is limited. Understanding the neurobiology of appetite and the important drivers of energy intake (EI) can lead to the development of more effective strategies for the prevention and treatment of obesity. Appetite regulation is complex and is influenced by genetic, social, and environmental factors. It is intricately regulated by a complex interplay of endocrine, gastrointestinal, and neural systems. Hormonal and neural signals generated in response to the energy state of the organism and the quality of food eaten are communicated by paracrine, endocrine, and gastrointestinal signals to the nervous system. The central nervous system integrates homeostatic and hedonic signals to regulate appetite. Although there has been an enormous amount of research over many decades regarding the regulation of EI and body weight, research is only now yielding potentially effective treatment strategies for obesity. The purpose of this article is to summarize the key findings presented in June 2022 at the 23rd annual Harvard Nutrition Obesity Symposium entitled "The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets." Findings presented at the symposium, sponsored by NIH P30 Nutrition Obesity Research Center at Harvard, enhance our current understanding of appetite biology, including innovative techniques used to assess and systematically manipulate critical hedonic processes, which will shape future research and the development of therapeutics for obesity prevention and treatment.


Assuntos
Ingestão de Alimentos , Comportamento Alimentar , Humanos , Ingestão de Alimentos/fisiologia , Comportamento Alimentar/fisiologia , Obesidade/terapia , Apetite/fisiologia , Peso Corporal
16.
J Physiol ; 590(4): 953-72, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22219333

RESUMO

Animals, including humans, can achieve precise regulation of caloric intake by adjusting consumption in response to covert changes in energy density. It remains unknown, however, whether the presence of flavour cues are required for the ability to maintain constant caloric intake. Also unknown are the brain circuits that may function as the central calorie monitors that control adaptive adjustments in energy intake. Here we show that mice trained to lick a dry spout in order to receive intra-gastric infusions of a fat emulsion maintained constant hourly caloric intake by adjusting the number of dry licks in response to changes in caloric density. Animals also increased dry licking according to hunger levels, and developed conditioned preferences for dry sippers associated with high calorie infusions. Importantly, striatal dopamine levels were closely associated with the amount of calories ingested, rather than with the number of dry licks produced. Dopamine levels in dorsal and ventral striatum also reflected caloric density in mice passively receiving intra-gastric infusions of fat emulsions. Consistent with the above, systemic administration of the dopamine receptor blocker haloperidol markedly increased the production of dry licks needed to obtain high-calorie infusions, as if the caloric density of the infusions had been diluted. Conversely, haloperidol markedly decreased the production of dry licks needed to obtain low-calorie infusions. Taken together, our results support the proposition that brain dopamine circuits function as one central sensor of calorie ingestion, since (1) extracellular striatal dopamine levels fluctuate in proportion to the caloric density of nutrients infused in the gut; and (2) inhibiting dopamine receptor signalling disrupts the animals' ability to maintain constant caloric intake across experimental sessions.


Assuntos
Gânglios da Base/fisiologia , Gorduras na Dieta , Dopamina/fisiologia , Ingestão de Energia/fisiologia , Estômago/fisiologia , Paladar/fisiologia , Animais , Antagonistas de Dopamina/farmacologia , Comportamento Alimentar/fisiologia , Haloperidol/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Dopaminérgicos/fisiologia
17.
J Nutr ; 142(6): 1134S-41S, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22573784

RESUMO

A remarkable amount of information has emerged in the past decade regarding sweet taste physiology. This article reviews these data, with a particular focus on the elucidation of the sweet taste receptor, its location and actions in taste transduction in the mouth, its nontaste functions in the gastrointestinal tract (e.g., in enteroendocrine cells), and the brain circuitry involved in the sensory processing of sweet taste. Complications in the use of rodents to model human sweet taste perception and responses are also considered. In addition, information relating to low-calorie sweeteners (LCS) is discussed in the context of these issues. Particular consideration is given to the known effects of LCS on enteroendocrine cell function.


Assuntos
Edulcorantes/farmacologia , Percepção Gustatória/fisiologia , Paladar/fisiologia , Animais , Humanos , Modelos Animais , Edulcorantes/química , Papilas Gustativas/fisiologia
18.
J Neurosci ; 30(23): 8012-23, 2010 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-20534849

RESUMO

When allowed to choose between different macronutrients, most animals display a strong attraction toward carbohydrates compared with proteins. It remains uncertain, however, whether this food selection pattern depends primarily on the sensory properties intrinsic to each nutrient or, alternatively, metabolic signals can act independently of the hedonic value of sweetness to stimulate elevated sugar intake. Here we show that Trpm5(-/-) mice, which lack the cellular mechanisms required for sweet and several forms of l-amino acid taste transduction, develop a robust preference for d-glucose compared with isocaloric l-serine independently of the perception of sweetness. Moreover, a close relationship was found between glucose oxidation and taste-independent nutrient intake levels, with animals increasing intake as a function of glucose oxidation rates. Furthermore, microdialysis measurements revealed nutrient-specific dopaminergic responses in accumbens and dorsal striatum during intragastric infusions of glucose or serine. Specifically, intragastric infusions of glucose induced significantly higher levels of dopamine release compared with isocaloric serine in both ventral and dorsal striatum. Intragastric stimulation of dopamine release seemed to depend on glucose utilization, because administration of an anti-metabolic glucose analog resulted in lower dopamine levels in striatum, an effect that was reversed by intravenous glucose infusions. Together, our findings suggest that carbohydrate-specific preferences can develop independently of taste quality or caloric load, an effect associated with the ability of a given nutrient to regulate glucose metabolism and stimulate brain dopamine centers.


Assuntos
Preferências Alimentares/fisiologia , Glucose/administração & dosagem , Transdução de Sinais/fisiologia , Edulcorantes/administração & dosagem , Percepção Gustatória/fisiologia , Paladar/fisiologia , Administração Oral , Animais , Comportamento de Escolha/efeitos dos fármacos , Comportamento de Escolha/fisiologia , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Dopamina/metabolismo , Preferências Alimentares/efeitos dos fármacos , Injeções Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microdiálise , Núcleo Accumbens/efeitos dos fármacos , Núcleo Accumbens/metabolismo , Serina/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Canais de Cátion TRPM/deficiência , Paladar/efeitos dos fármacos , Paladar/genética , Percepção Gustatória/efeitos dos fármacos , Percepção Gustatória/genética
19.
Digestion ; 83 Suppl 1: 32-6, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21389726

RESUMO

Although the umami compound monosodium glutamate (MSG) is a widely used flavor enhancer, controversy still persists regarding the effects of MSG intake on body weight. It has been claimed, in particular, that chronic MSG intake may result in excessive body weight gain and obesity. In this study we assessed the effects of chronic (16 weeks) ad libitum MSG on body weight and metabolism of C57BL6/J mice. Adult male mice were divided in four experimental groups and fed with either a low-fat (LF) or high-fat (HF) diet and with either two bottles of plain water or one bottle containing 1% MSG and another one containing water according to a factorial design. Mice were monitored weekly for body weight and food/fluid intake for 15 weeks. At the end of the experiments, the circulating levels of leptin, insulin, total protein, total cholesterol, triglyceride, blood urea nitrogen, and non-esterified fatty acids were also analyzed. Our results show that MSG intake did not influence body weight in either LF or HF groups. Interestingly, although animals overall displayed strong preferences for MSG against water, preferences were relatively higher in LF compared to HF group. Consistent with the body weight data, while significant differences in leptin, insulin, total cholesterol, and non-esterified fatty acids were found between HF and LF groups, such an effect was not influenced by MSG intake. Finally, indirect calorimetry measurements revealed similar energy expenditure levels between animals being presented water only and MSG only. In summary, our data does not support the notion that ad libitum MSG intake should trigger the development of obesity or other metabolic abnormalities.


Assuntos
Metabolismo Basal/efeitos dos fármacos , Ingestão de Líquidos/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Glutamato de Sódio/farmacologia , Aumento de Peso/efeitos dos fármacos , Análise de Variância , Animais , Glicemia/efeitos dos fármacos , Proteínas Sanguíneas/efeitos dos fármacos , Nitrogênio da Ureia Sanguínea , Calorimetria Indireta , Colesterol/sangue , Dieta , Ácidos Graxos não Esterificados/sangue , Glicogênio/metabolismo , Insulina/sangue , Leptina/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Glutamato de Sódio/administração & dosagem , Triglicerídeos/sangue
20.
STAR Protoc ; 2(2): 100474, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-33997807

RESUMO

The jugular-nodose ganglia contain the sensory peripheral neurons of the vagus nerve, linking visceral organs to the medulla oblongata. Accessing these ganglia in smaller animals without damaging the vascular and neural structures may be challenging, as ganglionic fibers imbed deeply into the carotid sheath, and vagal parasympathetic fibers cross through the interior of the ganglia. We describe a practical protocol for locating and accessing the mouse jugular-nodose ganglia in vivo, including instructions for intraganglionic injections and postperfusion dissection. For complete details on the use and execution of this protocol, please refer to Han et al. (2018).


Assuntos
Dissecação/métodos , Gânglio Nodoso , Animais , Feminino , Forâmen Jugular/inervação , Masculino , Camundongos , Gânglio Nodoso/anatomia & histologia , Gânglio Nodoso/cirurgia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA