Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters










Publication year range
1.
Res Sq ; 2023 Oct 27.
Article in English | MEDLINE | ID: mdl-37961128

ABSTRACT

Chronic stress underlies the etiology of both major depressive disorder (MDD) and irritable bowel syndrome (IBS), two highly prevalent and debilitating conditions with high rates of co-morbidity. However, it is not fully understood how the brain and gut bi-directionally communicate during stress to impact intestinal homeostasis and stress-relevant behaviours. Using the chronic social defeat stress (CSDS) model, we find that stressed mice display greater intestinal permeability and circulating levels of the endotoxin lipopolysaccharide (LPS) compared to unstressed control (CON) mice. Interestingly, the microbiota in the colon also exhibit elevated LPS biosynthesis gene expression following CSDS. Additionally, CSDS triggers an increase in pro-inflammatory colonic IFNγ+ Th1 cells and a decrease in IL4+ Th2 cells compared to CON mice, and this gut inflammation contributes to stress-induced intestinal barrier permeability and social avoidance behaviour. We next investigated the role of enteric neurons and identified that noradrenergic dopamine beta-hydroxylase (DBH)+ neurons in the colon are activated by CSDS, and that their ablation protects against gut pathophysiology and disturbances in social behaviour. Retrograde tracing from the colon identified a population of corticotropin-releasing hormone-expressing (CRH+) neurons in the paraventricular nucleus of the hypothalamus (PVH) that innervate the colon and are activated by stress. Chemogenetically activating these PVH CRH+ neurons is sufficient to induce gut inflammation, barrier permeability, and social avoidance behaviour, while inhibiting these cells prevents these effects following exposure to CSDS. Thus, we define a stress-activated brain-to-gut circuit that confers colonic inflammation, leading to impaired intestinal barrier function, and consequent behavioural deficits.

2.
Am J Clin Nutr ; 118(1): 314-328, 2023 07.
Article in English | MEDLINE | ID: mdl-37149092

ABSTRACT

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.


Subject(s)
Eating , Feeding Behavior , Humans , Eating/physiology , Feeding Behavior/physiology , Obesity/therapy , Appetite/physiology , Body Weight
3.
Cell Rep ; 42(3): 112190, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36857179

ABSTRACT

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.


Subject(s)
Neurons , Nucleus Accumbens , Mice , Animals , Nucleus Accumbens/metabolism , Neurons/metabolism , Brain/metabolism , Sugars/metabolism , Receptors, Dopamine D1/metabolism
4.
Hum Brain Mapp ; 44(4): 1309-1319, 2023 03.
Article in English | MEDLINE | ID: mdl-36217737

ABSTRACT

The neuroanatomical circuitry of jaw muscles has been mostly explored in non-human animals. A recent rodent study revealed a novel circuit from the central amygdala (CeA) to the trigeminal motor nucleus (5M), which controls biting attacks. This circuit has yet to be delineated in humans. Ultra-high diffusion-weighted imaging data from the Human Connectome Project (HCP) allow in vivo delineation of circuits identified in other species-for example, the CeA-5M pathway-in humans. We hypothesized that the CeA-5M circuit could be resolved in humans at both 7 and 3 T. We performed probabilistic tractography between the CeA and 5M in 30 healthy young adults from the HCP database. As a negative control, we performed tractography between the basolateral amygdala (BLAT) and 5M, as CeA is the only amygdalar nucleus with extensive projections to the brainstem. Connectivity strength was operationalized as the number of streamlines between each region of interest. Connectivity strength between CeA-5M and BLAT-5M within each hemisphere was compared, and CeA-5M circuit had significantly stronger connectivity than the BLAT-5M circuit, bilaterally at both 7 T (all p < .001) and 3 T (all p < .001). This study is the first to delineate the CeA-5M circuit in humans.


Subject(s)
Central Amygdaloid Nucleus , Trigeminal Motor Nucleus , Animals , Humans , Central Amygdaloid Nucleus/diagnostic imaging , Neural Pathways/diagnostic imaging , Neural Pathways/physiology , Diffusion Magnetic Resonance Imaging , Brain Stem
5.
Cell ; 185(14): 2478-2494.e28, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35662413

ABSTRACT

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.


Subject(s)
Appetite , Glucagon-Like Peptide 1/metabolism , Ileum , Neurons , Stomach , Abdomen , Animals , Cell Communication , Glucagon-Like Peptide-1 Receptor/metabolism , Ileum/innervation , Ileum/metabolism , Male , Mice , Neurons/metabolism , Nitric Oxide/metabolism , Signal Transduction , Stomach/innervation , Stomach/metabolism
6.
PLoS One ; 17(2): e0263527, 2022.
Article in English | MEDLINE | ID: mdl-35143525

ABSTRACT

Chronic pain is associated with anhedonia and decreased motivation. These behavioral alterations have been linked to alterations in the limbic brain and could explain the increased risk for obesity in pain patients. The mechanism of these behavioral changes and how they set in in relation to the development of chronic pain remain however poorly understood. Here we asked how eating behavior was affected in low-back pain patients before and after they transitioned to chronic pain, compared to patients whose pain subsided. Additionally, we assessed how the hedonic perception of fat-rich food, which is altered in chronic pain patients, related to the properties of the nucleus accumbens in this patients' population. We hypothesized that the accumbens would be directly implicated in the hedonic processing of fat-rich food in pain patients because of its well-established role in hedonic feeding and fat ingestion, and its emerging role in chronic pain. Accordingly, we used behavioral assays and structural brain imaging to test sub-acute back pain patients (SBP) and healthy control subjects at baseline and at approximately one-year follow-up. We also studied a sample of chronic low-back pain patients (CLBP) at one time point only. We found that SBP patients who recovered at follow-up (SBPr) and CLBP patients showed disrupted eating behaviors. In contrast, SBP patients who persisted in having pain at follow-up (SBPp) showed intact eating behavior. From a neurological standpoint, only SBPp and CLBP patients showed a strong and direct relationship between hedonic perception of fat-rich food and nucleus accumbens volume. This suggests that accumbens alterations observed in SBPp patients in previous works might protect them from hedonic eating disruptions during the early course of the illness. We conclude that disrupted eating behavior specifically sets in after pain chronification and is accompanied by structural changes in the nucleus accumbens.


Subject(s)
Feeding Behavior , Low Back Pain/physiopathology , Nucleus Accumbens , Adult , Appetite , Chronic Pain , Dietary Fats , Female , Follow-Up Studies , Food Preferences , Humans , Low Back Pain/psychology , Magnetic Resonance Imaging , Male , Nucleus Accumbens/physiopathology , Pleasure
7.
STAR Protoc ; 2(2): 100474, 2021 06 18.
Article in English | MEDLINE | ID: mdl-33997807

ABSTRACT

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).


Subject(s)
Dissection/methods , Nodose Ganglion , Animals , Female , Jugular Foramina/innervation , Male , Mice , Nodose Ganglion/anatomy & histology , Nodose Ganglion/surgery
8.
Neuron ; 109(3): 391-393, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33539771

ABSTRACT

Area postrema in brainstem has long been known to trigger emesis by detecting blood-borne toxins and pathogens. In this issue, Zhang and colleagues provide a single-cell molecular atlas of this region, opening new possibilities for harnessing its neurons in vivo.


Subject(s)
Area Postrema , Nausea , Brain Stem , Humans , Neurons , Vomiting
10.
Nature ; 583(7816): 441-446, 2020 07.
Article in English | MEDLINE | ID: mdl-32641826

ABSTRACT

Connections between the gut and brain monitor the intestinal tissue and its microbial and dietary content1, regulating both physiological intestinal functions such as nutrient absorption and motility2,3, and brain-wired feeding behaviour2. It is therefore plausible that circuits exist to detect gut microorganisms and relay this information to areas of the central nervous system that, in turn, regulate gut physiology4. Here we characterize the influence of the microbiota on enteric-associated neurons by combining gnotobiotic mouse models with transcriptomics, circuit-tracing methods and functional manipulations. We find that the gut microbiome modulates gut-extrinsic sympathetic neurons: microbiota depletion leads to increased expression of the neuronal transcription factor cFos, and colonization of germ-free mice with bacteria that produce short-chain fatty acids suppresses cFos expression in the gut sympathetic ganglia. Chemogenetic manipulations, translational profiling and anterograde tracing identify a subset of distal intestine-projecting vagal neurons that are positioned to have an afferent role in microbiota-mediated modulation of gut sympathetic neurons. Retrograde polysynaptic neuronal tracing from the intestinal wall identifies brainstem sensory nuclei that are activated during microbial depletion, as well as efferent sympathetic premotor glutamatergic neurons that regulate gastrointestinal transit. These results reveal microbiota-dependent control of gut-extrinsic sympathetic activation through a gut-brain circuit.


Subject(s)
Gastrointestinal Microbiome/physiology , Intestines/innervation , Neurons/physiology , Sympathetic Nervous System/cytology , Sympathetic Nervous System/physiology , Animals , Dysbiosis/physiopathology , Female , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/physiology , Gastrointestinal Motility , Germ-Free Life , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Models, Animal , Neural Pathways/physiology , Proto-Oncogene Proteins c-fos/metabolism , Transcriptome
11.
J Clin Invest ; 130(9): 4985-4998, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32516139

ABSTRACT

The brain has evolved in an environment where food sources are scarce, and foraging for food is one of the major challenges for survival of the individual and species. Basic and clinical studies show that obesity or overnutrition leads to overwhelming changes in the brain in animals and humans. However, the exact mechanisms underlying the consequences of excessive energy intake are not well understood. Neurons expressing the neuropeptide hypocretin/orexin (Hcrt) in the lateral/perifonical hypothalamus (LH) are critical for homeostatic regulation, reward seeking, stress response, and cognitive functions. In this study, we examined adaptations in Hcrt cells regulating behavioral responses to salient stimuli in diet-induced obese mice. Our results demonstrated changes in primary cilia, synaptic transmission and plasticity, cellular responses to neurotransmitters necessary for reward seeking, and stress responses in Hcrt neurons from obese mice. Activities of neuronal networks in the LH and hippocampus were impaired as a result of decreased hypocretinergic function. The weakened Hcrt system decreased reward seeking while altering responses to acute stress (stress-coping strategy), which were reversed by selectively activating Hcrt cells with chemogenetics. Taken together, our data suggest that a deficiency in Hcrt signaling may be a common cause of behavioral changes (such as lowered arousal, weakened reward seeking, and altered stress response) in obese animals.


Subject(s)
Feeding Behavior , Hypothalamus , Nerve Net , Neurons , Obesity , Orexins , Animals , Hypothalamus/metabolism , Hypothalamus/pathology , Hypothalamus/physiopathology , Male , Mice , Mice, Transgenic , Nerve Net/metabolism , Nerve Net/pathology , Nerve Net/physiopathology , Neurons/metabolism , Neurons/pathology , Obesity/genetics , Obesity/metabolism , Obesity/pathology , Obesity/physiopathology , Orexins/genetics , Orexins/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism , Stress, Psychological/pathology , Stress, Psychological/physiopathology
12.
eNeuro ; 7(4)2020.
Article in English | MEDLINE | ID: mdl-32471848

ABSTRACT

The olfactory system is uniquely heterogeneous, performing multifaceted functions (beyond basic sensory processing) across diverse, widely distributed neural substrates. While knowledge of human olfaction continues to grow, it remains unclear how the olfactory network is organized to serve this unique set of functions. Leveraging a large and high-quality resting-state functional magnetic resonance imaging (rs-fMRI) dataset of nearly 900 participants from the Human Connectome Project (HCP), we identified a human olfactory network encompassing cortical and subcortical regions across the temporal and frontal lobes. Highlighting its reliability and generalizability, the connectivity matrix of this olfactory network mapped closely onto that extracted from an independent rs-fMRI dataset. Graph theoretical analysis further explicated the organizational principles of the network. The olfactory network exhibits a modular composition of three (i.e., the sensory, limbic, and frontal) subnetworks and demonstrates strong small-world properties, high in both global integration and local segregation (i.e., circuit specialization). This network organization thus ensures the segregation of local circuits, which are nonetheless integrated via connecting hubs [i.e., amygdala (AMY) and anterior insula (INSa)], thereby enabling the specialized, yet integrative, functions of olfaction. In particular, the degree of local segregation positively predicted olfactory discrimination performance in the independent sample, which we infer as a functional advantage of the network organization. In sum, an olfactory functional network has been identified through the large HCP dataset, affording a representative template of the human olfactory functional neuroanatomy. Importantly, the topological analysis of the olfactory network provides network-level insights into the remarkable functional specialization and spatial segregation of the olfactory system.


Subject(s)
Connectome , Brain/diagnostic imaging , Humans , Magnetic Resonance Imaging , Nerve Net/diagnostic imaging , Reproducibility of Results , Smell
13.
Annu Rev Psychol ; 71: 139-164, 2020 01 04.
Article in English | MEDLINE | ID: mdl-31561741

ABSTRACT

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.


Subject(s)
Brain , Feeding Behavior , Food , Gastrointestinal Tract , Reward , Animals , Humans
14.
Appetite ; 139: 145-151, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31029689

ABSTRACT

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.


Subject(s)
Brain/physiology , Duodenum/innervation , Food Preferences/physiology , Nutrients/physiology , Vagus Nerve/physiology , Animals , Digestion/physiology , Duodenum/surgery , Gastric Bypass , Learning/physiology , Male , Mice , Mice, Inbred C57BL , Vagus Nerve/surgery
15.
Front Integr Neurosci ; 12: 57, 2018.
Article in English | MEDLINE | ID: mdl-30519164

ABSTRACT

In most species, including humans, food preference is primarily controlled by nutrient value. In particular, glucose-containing sugars exert exquisitely strong effects on food choice via gut-generated signals. However, the identity of the visceral signals underlying glucose's rewarding effects remains uncertain. In particular, it is unknown whether sugar metabolism mediates the formation of preferences for glucose-containing sugars. Using the mouse as a model organism, we made use of a combination of conditioning schedules, gastrointestinal nutrient administration, and chromatographic/electrochemical methods to assess the behavioral and neural effects of activating the gut with either metabolizable glucose or a non-metabolizable glucose analog. We show that mice display much superior preferences for flavors associated with intra-gastric infusions of glucose compared to flavors associated with intra-gastric infusions of the non-metabolizable glucose analog α-methyl-D-glucopyranoside ("MDG," an activator of intestinal sodium/glucose co-transporters). These effects were unaffected by surgical bypassing of the duodenum, suggesting glucose-specific post-absorptive sensing mechanisms. Consistently, intra-portal infusions of glucose, but not of MDG, induced significant rises in dopamine (DA) levels within brain reward circuits. Our data reveal that the unmatched rewarding effects of glucose-containing sugars cannot be accounted for by metabolism-independent activation of sodium/glucose cotransporters; rather, they point to glucose metabolism as the physiological mechanism underlying the potent reward value of sugar-sweetened flavored beverages. In particular, no circulating "gut factors" need to be invoked to explain the reward value of ingested glucose. Thus, instead of circulating gut hormones, portal-mesenteric sensing of glucose emerges as the preferential physiological pathway for sugar reward.

17.
Cell ; 175(3): 665-678.e23, 2018 10 18.
Article in English | MEDLINE | ID: mdl-30245012

ABSTRACT

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.


Subject(s)
Intestines/physiology , Reward , Substantia Nigra/physiology , Vagus Nerve/physiology , Afferent Pathways/metabolism , Afferent Pathways/physiology , Animals , Dopamine/metabolism , Dopaminergic Neurons/physiology , Glutamic Acid/metabolism , Intestines/innervation , Male , Mice , Mice, Inbred C57BL , Optogenetics
18.
Cell ; 168(1-2): 311-324.e18, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28086095

ABSTRACT

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.


Subject(s)
Central Amygdaloid Nucleus/physiology , Predatory Behavior , Animals , Anxiety/metabolism , Central Amygdaloid Nucleus/anatomy & histology , Electromyography , Interneurons/metabolism , Mandible/anatomy & histology , Mandible/innervation , Mandible/physiology , Mice , Neck/anatomy & histology , Neck/innervation , Neck/physiology , Neurons/cytology , Neurons/physiology , Periaqueductal Gray/physiology
19.
Cell Metab ; 25(2): 335-344, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28065827

ABSTRACT

Bariatric surgery remains the single most effective long-term treatment modality for morbid obesity, achieved mainly by lowering caloric intake through as yet ill-defined mechanisms. Here we show in rats that Roux-en-Y gastric bypass (RYGB)-like rerouting of ingested fat mobilizes lower small intestine production of the fat-satiety molecule oleoylethanolamide (OEA). This was associated with vagus nerve-driven increases in dorsal striatal dopamine release. We also demonstrate that RYGB upregulates striatal dopamine 1 receptor (D1R) expression specifically under high-fat diet feeding conditions. Mechanistically, interfering with local OEA, vagal, and dorsal striatal D1R signaling negated the beneficial effects of RYGB on fat intake and preferences. These findings delineate a molecular/systems pathway through which bariatric surgery improves feeding behavior and may aid in the development of novel weight loss strategies that similarly modify brain reward circuits compromised in obesity.


Subject(s)
Appetite/drug effects , Dietary Fats/pharmacology , Gastric Bypass , Gastrointestinal Tract/metabolism , Neostriatum/metabolism , Obesity/metabolism , Obesity/physiopathology , PPAR alpha/metabolism , Receptors, Dopamine D1/metabolism , Signal Transduction , Administration, Oral , Animals , Body Weight/drug effects , Diet, High-Fat , Dopamine/metabolism , Endocannabinoids/metabolism , Feeding Behavior/drug effects , Food Preferences/drug effects , Gastrointestinal Tract/drug effects , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice, Obese , Models, Biological , Neostriatum/drug effects , Oleic Acids/metabolism , Rats, Wistar , Signal Transduction/drug effects , Vagus Nerve/drug effects , Vagus Nerve/metabolism , Weight Loss/drug effects
20.
eNeuro ; 3(2)2016.
Article in English | MEDLINE | ID: mdl-27257625

ABSTRACT

The influence of micronutrients on dopamine systems is not well defined. Using mice, we show a potential role for reduced dietary vitamin D3 (cholecalciferol) in promoting diet-induced obesity (DIO), food intake, and drug consumption while on a high fat diet. To complement these deficiency studies, treatments with exogenous fully active vitamin D3 (calcitriol, 10 µg/kg, i.p.) were performed. Nondeficient mice that were made leptin resistant with a high fat diet displayed reduced food intake and body weight after an acute treatment with exogenous calcitriol. Dopamine neurons in the midbrain and their target neurons in the striatum were found to express vitamin D3 receptor protein. Acute calcitriol treatment led to transcriptional changes of dopamine-related genes in these regions in naive mice, enhanced amphetamine-induced dopamine release in both naive mice and rats, and increased locomotor activity after acute amphetamine treatment (2.5 mg/kg, i.p.). Alternatively, mice that were chronically fed either the reduced D3 high fat or chow diets displayed less activity after acute amphetamine treatment compared with their respective controls. Finally, high fat deficient mice that were trained to orally consume liquid amphetamine (90 mg/L) displayed increased consumption, while nondeficient mice treated with calcitriol showed reduced consumption. Our findings suggest that reduced dietary D3 may be a contributing environmental factor enhancing DIO as well as drug intake while eating a high fat diet. Moreover, these data demonstrate that dopamine circuits are modulated by D3 signaling, and may serve as direct or indirect targets for exogenous calcitriol.


Subject(s)
Amphetamine/pharmacology , Calcitriol/pharmacology , Central Nervous System Agents/pharmacology , Corpus Striatum/drug effects , Diet, High-Fat/adverse effects , Obesity/drug therapy , Animals , Body Weight/drug effects , Body Weight/physiology , Cholecalciferol/deficiency , Cholecalciferol/metabolism , Corpus Striatum/metabolism , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Eating/drug effects , Eating/physiology , Male , Mice, Inbred C57BL , Motor Activity/drug effects , Motor Activity/physiology , Obesity/metabolism , Obesity/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...