Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters











Publication year range
1.
Nature ; 632(8025): 585-593, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38987598

ABSTRACT

The most successful obesity therapeutics, glucagon-like peptide-1 receptor (GLP1R) agonists, cause aversive responses such as nausea and vomiting1,2, effects that may contribute to their efficacy. Here, we investigated the brain circuits that link satiety to aversion, and unexpectedly discovered that the neural circuits mediating these effects are functionally separable. Systematic investigation across drug-accessible GLP1R populations revealed that only hindbrain neurons are required for the efficacy of GLP1-based obesity drugs. In vivo two-photon imaging of hindbrain GLP1R neurons demonstrated that most neurons are tuned to either nutritive or aversive stimuli, but not both. Furthermore, simultaneous imaging of hindbrain subregions indicated that area postrema (AP) GLP1R neurons are broadly responsive, whereas nucleus of the solitary tract (NTS) GLP1R neurons are biased towards nutritive stimuli. Strikingly, separate manipulation of these populations demonstrated that activation of NTSGLP1R neurons triggers satiety in the absence of aversion, whereas activation of APGLP1R neurons triggers strong aversion with food intake reduction. Anatomical and behavioural analyses revealed that NTSGLP1R and APGLP1R neurons send projections to different downstream brain regions to drive satiety and aversion, respectively. Importantly, GLP1R agonists reduce food intake even when the aversion pathway is inhibited. Overall, these findings highlight NTSGLP1R neurons as a population that could be selectively targeted to promote weight loss while avoiding the adverse side effects that limit treatment adherence.


Subject(s)
Anti-Obesity Agents , Avoidance Learning , Glucagon-Like Peptide-1 Receptor , Neural Pathways , Rhombencephalon , Satiety Response , Animals , Female , Male , Mice , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/pharmacology , Area Postrema/metabolism , Area Postrema/drug effects , Avoidance Learning/drug effects , Avoidance Learning/physiology , Eating/drug effects , Eating/physiology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Mice, Inbred C57BL , Neural Pathways/drug effects , Neurons/metabolism , Neurons/physiology , Neurons/drug effects , Obesity/metabolism , Rhombencephalon/cytology , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Rhombencephalon/physiology , Satiety Response/drug effects , Satiety Response/physiology , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Solitary Nucleus/physiology , Food
2.
Cell Rep ; 42(11): 113338, 2023 11 28.
Article in English | MEDLINE | ID: mdl-37910501

ABSTRACT

Caloric restriction has anti-inflammatory effects. However, the coordinated physiological actions that lead to reduced inflammation in a state of caloric deficit (hunger) are largely unknown. Using a mouse model of injury-induced peripheral inflammation, we find that food deprivation reduces edema, temperature, and cytokine responses that occur after injury. The magnitude of the anti-inflammatory effect that occurs during hunger is more robust than that of non-steroidal anti-inflammatory drugs. The effects of hunger are recapitulated centrally by activity in nutrient-sensing hypothalamic agouti-related protein (AgRP)-expressing neurons. We find that AgRP neurons projecting to the paraventricular nucleus of the hypothalamus rapidly and robustly reduce inflammation and mediate the majority of hunger's anti-inflammatory effects. Intact vagal efferent signaling is required for the anti-inflammatory action of hunger, revealing a brain-to-periphery pathway for this reduction in inflammation. Taken together, these data begin to unravel a potent anti-inflammatory pathway engaged by hypothalamic AgRP neurons to reduce inflammation.


Subject(s)
Hunger , Hypothalamus , Humans , Hunger/physiology , Agouti-Related Protein/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Inflammation/metabolism
3.
Mol Metab ; 78: 101833, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37925021

ABSTRACT

OBJECTIVE: The learned associations between sensory cues (e.g., taste, smell) and nutritive value (e.g., calories, post-ingestive signaling) of foods powerfully influences our eating behavior [1], but the neural circuits that mediate these associations are not well understood. Here, we examined the role of agouti-related protein (AgRP)-expressing neurons - neurons which are critical drivers of feeding behavior [2; 3] - in mediating flavor-nutrient learning (FNL). METHODS: Because mice prefer flavors associated with AgRP neuron activity suppression [4], we examined how optogenetic stimulation of AgRP neurons during intake influences FNL, and used fiber photometry to determine how endogenous AgRP neuron activity tracks associations between flavors and nutrients. RESULTS: We unexpectedly found that tonic activity in AgRP neurons during FNL potentiated, rather than prevented, the development of flavor preferences. There were notable sex differences in the mechanisms for this potentiation. Specifically, in male mice, AgRP neuron activity increased flavor consumption during FNL training, thereby strengthening the association between flavors and nutrients. In female mice, AgRP neuron activity enhanced flavor-nutrient preferences independently of consumption during training, suggesting that AgRP neuron activity enhances the reward value of the nutrient-paired flavor. Finally, in vivo neural activity analyses demonstrated that acute AgRP neuron dynamics track the association between flavors and nutrients in both sexes. CONCLUSIONS: Overall, these data (1) demonstrate that AgRP neuron activity enhances associations between flavors and nutrients in a sex-dependent manner and (2) reveal that AgRP neurons track and rapidly update these associations. Taken together, our findings provide new insight into the role of AgRP neurons in assimilating sensory and nutritive signals for food reinforcement.


Subject(s)
Eating , Feeding Behavior , Animals , Female , Male , Mice , Agouti-Related Protein/metabolism , Eating/physiology , Energy Intake , Feeding Behavior/physiology , Neurons/metabolism
4.
bioRxiv ; 2023 Sep 22.
Article in English | MEDLINE | ID: mdl-37786670

ABSTRACT

Objective: The learned associations between sensory cues (e.g., taste, smell) and nutritive value (e.g., calories, post-ingestive signaling) of foods powerfully influences our eating behavior [1], but the neural circuits that mediate these associations are not well understood. Here, we examined the role of agouti-related protein (AgRP)-expressing neurons - neurons which are critical drivers of feeding behavior [2; 3] - in mediating flavor-nutrient learning (FNL). Methods: Because mice prefer flavors associated with AgRP neuron activity suppression [4], we examined how optogenetic stimulation of AgRP neurons during intake influences FNL, and used fiber photometry to determine how endogenous AgRP neuron activity tracks associations between flavors and nutrients. Results: We unexpectedly found that tonic activity in AgRP neurons during FNL potentiated, rather than prevented, the development of flavor preferences. There were notable sex differences in the mechanisms for this potentiation. Specifically, in male mice, AgRP neuron activity increased flavor consumption during FNL training, thereby strengthening the association between flavors and nutrients. In female mice, AgRP neuron activity enhanced flavor-nutrient preferences independently of consumption during training, suggesting that AgRP neuron activity enhances the reward value of the nutrient-paired flavor. Finally, in vivo neural activity analyses demonstrated that acute AgRP neuron dynamics track the association between flavors and nutrients in both sexes. Conclusions: Overall, these data (1) demonstrate that AgRP neuron activity enhances associations between flavors and nutrients in a sex-dependent manner and (2) reveal that AgRP neurons track and update these associations on fast timescales. Taken together, our findings provide new insight into the role of AgRP neurons in assimilating sensory and nutritive signals for food reinforcement.

5.
Cell Mol Gastroenterol Hepatol ; 16(2): 189-199, 2023.
Article in English | MEDLINE | ID: mdl-37172823

ABSTRACT

The detection of nutrients in the gut influences ongoing and future feeding behavior as well as the development of food preferences. In addition to nutrient sensing in the intestine, the hepatic portal vein plays a considerable role in detecting ingested nutrients and conveying this information to brain nuclei involved in metabolism, learning, and reward. Here, we review mechanisms underlying hepatic portal vein sensing of nutrients, particularly glucose, and how this is relayed to the brain to influence feeding behavior and reward. We additionally highlight several gaps where future research can provide new insights into the effects of portal nutrients on neural activity in the brain and feeding behavior.


Subject(s)
Glucose , Portal Vein , Portal Vein/metabolism , Glucose/metabolism , Feeding Behavior , Reward , Eating
6.
Nature ; 612(7941): 739-747, 2022 12.
Article in English | MEDLINE | ID: mdl-36517598

ABSTRACT

Exercise exerts a wide range of beneficial effects for healthy physiology1. However, the mechanisms regulating an individual's motivation to engage in physical activity remain incompletely understood. An important factor stimulating the engagement in both competitive and recreational exercise is the motivating pleasure derived from prolonged physical activity, which is triggered by exercise-induced neurochemical changes in the brain. Here, we report on the discovery of a gut-brain connection in mice that enhances exercise performance by augmenting dopamine signalling during physical activity. We find that microbiome-dependent production of endocannabinoid metabolites in the gut stimulates the activity of TRPV1-expressing sensory neurons and thereby elevates dopamine levels in the ventral striatum during exercise. Stimulation of this pathway improves running performance, whereas microbiome depletion, peripheral endocannabinoid receptor inhibition, ablation of spinal afferent neurons or dopamine blockade abrogate exercise capacity. These findings indicate that the rewarding properties of exercise are influenced by gut-derived interoceptive circuits and provide a microbiome-dependent explanation for interindividual variability in exercise performance. Our study also suggests that interoceptomimetic molecules that stimulate the transmission of gut-derived signals to the brain may enhance the motivation for exercise.


Subject(s)
Brain-Gut Axis , Dopamine , Exercise , Gastrointestinal Microbiome , Motivation , Running , Animals , Mice , Brain/cytology , Brain/metabolism , Dopamine/metabolism , Endocannabinoids/antagonists & inhibitors , Endocannabinoids/metabolism , Sensory Receptor Cells/metabolism , Brain-Gut Axis/physiology , Gastrointestinal Microbiome/physiology , Exercise/physiology , Exercise/psychology , Physical Conditioning, Animal/physiology , Physical Conditioning, Animal/psychology , Models, Animal , Humans , Ventral Striatum/cytology , Ventral Striatum/metabolism , Running/physiology , Running/psychology , Reward , Individuality
7.
Neuron ; 110(2): 180-182, 2022 01 19.
Article in English | MEDLINE | ID: mdl-35051361

ABSTRACT

Humans and animals alike perform behaviors-like putting on a sweater or building a warm nest-to regulate body temperature. In this issue of Neuron, Jung et al. (2022) reveal a parabrachial nucleus-to-lateral hypothalamus circuit that regulates thermoregulatory behavior, a circuit distinct from that which governs motivated feeding behavior.


Subject(s)
Hunger , Hypothalamic Area, Lateral , Animals , Body Temperature Regulation , Feeding Behavior/physiology , Hypothalamic Area, Lateral/physiology , Neurons/physiology
8.
Nature ; 600(7888): 269-273, 2021 12.
Article in English | MEDLINE | ID: mdl-34789878

ABSTRACT

The brain is the seat of body weight homeostasis. However, our inability to control the increasing prevalence of obesity highlights a need to look beyond canonical feeding pathways to broaden our understanding of body weight control1-3. Here we used a reverse-translational approach to identify and anatomically, molecularly and functionally characterize a neural ensemble that promotes satiation. Unbiased, task-based functional magnetic resonance imaging revealed marked differences in cerebellar responses to food in people with a genetic disorder characterized by insatiable appetite. Transcriptomic analyses in mice revealed molecularly and topographically -distinct neurons in the anterior deep cerebellar nuclei (aDCN) that are activated by feeding or nutrient infusion in the gut. Selective activation of aDCN neurons substantially decreased food intake by reducing meal size without compensatory changes to metabolic rate. We found that aDCN activity terminates food intake by increasing striatal dopamine levels and attenuating the phasic dopamine response to subsequent food consumption. Our study defines a conserved satiation centre that may represent a novel therapeutic target for the management of excessive eating, and underscores the utility of a 'bedside-to-bench' approach for the identification of neural circuits that influence behaviour.


Subject(s)
Body Weight Maintenance/genetics , Body Weight Maintenance/physiology , Cerebellum/physiology , Food , Protein Biosynthesis , Reverse Genetics , Satiety Response/physiology , Adult , Animals , Appetite Regulation/genetics , Appetite Regulation/physiology , Cerebellar Nuclei/cytology , Cerebellar Nuclei/physiology , Cerebellum/cytology , Cues , Dopamine/metabolism , Eating/genetics , Eating/physiology , Feeding Behavior/physiology , Female , Homeostasis , Humans , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Neostriatum/metabolism , Neurons/physiology , Obesity/genetics , Philosophy , Young Adult
9.
Science ; 374(6567): 547-548, 2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34709901
10.
Endocrinology ; 162(5)2021 05 01.
Article in English | MEDLINE | ID: mdl-33558881

ABSTRACT

Appropriate food intake requires exquisite coordination between the gut and the brain. Indeed, it has long been known that gastrointestinal signals communicate with the brain to promote or inhibit feeding behavior. Recent advances in the ability to monitor and manipulate neural activity in awake, behaving rodents has facilitated important discoveries about how gut signaling influences neural activity and feeding behavior. This review emphasizes recent studies that have advanced our knowledge of gut-brain signaling and food intake control, with a focus on how gut signaling influences in vivo neural activity in animal models. Moving forward, dissecting the complex pathways and circuits that transmit nutritive signals from the gut to the brain will reveal fundamental principles of energy balance, ultimately enabling new treatment strategies for diseases rooted in body weight control.


Subject(s)
Brain/metabolism , Eating , Gastrointestinal Tract/metabolism , Animals , Feeding Behavior , Gastrointestinal Tract/innervation , Humans , Signal Transduction
11.
Cell Metab ; 33(3): 676-687.e5, 2021 03 02.
Article in English | MEDLINE | ID: mdl-33450178

ABSTRACT

Food intake is tightly regulated by complex and coordinated gut-brain interactions. Nutrients rapidly modulate activity in key populations of hypothalamic neurons that regulate food intake, including hunger-sensitive agouti-related protein (AgRP)-expressing neurons. Because individual macronutrients engage specific receptors in the gut to communicate with the brain, we reasoned that macronutrients may utilize different pathways to reduce activity in AgRP neurons. Here, we revealed that AgRP neuron activity in hungry mice is inhibited by site-specific intestinal detection of different macronutrients. We showed that vagal gut-brain signaling is required for AgRP neuron inhibition by fat. In contrast, spinal gut-brain signaling relays the presence of intestinal glucose. Further, we identified glucose sensors in the intestine and hepatic portal vein that mediate glucose-dependent AgRP neuron inhibition. Therefore, distinct pathways are activated by individual macronutrients to inhibit AgRP neuron activity.


Subject(s)
Intestines/physiology , Neurons/metabolism , Nutrients/metabolism , Agouti-Related Protein/metabolism , Animals , Brain/drug effects , Brain/physiology , Dietary Fats/metabolism , Dietary Fats/pharmacology , Glucose/metabolism , Glucose/pharmacology , Intestinal Mucosa/metabolism , Intestines/drug effects , Mice , Mice, Inbred C57BL , Nutrients/pharmacology , Signal Transduction/drug effects , Sodium-Glucose Transporter 1/metabolism , Vagus Nerve/drug effects , Vagus Nerve/physiology
12.
Am J Clin Nutr ; 113(1): 232-245, 2021 Jan 04.
Article in English | MEDLINE | ID: mdl-33300030

ABSTRACT

In November 2019, the NIH held the "Sensory Nutrition and Disease" workshop to challenge multidisciplinary researchers working at the interface of sensory science, food science, psychology, neuroscience, nutrition, and health sciences to explore how chemosensation influences dietary choice and health. This report summarizes deliberations of the workshop, as well as follow-up discussion in the wake of the current pandemic. Three topics were addressed: A) the need to optimize human chemosensory testing and assessment, B) the plasticity of chemosensory systems, and C) the interplay of chemosensory signals, cognitive signals, dietary intake, and metabolism. Several ways to advance sensory nutrition research emerged from the workshop: 1) refining methods to measure chemosensation in large cohort studies and validating measures that reflect perception of complex chemosensations relevant to dietary choice; 2) characterizing interindividual differences in chemosensory function and how they affect ingestive behaviors, health, and disease risk; 3) defining circuit-level organization and function that link and interact with gustatory, olfactory, homeostatic, visceral, and cognitive systems; and 4) discovering new ligands for chemosensory receptors (e.g., those produced by the microbiome) and cataloging cell types expressing these receptors. Several of these priorities were made more urgent by the current pandemic because infection with sudden acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the ensuing coronavirus disease of 2019 has direct short- and perhaps long-term effects on flavor perception. There is increasing evidence of functional interactions between the chemosensory and nutritional sciences. Better characterization of this interface is expected to yield insights to promote health, mitigate disease risk, and guide nutrition policy.

13.
Physiol Behav ; 224: 113039, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32610101

ABSTRACT

Dysfunction in neurophysiological systems that regulate food intake and metabolism are at least partly responsible for obesity and related comorbidities. An important component of this process is the hypothalamic melanocortin system, where an imbalance can result in severe obesity and deficits in glucose metabolism. Exercise offers many health benefits related to cardiovascular improvements, hunger control, and blood glucose homeostasis. However, the molecular mechanism underlying the exercise-induced improvements to the melanocortin system remain undefined. Here, we review the role of the melanocortin system to sense hormonal, nutrient, and neuronal signals of energy status. This information is then relayed onto secondary neurons in order to regulate physiological parameters, which promote proper energy and glucose balance. We also provide an overview on the effects of physical exercise to induce biophysical changes in the melanocortin circuit which may regulate food intake, glucose metabolism and improve overall metabolic health.


Subject(s)
Energy Metabolism , Melanocortins , Homeostasis , Humans , Hypothalamus , Obesity
14.
Neuroscience ; 450: 126-134, 2020 12 01.
Article in English | MEDLINE | ID: mdl-32540365

ABSTRACT

Unpleasant somatosensory stimuli such as pain and itch can interrupt normal behavior. But survival can depend on resuming normal behavior before these challenges are fully resolved. The neural mechanisms that prioritize behavior when individuals are challenged with unpleasant somatosensory sensations, however, are not fully understood. Recently, we identified a neural circuit activated by hunger that can inhibit pain, prioritizing food seeking over tending to an injury. Here, we examine the ability of hunger, and neurons activated by hunger, to inhibit behavioral responses to another unpleasant somatosensory sensation - itch. We demonstrate that food deprivation inhibits scratching induced by three different pruritogenic stimuli: histamine, serotonin, and chloroquine. The inhibition of scratching correlates with the level of food deprivation, suggesting a cross-competition of alarm systems in the brain whereby more energy need more efficiently inhibits competing drives. Finally, we show that activity in hunger-sensitive, hypothalamic agouti-related protein (AgRP)-expressing neurons is sufficient to inhibit itch. Taken together, we showed that hunger or AgRP neuron activity inhibits itch, demonstrating that organisms have neural systems to filter and process ascending spinal signals activated by unpleasant somatosensory stimuli to prioritize salient needs.


Subject(s)
Hunger , Neurons , Agouti-Related Protein/metabolism , Food Deprivation , Humans , Hypothalamus/metabolism , Neurons/metabolism
15.
Brain Res ; 1724: 146441, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31513793

ABSTRACT

Hunger resulting from food deprivation is associated with negative affect. This is supported by recent evidence showing that hunger-sensitive neurons drive feeding through a negative valence teaching signal. However, the complementary hypothesis that hormonal signals of energy surfeit counteract this negative valence, or even transmit positive valence, has received less attention. The adipose-derived hormone leptin signals in proportion to fat mass, is an indicator of energy surplus, and reduces food intake. Here, we showed that centrally-delivered leptin reduced food intake and conditioned a place preference in food-restricted as well as ad libitum fed rats. In contrast, leptin did not reduce food intake nor condition a place preference in obese rats, likely due to leptin resistance. Despite a well-known role for hindbrain leptin receptor signaling in energy balance control, hindbrain leptin delivery did not condition a place preference in food-restricted rats, suggesting that leptin acting in midbrain or forebrain sites mediates place preference conditioning. Supporting the hypothesis that leptin signaling induces a positive affective state, leptin also decreased the threshold for ventral tegmental area brain stimulation reward. Together, these data suggest that leptin signaling is intrinsically preferred, and support the view that signals of energy surfeit are associated with positive affect. Harnessing the positive valence of signals such as leptin may attenuate the negative affect associated with hunger, providing a compelling new approach for weight loss maintenance.


Subject(s)
Eating/drug effects , Feeding Behavior/physiology , Leptin/metabolism , Affect/physiology , Animals , Conditioning, Classical/physiology , Emotions/physiology , Energy Metabolism/physiology , Feeding Behavior/drug effects , Food , Food Deprivation/physiology , Leptin/physiology , Male , Obesity , Rats , Rats, Sprague-Dawley , Receptors, Leptin/metabolism , Reward , Rhombencephalon/metabolism , Signal Transduction/drug effects , Ventral Tegmental Area/metabolism
16.
Aging Cell ; 18(5): e13014, 2019 10.
Article in English | MEDLINE | ID: mdl-31373126

ABSTRACT

The mechanistic target of rapamycin (mTOR) is an evolutionarily conserved protein kinase that regulates growth and metabolism. mTOR is found in two protein complexes, mTORC1 and mTORC2, that have distinct components and substrates and are both inhibited by rapamycin, a macrolide drug that robustly extends lifespan in multiple species including worms and mice. Although the beneficial effect of rapamycin on longevity is generally attributed to reduced mTORC1 signaling, disruption of mTORC2 signaling can also influence the longevity of worms, either positively or negatively depending on the temperature and food source. Here, we show that loss of hypothalamic mTORC2 signaling in mice decreases activity level, increases the set point for adiposity, and renders the animals susceptible to diet-induced obesity. Hypothalamic mTORC2 signaling normally increases with age, and mice lacking this pathway display higher fat mass and impaired glucose homeostasis throughout life, become more frail with age, and have decreased overall survival. We conclude that hypothalamic mTORC2 is essential for the normal metabolic health, fitness, and lifespan of mice. Our results have implications for the use of mTORC2-inhibiting pharmaceuticals in the treatment of brain cancer and diseases of aging.


Subject(s)
Hypothalamus/metabolism , Longevity , Mechanistic Target of Rapamycin Complex 2/metabolism , Animals , Female , Mice , Mice, Inbred C57BL
17.
Neuron ; 103(5): 891-908.e6, 2019 09 04.
Article in English | MEDLINE | ID: mdl-31277924

ABSTRACT

Motivated behavior is influenced by neural networks that integrate physiological needs. Here, we describe coordinated regulation of hypothalamic feeding and midbrain reward circuits in awake behaving mice. We find that alcohol and other non-nutritive drugs inhibit activity in hypothalamic feeding neurons. Interestingly, nutrients and drugs utilize different pathways for the inhibition of hypothalamic neuron activity, as alcohol signals hypothalamic neurons in a vagal-independent manner, while fat and satiation signals require the vagus nerve. Concomitantly, nutrients, alcohol, and drugs also increase midbrain dopamine signaling. We provide evidence that these changes are interdependent, as modulation of either hypothalamic neurons or midbrain dopamine signaling influences reward-evoked activity changes in the other population. Taken together, our results demonstrate that (1) food and drugs can engage at least two peripheral→central pathways to influence hypothalamic neuron activity, and (2) hypothalamic and dopamine circuits interact in response to rewards.


Subject(s)
Central Nervous System Depressants/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopaminergic Neurons/drug effects , Ethanol/pharmacology , Feeding Behavior/drug effects , Hypothalamus/drug effects , Nicotinic Agonists/pharmacology , Reward , Agouti-Related Protein/metabolism , Amphetamine/pharmacology , Animals , Cocaine/pharmacology , Dopamine Antagonists/pharmacology , Dopaminergic Neurons/metabolism , Hypothalamus/metabolism , Mice , Neural Pathways/drug effects , Neurons/drug effects , Neurons/metabolism , Nicotine/pharmacology , Pro-Opiomelanocortin/metabolism , Vagotomy , Vagus Nerve/physiology
18.
Neuron ; 100(6): 1277-1279, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30571939

ABSTRACT

The brain orchestrates a variety of responses to noxious environmental stimuli, from reflexive movements to coordinated defensive behaviors. In this issue of Neuron, Barik et al. identify a hindbrain circuit essential for escape behaviors (Barik et al., 2018).


Subject(s)
Hot Temperature , Rhombencephalon , Animals , Behavior, Animal , Brain Stem , Escape Reaction
19.
Mol Metab ; 18: 107-119, 2018 12.
Article in English | MEDLINE | ID: mdl-30292523

ABSTRACT

OBJECTIVE: Hypothalamic Pro-opiomelanocortin (POMC) and Neuropeptide Y/Agouti-Related Peptide (NPY/AgRP) neurons are critical nodes of a circuit within the brain that sense key metabolic cues as well as regulate metabolism. Importantly, these neurons retain an innate ability to rapidly reorganize synaptic inputs and electrophysiological properties in response to metabolic state. While the cellular properties of these neurons have been investigated in the context of obesity, much less is known about the effects of exercise training. METHODS: In order to further investigate this issue, we utilized neuron-specific transgenic mouse models to identify POMC and NPY/AgRP neurons for patch-clamp electrophysiology experiments. RESULTS: Using whole-cell patch-clamp electrophysiology, we found exercise depolarized and increased firing rate of arcuate POMC neurons. The increased excitability of POMC neurons was concomitant with increased excitatory inputs to these neurons. In agreement with recent work suggesting leptin plays an important role in the synaptic (re)organization of POMC neurons, POMC neurons which express leptin receptors were more sensitive to exercise-induced changes in biophysical properties. Opposite to effects observed in POMC neurons, NPY neurons were shunted toward inhibition following exercise. CONCLUSIONS: Together, these data support a rapid reorganization of synaptic inputs and biophysical properties in response to exercise, which may facilitate adaptations to altered energy balance and glucose metabolism.


Subject(s)
Hypothalamus/physiology , Neurons/physiology , Neuropeptide Y/metabolism , Physical Conditioning, Animal , Pro-Opiomelanocortin/metabolism , Synapses/physiology , Action Potentials , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Hypothalamus/cytology , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neuropeptide Y/genetics , Pro-Opiomelanocortin/genetics , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Synapses/metabolism , Synaptic Potentials
20.
Cell ; 173(1): 140-152.e15, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29570993

ABSTRACT

Hunger and pain are two competing signals that individuals must resolve to ensure survival. However, the neural processes that prioritize conflicting survival needs are poorly understood. We discovered that hunger attenuates behavioral responses and affective properties of inflammatory pain without altering acute nociceptive responses. This effect is centrally controlled, as activity in hunger-sensitive agouti-related protein (AgRP)-expressing neurons abrogates inflammatory pain. Systematic analysis of AgRP projection subpopulations revealed that the neural processing of hunger and inflammatory pain converge in the hindbrain parabrachial nucleus (PBN). Strikingly, activity in AgRP → PBN neurons blocked the behavioral response to inflammatory pain as effectively as hunger or analgesics. The anti-nociceptive effect of hunger is mediated by neuropeptide Y (NPY) signaling in the PBN. By investigating the intersection between hunger and pain, we have identified a neural circuit that mediates competing survival needs and uncovered NPY Y1 receptor signaling in the PBN as a target for pain suppression.


Subject(s)
Neurons/metabolism , Pain/pathology , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Analgesics, Opioid/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Behavior, Animal/drug effects , Diet , Feeding Behavior/drug effects , Formaldehyde/toxicity , Glutamate Decarboxylase/metabolism , Locomotion/drug effects , Mice , Mice, Inbred C57BL , Morphine/pharmacology , Neurons/drug effects , Pain/etiology , Pain/metabolism , Parabrachial Nucleus/drug effects , Parabrachial Nucleus/metabolism , Receptors, Neuropeptide Y/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL