Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Curr Biol ; 34(4): R155-R157, 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38412828

RESUMEN

Satiety-promoting neurons of the hindbrain have long been known for their role in meal termination. An innovative new study now reveals how different hindbrain cell types mediate appetite on distinct timescales.


Asunto(s)
Apetito , Ingestión de Alimentos , Apetito/fisiología , Saciedad , Rombencéfalo , Neuronas
2.
bioRxiv ; 2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-37961449

RESUMEN

Liraglutide and other agonists of the glucagon-like peptide 1 receptor (GLP-1RAs) are effective weight loss drugs, but how they suppress appetite remains unclear. GLP-1RAs inhibit hunger-promoting Agouti-related peptide (AgRP) neurons of the arcuate hypothalamus (Arc) but only indirectly, implicating synaptic afferents to AgRP neurons. To investigate, we developed a method combining rabies-based connectomics with single-nuclei transcriptomics. Applying this method to AgRP neurons in mice predicts 21 afferent subtypes in the mediobasal and paraventricular hypothalamus. Among these are Trh+ Arc neurons (TrhArc), which express the Glp1r gene and are activated by the GLP-1RA liraglutide. Activating TrhArc neurons inhibits AgRP neurons and decreases feeding in an AgRP neuron-dependent manner. Silencing TrhArc neurons increases feeding and body weight and reduces liraglutide's satiating effects. Our results thus demonstrate a widely applicable method for molecular connectomics, reveal the molecular organization of AgRP neuron afferents, and shed light on a neurocircuit through which GLP-1RAs suppress appetite.

3.
Neuron ; 111(18): 2899-2917.e6, 2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37442130

RESUMEN

Motivated behaviors are often studied in isolation to assess labeled lines of neural connections underlying innate actions. However, in nature, multiple systems compete for expression of goal-directed behaviors via complex neural networks. Here, we examined flexible survival decisions in animals tasked with food seeking under predation threat. We found that predator exposure rapidly induced physiological, neuronal, and behavioral adaptations in mice highlighted by reduced food seeking and consumption contingent on current threat level. Diminishing conflict via internal state or external environment perturbations shifted feeding strategies. Predator introduction and/or selective manipulation of danger-responsive cholecystokinin (Cck) cells of the dorsal premammilary nucleus (PMd) suppressed hunger-sensitive Agouti-related peptide (AgRP) neurons, providing a mechanism for threat-evoked hypophagia. Increased caloric need enhanced food seeking under duress through AgRP pathways to the bed nucleus of the stria terminalis (BNST) and/or lateral hypothalamus (LH). Our results suggest oscillating interactions between systems underlying self-preservation and food seeking to promote optimal behavior.


Asunto(s)
Hipotálamo , Neuronas , Ratones , Animales , Proteína Relacionada con Agouti/metabolismo , Hipotálamo/metabolismo , Neuronas/fisiología , Hambre/fisiología , Área Hipotalámica Lateral/fisiología
4.
Trends Endocrinol Metab ; 34(4): 191-193, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36841699

RESUMEN

Calculating and selecting what sensory and homeostatic requirements to attend to at any given time is vital for animals' survival. Tang et al. uncovered a circuit emanating from excitatory cortical neurons that transmit nociceptive information via the hypothalamus to blunt appetite during periods of chronic pain.


Asunto(s)
Apetito , Hambre , Animales , Hambre/fisiología , Dolor , Hipotálamo , Neuronas/fisiología
5.
Mol Psychiatry ; 28(4): 1622-1635, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36577844

RESUMEN

Anorexia nervosa (AN) is a debilitating and deadly disease characterized by low body mass index due to diminished food intake, and oftentimes concurrent hyperactivity. A high percentage of AN behavioral and metabolic phenotypes can be replicated in rodents given access to a voluntary running wheel and subject to food restriction, termed activity-based anorexia (ABA). Despite the well-documented bodyweight loss observed in AN human patients and ABA rodents, much less is understood regarding the neurobiological underpinnings of these maladaptive behaviors. Hunger-promoting hypothalamic agouti-related peptide (AgRP) neurons have been well characterized in their ability to regulate appetite, yet much less is known regarding their activity and function in the mediation of food intake during ABA. Here, feeding microstructure analysis revealed ABA mice decreased food intake due to increased interpellet interval retrieval and diminished meal number. Longitudinal activity recordings of AgRP neurons in ABA animals exhibited a maladaptive inhibitory response to food, independent of basal activity changes. We then demonstrated that ABA development or progression can be mitigated by chemogenetic AgRP activation through the reprioritization of food intake (increased meal number) over hyperactivity, but only during periods of food availability. These results elucidate a potential neural target for the amelioration of behavioral maladaptations present in AN patients.


Asunto(s)
Anorexia Nerviosa , Anorexia , Ratones , Humanos , Animales , Anorexia/metabolismo , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Hipotálamo/metabolismo , Anorexia Nerviosa/metabolismo , Neuronas/metabolismo , Ingestión de Alimentos
6.
Sci Adv ; 8(33): eabn9134, 2022 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-35984878

RESUMEN

Recent data demonstrate that noradrenergic neurons of the locus coeruleus (LC-NE) are required for fear-induced suppression of feeding, but the role of endogenous LC-NE activity in natural, homeostatic feeding remains unclear. Here, we found that LC-NE activity was suppressed during food consumption, and the magnitude of this neural response was attenuated as mice consumed more pellets throughout the session, suggesting that LC responses to food are modulated by satiety state. Visual-evoked LC-NE activity was also attenuated in sated mice, suggesting that satiety state modulates LC-NE encoding of multiple behavioral states. We also found that food intake could be attenuated by brief or longer durations of LC-NE activation. Last, we found that activation of the LC to the lateral hypothalamus pathway suppresses feeding and enhances avoidance and anxiety-like responding. Our findings suggest that LC-NE neurons modulate feeding by integrating both external cues (e.g., anxiogenic environmental cues) and internal drives (e.g., satiety).

7.
Nat Metab ; 4(7): 836-847, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35879462

RESUMEN

The overconsumption of highly caloric and palatable foods has caused a surge in obesity rates in the past half century, thereby posing a healthcare challenge due to the array of comorbidities linked to heightened body fat accrual. Developing treatments to manage body weight requires a grasp of the neurobiological basis of appetite. In this Review, we discuss advances in neuroscience that have identified brain regions and neural circuits that coordinate distinct phases of eating: food procurement, food consumption, and meal termination. While pioneering work identified several hypothalamic nuclei to be involved in feeding, more recent studies have explored how neuronal populations beyond the hypothalamus, such as the mesolimbic pathway and nodes in the hindbrain, interconnect to modulate appetite. We also examine how long-term exposure to a calorically dense diet rewires feeding circuits and alters the response of motivational systems to food. Understanding how the nervous system regulates eating behaviour will bolster the development of medical strategies that will help individuals to maintain a healthy body weight.


Asunto(s)
Apetito , Conducta Alimentaria , Apetito/fisiología , Peso Corporal , Dieta , Conducta Alimentaria/fisiología , Humanos , Obesidad
8.
Elife ; 112022 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-35507386

RESUMEN

Food intake behavior is regulated by a network of appetite-inducing and appetite-suppressing neuronal populations throughout the brain. The parasubthalamic nucleus (PSTN), a relatively unexplored population of neurons in the posterior hypothalamus, has been hypothesized to regulate appetite due to its connectivity with other anorexigenic neuronal populations and because these neurons express Fos, a marker of neuronal activation, following a meal. However, the individual cell types that make up the PSTN are not well characterized, nor are their functional roles in food intake behavior. Here, we identify and distinguish between two discrete PSTN subpopulations, those that express tachykinin-1 (PSTNTac1 neurons) and those that express corticotropin-releasing hormone (PSTNCRH neurons), and use a panel of genetically encoded tools in mice to show that PSTNTac1 neurons play an important role in appetite suppression. Both subpopulations increase activity following a meal and in response to administration of the anorexigenic hormones amylin, cholecystokinin (CCK), and peptide YY (PYY). Interestingly, chemogenetic inhibition of PSTNTac1, but not PSTNCRH neurons, reduces the appetite-suppressing effects of these hormones. Consistently, optogenetic and chemogenetic stimulation of PSTNTac1 neurons, but not PSTNCRH neurons, reduces food intake in hungry mice. PSTNTac1 and PSTNCRH neurons project to distinct downstream brain regions, and stimulation of PSTNTac1 projections to individual anorexigenic populations reduces food consumption. Taken together, these results reveal the functional properties and projection patterns of distinct PSTN cell types and demonstrate an anorexigenic role for PSTNTac1 neurons in the hormonal and central regulation of appetite.


Asunto(s)
Regulación del Apetito , Apetito , Animales , Hormona Liberadora de Corticotropina/metabolismo , Ingestión de Alimentos/fisiología , Ratones , Neuronas/fisiología , Optogenética
9.
Cell Metab ; 34(2): 285-298.e7, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35108515

RESUMEN

The central nervous system has long been thought to regulate insulin secretion, an essential process in the maintenance of blood glucose levels. However, the anatomical and functional connections between the brain and insulin-producing pancreatic ß cells remain undefined. Here, we describe a functional transneuronal circuit connecting the hypothalamus to ß cells in mice. This circuit originates from a subpopulation of oxytocin neurons in the paraventricular hypothalamic nucleus (PVNOXT), and it reaches the islets of the endocrine pancreas via the sympathetic autonomic branch to innervate ß cells. Stimulation of PVNOXT neurons rapidly suppresses insulin secretion and causes hyperglycemia. Conversely, silencing of these neurons elevates insulin levels by dysregulating neuronal signaling and secretory pathways in ß cells and induces hypoglycemia. PVNOXT neuronal activity is triggered by glucoprivation. Our findings reveal that a subset of PVNOXT neurons form functional multisynaptic circuits with ß cells in mice to regulate insulin secretion, and their function is necessary for the ß cell response to hypoglycemia.


Asunto(s)
Células Secretoras de Insulina , Animales , Hipotálamo/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo
10.
Front Behav Neurosci ; 15: 735387, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630052

RESUMEN

The reproducibility crisis (or replication crisis) in biomedical research is a particularly existential and under-addressed issue in the field of behavioral neuroscience, where, in spite of efforts to standardize testing and assay protocols, several known and unknown sources of confounding environmental factors add to variance. Human interference is a major contributor to variability both within and across laboratories, as well as novelty-induced anxiety. Attempts to reduce human interference and to measure more "natural" behaviors in subjects has led to the development of automated home-cage monitoring systems. These systems enable prolonged and longitudinal recordings, and provide large continuous measures of spontaneous behavior that can be analyzed across multiple time scales. In this review, a diverse team of neuroscientists and product developers share their experiences using such an automated monitoring system that combines Noldus PhenoTyper® home-cages and the video-based tracking software, EthoVision® XT, to extract digital biomarkers of motor, emotional, social and cognitive behavior. After presenting our working definition of a "home-cage", we compare home-cage testing with more conventional out-of-cage tests (e.g., the open field) and outline the various advantages of the former, including opportunities for within-subject analyses and assessments of circadian and ultradian activity. Next, we address technical issues pertaining to the acquisition of behavioral data, such as the fine-tuning of the tracking software and the potential for integration with biotelemetry and optogenetics. Finally, we provide guidance on which behavioral measures to emphasize, how to filter, segment, and analyze behavior, and how to use analysis scripts. We summarize how the PhenoTyper has applications to study neuropharmacology as well as animal models of neurodegenerative and neuropsychiatric illness. Looking forward, we examine current challenges and the impact of new developments. Examples include the automated recognition of specific behaviors, unambiguous tracking of individuals in a social context, the development of more animal-centered measures of behavior and ways of dealing with large datasets. Together, we advocate that by embracing standardized home-cage monitoring platforms like the PhenoTyper, we are poised to directly assess issues pertaining to reproducibility, and more importantly, measure features of rodent behavior under more ethologically relevant scenarios.

11.
Nature ; 595(7869): 695-700, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34262177

RESUMEN

Agouti-related peptide (AGRP)-expressing neurons are activated by fasting-this causes hunger1-4, an aversive state that motivates the seeking and consumption of food5,6. Eating returns AGRP neuron activity towards baseline on three distinct timescales: rapidly and transiently following sensory detection of food cues6-8, slowly and longer-lasting in response to nutrients in the gut9,10, and even more slowly and permanently with restoration of energy balance9,11. The rapid regulation by food cues is of particular interest as its neurobiological basis and purpose are unknown. Given that AGRP neuron activity is aversive6, the sensory cue-linked reductions in activity could function to guide behaviour. To evaluate this, we first identified the circuit mediating sensory cue inhibition and then selectively perturbed it to determine function. Here, we show that a lateral hypothalamic glutamatergic â†’ dorsomedial hypothalamic GABAergic (γ-aminobutyric acid-producing)12 → AGRP neuron circuit mediates this regulation. Interference with this circuit impairs food cue inhibition of AGRP neurons and, notably, greatly impairs learning of a sensory cue-initiated food-acquisition task. This is specific for food, as learning of an identical water-acquisition task is unaffected. We propose that decreases in aversive AGRP neuron activity6 mediated by this food-specific circuit increases the incentive salience13 of food cues, and thus facilitates the learning of food-acquisition tasks.


Asunto(s)
Proteína Relacionada con Agouti/metabolismo , Señales (Psicología) , Alimentos , Hambre/fisiología , Vías Nerviosas , Neuronas/fisiología , Animales , Área Hipotalámica Lateral/fisiología , Aprendizaje , Masculino , Ratones , Ratones Endogámicos C57BL , Optogenética
12.
Cell Metab ; 33(7): 1389-1403.e6, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-34038711

RESUMEN

The preoptic area (POA) is a key brain region for regulation of body temperature (Tb), dictating thermogenic, cardiovascular, and behavioral responses that control Tb. Previously characterized POA neuronal populations all reduced Tb when activated. Using mice, we now identify POA neurons expressing bombesin-like receptor 3 (POABRS3) as a population whose activation increased Tb; inversely, acute inhibition of these neurons reduced Tb. POABRS3 neurons that project to either the paraventricular nucleus of the hypothalamus or the dorsomedial hypothalamus increased Tb, heart rate, and blood pressure via the sympathetic nervous system. Long-term inactivation of POABRS3 neurons caused increased Tb variability, overshooting both increases and decreases in Tb set point, with RNA expression profiles suggesting multiple types of POABRS3 neurons. Thus, POABRS3 neuronal populations regulate Tb and heart rate, contribute to cold defense, and fine-tune feedback control of Tb. These findings advance understanding of homeothermy, a defining feature of mammalian biology.


Asunto(s)
Regulación de la Temperatura Corporal , Frecuencia Cardíaca , Neuronas/fisiología , Área Preóptica/metabolismo , Receptores de Bombesina/metabolismo , Animales , Temperatura Corporal/genética , Regulación de la Temperatura Corporal/genética , Frecuencia Cardíaca/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/citología , Neuronas/metabolismo , Área Preóptica/citología , Receptores de Bombesina/genética , Transducción de Señal/genética , Sistema Nervioso Simpático/fisiología , Termogénesis/genética
13.
Elife ; 102021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33779547

RESUMEN

Feeding is critical for survival, and disruption in the mechanisms that govern food intake underlies disorders such as obesity and anorexia nervosa. It is important to understand both food intake and food motivation to reveal mechanisms underlying feeding disorders. Operant behavioral testing can be used to measure the motivational component to feeding, but most food intake monitoring systems do not measure operant behavior. Here, we present a new solution for monitoring both food intake and motivation in rodent home-cages: the Feeding Experimentation Device version 3 (FED3). FED3 measures food intake and operant behavior in rodent home-cages, enabling longitudinal studies of feeding behavior with minimal experimenter intervention. It has a programmable output for synchronizing behavior with optogenetic stimulation or neural recordings. Finally, FED3 design files are open-source and freely available, allowing researchers to modify FED3 to suit their needs.


Obesity and anorexia nervosa are two health conditions related to food intake. Researchers studying these disorders in animal models need to both measure food intake and assess behavioural factors: that is, why animals seek and consume food. Measuring an animal's food intake is usually done by weighing food containers. However, this can be inaccurate due to the small amount of food that rodents eat. As for studying feeding motivation, this can involve calculating the number of times an animal presses a lever to receive a food pellet. These tests are typically conducted in hour-long sessions in temporary testing cages, called operant boxes. Yet, these tests only measure a brief period of a rodent's life. In addition, it takes rodents time to adjust to these foreign environments, which can introduce stress and may alter their feeding behaviour. To address this, Matikainen-Ankney, Earnest, Ali et al. developed a device for monitoring food intake and feeding behaviours around the clock in rodent home cages with minimal experimenter intervention. This 'Feeding Experimentation Device' (FED3) features a pellet dispenser and two 'nose-poke' sensors to measure total food intake, as well as motivation for and learning about food rewards. The battery-powered, wire-free device fits in standard home cages, enabling long-term studies of feeding behaviour with minimal intervention from investigators and less stress on the animals. This means researchers can relate data to circadian rhythms and meal patterns, as Matikainen-Ankney did here. Moreover, the device software is open-source so researchers can customise it to suit their experimental needs. It can also be programmed to synchronise with other instruments used in animal experiments, or across labs running the same behavioural tasks for multi-site studies. Used in this way, it could help improve reproducibility and reliability of results from such studies. In summary, Matikainen-Ankney et al. have presented a new practical solution for studying food-related behaviours in mice and rats. Not only could the device be useful to researchers, it may also be suitable to use in educational settings such as teaching labs and classrooms.


Asunto(s)
Crianza de Animales Domésticos , Condicionamiento Operante , Diseño de Equipo/instrumentación , Conducta Alimentaria , Vivienda para Animales , Roedores/fisiología , Animales , Ingestión de Alimentos , Femenino , Masculino , Ratones
14.
Cell Rep ; 34(7): 108755, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33596417

RESUMEN

Obesity is frequently caused by calorie-rich dietary choices across the animal kingdom. As prandial preference toward a high-fat diet develops in mice, an anti-preference or devaluation of a nutritionally balanced but less palatable standard chow diet occurs concomitantly. Although mechanistic insights underlying devaluation have been observed physiologically in the brain, it is unclear how peripheral sensory processing affects food choice. Because olfactory cues and odor perception help coordinate food preference and intake, we determine the role of smell in the targeted consumption of a high-fat diet and simultaneous devaluation of a standard chow diet. Using inaccessible food and loss-of-function manipulations, we find that olfactory information is neither sufficient nor necessary for both the acute and chronic selection of high-fat diet and coincident diminished value of standard diet. This work suggests alternative means are behind the immediate and sustained consumption of high-fat diet and concurrent standard diet devaluation.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Preferencias Alimentarias/fisiología , Obesidad/fisiopatología , Olfato/fisiología , Animales , Ratones
15.
Nat Neurosci ; 23(10): 1253-1266, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32747789

RESUMEN

Maintaining healthy body weight is increasingly difficult in our obesogenic environment. Dieting efforts are often overpowered by the internal drive to consume energy-dense foods. Although the selection of calorically rich substrates over healthier options is identifiable across species, the mechanisms behind this choice remain poorly understood. Using a passive devaluation paradigm, we found that exposure to high-fat diet (HFD) suppresses the intake of nutritionally balanced standard chow diet (SD) irrespective of age, sex, body mass accrual and functional leptin or melanocortin-4 receptor signaling. Longitudinal recordings revealed that this SD devaluation and subsequent shift toward HFD consumption is encoded at the level of hypothalamic agouti-related peptide neurons and mesolimbic dopamine signaling. Prior HFD consumption vastly diminished the capacity of SD to alleviate the negative valence associated with hunger and the rewarding properties of food discovery even after periods of HFD abstinence. These data reveal a neural basis behind the hardships of dieting.


Asunto(s)
Núcleo Arqueado del Hipotálamo/fisiología , Conducta Consumatoria/fisiología , Dieta Alta en Grasa , Preferencias Alimentarias/fisiología , Neuronas/fisiología , Área Tegmental Ventral/fisiología , Proteína Relacionada con Agouti/fisiología , Animales , Dopamina/fisiología , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Vías Nerviosas/fisiología , Optogenética
16.
Trends Endocrinol Metab ; 31(7): 495-507, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32387196

RESUMEN

Motivated behaviors have fascinated neuroscientists and ethologists for decades due to their necessity for organism survival. Motivations guide behavioral choice through an intricate synthesis of internal state detection, external stimulus exposure, and learned associations. One critical motivation, hunger, provides an accessible example for understanding purposeful behavior. Neuroscientists commonly focus research efforts on neural circuits underlying individual motivations, sacrificing ethological relevance for tight experimental control. This restrictive focus deprives the field of a more nuanced understanding of the unified nervous system in weighing multiple motivations simultaneously and choosing, moment-to-moment, optimal behaviors for survival. Here, we explore the reciprocal interplay between hunger, encoded via hypothalamic neurons marked by the expression of Agouti-related peptide, and alternative need-based motivational systems.


Asunto(s)
Conducta Alimentaria/fisiología , Hambre/fisiología , Motivación/fisiología , Neuronas/metabolismo , Animales , Humanos
17.
Cell ; 179(2): 287-289, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31585075

RESUMEN

Animals use their sense of taste to evaluate the quality and safety of food before ingestion. In this issue of Cell, Zhang and colleagues provide a comprehensive exploration into the elusive mechanisms underlying sour detection.


Asunto(s)
Gusto , Lengua , Animales , Encéfalo
18.
Diabetes ; 68(12): 2210-2222, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31530579

RESUMEN

Melanin-concentrating hormone (MCH) is an important regulator of food intake, glucose metabolism, and adiposity. However, the mechanisms mediating these actions remain largely unknown. We used pharmacological and genetic approaches to show that the sirtuin 1 (SIRT1)/FoxO1 signaling pathway in the hypothalamic arcuate nucleus (ARC) mediates MCH-induced feeding, adiposity, and glucose intolerance. MCH reduces proopiomelanocortin (POMC) neuronal activity, and the SIRT1/FoxO1 pathway regulates the inhibitory effect of MCH on POMC expression. Remarkably, the metabolic actions of MCH are compromised in mice lacking SIRT1 specifically in POMC neurons. Of note, the actions of MCH are independent of agouti-related peptide (AgRP) neurons because inhibition of γ-aminobutyric acid receptor in the ARC did not prevent the orexigenic action of MCH, and the hypophagic effect of MCH silencing was maintained after chemogenetic stimulation of AgRP neurons. Central SIRT1 is required for MCH-induced weight gain through its actions on the sympathetic nervous system. The central MCH knockdown causes hypophagia and weight loss in diet-induced obese wild-type mice; however, these effects were abolished in mice overexpressing SIRT1 fed a high-fat diet. These data reveal the neuronal basis for the effects of MCH on food intake, body weight, and glucose metabolism and highlight the relevance of SIRT1/FoxO1 pathway in obesity.


Asunto(s)
Adiposidad/efectos de los fármacos , Proteína Forkhead Box O1/metabolismo , Intolerancia a la Glucosa/metabolismo , Hiperfagia/metabolismo , Hormonas Hipotalámicas/farmacología , Melaninas/farmacología , Neuronas/efectos de los fármacos , Hormonas Hipofisarias/farmacología , Proopiomelanocortina/metabolismo , Sirtuina 1/metabolismo , Adiposidad/fisiología , Animales , Proteína Forkhead Box O1/genética , Intolerancia a la Glucosa/genética , Hiperfagia/genética , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Técnicas de Placa-Clamp , Ratas Sprague-Dawley , Sirtuina 1/genética
19.
Neuron ; 102(3): 653-667.e6, 2019 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-30879785

RESUMEN

SIM1-expressing paraventricular hypothalamus (PVH) neurons are key regulators of energy balance. Within the PVHSIM1 population, melanocortin-4 receptor-expressing (PVHMC4R) neurons are known to regulate satiety and bodyweight, yet they account for only half of PVHSIM1 neuron-mediated regulation. Here we report that PVH prodynorphin-expressing (PVHPDYN) neurons, which notably lack MC4Rs, function independently and additively with PVHMC4R neurons to account for the totality of PVHSIM1 neuron-mediated satiety. Moreover, PVHPDYN neurons are necessary for prevention of obesity in an independent but equipotent manner to PVHMC4R neurons. While PVHPDYN and PVHMC4R neurons both project to the parabrachial complex (PB), they synaptically engage distinct efferent nodes, the pre-locus coeruleus (pLC), and central lateral parabrachial nucleus (cLPBN), respectively. PB-projecting PVHPDYN neurons, like PVHMC4R neurons, receive input from interoceptive ARCAgRP neurons, respond to caloric state, and are sufficient and necessary to control food intake. This expands the CNS satiety circuitry to include two non-overlapping PVH to hindbrain circuits.


Asunto(s)
Conducta Alimentaria/fisiología , Neuronas/citología , Obesidad/fisiopatología , Núcleo Hipotalámico Paraventricular/citología , Respuesta de Saciedad/fisiología , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/citología , Núcleo Arqueado del Hipotálamo/metabolismo , Núcleo Arqueado del Hipotálamo/fisiología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Metabolismo Energético , Encefalinas/metabolismo , Locus Coeruleus/citología , Locus Coeruleus/metabolismo , Locus Coeruleus/fisiología , Ratones , Neuronas/metabolismo , Neuronas/fisiología , Núcleos Parabraquiales/citología , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/fisiología , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Hipotalámico Paraventricular/fisiología , Precursores de Proteínas/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Proteínas Represoras/metabolismo
20.
Elife ; 82019 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-30907726

RESUMEN

When presented with a choice, organisms need to assimilate internal information with external stimuli and past experiences to rapidly and flexibly optimize decisions on a moment-to-moment basis. We hypothesized that increasing hunger intensity would curb expression of social behaviors such as mating or territorial aggression; we further hypothesized social interactions, reciprocally, would influence food consumption. We assessed competition between these motivations from both perspectives of mice within a resident-intruder paradigm. We found that as hunger state escalated, resident animal social interactions with either a female or male intruder decreased. Furthermore, intense hunger states, especially those evoked via AgRP photoactivation, fundamentally altered sequences of behavioral choice; effects dependent on food availibility. Additionally, female, but not male, intrusion attenuated resident mouse feeding. Lastly, we noted environmental context-dependent gating of food intake in intruding mice, suggesting a dynamic influence of context cues on the expression of feeding behaviors.


Asunto(s)
Toma de Decisiones , Conducta Alimentaria , Hambre , Conducta Social , Animales , Femenino , Masculino , Ratones , Motivación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...