Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2024 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-38559032

RESUMEN

Central nervous system (CNS) control of metabolism plays a pivotal role in maintaining energy homeostasis. Glucagon-like peptide-1 (GLP-1, encoded by Gcg), secreted by a distinct population of neurons located within the nucleus tractus solitarius (NTS), suppresses feeding through projections to multiple brain targets1-3. Although GLP-1 analogs are proven clinically effective in treating type 2 diabetes and obesity4, the mechanisms of GLP-1 action within the brain remain unclear. Here, we investigate the involvement of GLP-1 receptor (GLP-1R) mediated signaling in a descending circuit formed by GLP-1R neurons in the paraventricular hypothalamic nucleus (PVNGLP-1R) that project to dorsal vagal complex (DVC) neurons of the brain stem in mice. PVNGLP- 1R→DVC synapses release glutamate that is augmented by GLP-1 via a presynaptic mechanism. Chemogenetic activation of PVNGLP-1R→DVC neurons suppresses feeding. The PVNGLP-1R→DVC synaptic transmission is dynamically regulated by energy states. In a state of energy deficit, synaptic strength is weaker but is more profoundly augmented by GLP-1R signaling compared to an energy-replete state. In an obese state, the dynamic synaptic strength changes in the PVNGLP-1R→DVC descending circuit are disrupted. Blocking PVNGLP-1R→DVC synaptic release or ablation of GLP-1R in the presynaptic compartment increases food intake and causes obesity, elevated blood glucose, and impaired insulin sensitivity. These findings suggest that the state-dependent synaptic plasticity in this PVNGLP-1R→DVC descending circuit mediated by GLP-1R signaling is an essential regulator of energy homeostasis.

2.
Am J Physiol Regul Integr Comp Physiol ; 324(4): R547-R555, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-36847494

RESUMEN

Hindbrain growth hormone secretagogue receptor (GHSR) agonism increases food intake, yet the underlying neural mechanisms remain unclear. The functional effects of hindbrain GHSR antagonism by its endogenous antagonist liver-expressed antimicrobial peptide 2 (LEAP2) are also yet unexplored. To test the hypothesis that hindbrain GHSR agonism attenuates the food intake inhibitory effect of gastrointestinal (GI) satiation signals, ghrelin (at a feeding subthreshold dose) was administered to the fourth ventricle (4V) or directly to the nucleus tractus solitarius (NTS) before systemic delivery of the GI satiation signal cholecystokinin (CCK). Also examined, was whether hindbrain GHSR agonism attenuated CCK-induced NTS neural activation (c-Fos immunofluorescence). To investigate an alternate hypothesis that hindbrain GHSR agonism enhances feeding motivation and food seeking, intake stimulatory ghrelin doses were administered to the 4V and fixed ratio 5 (FR-5), progressive ratio (PR), and operant reinstatement paradigms for palatable food responding were evaluated. Also assessed were 4V LEAP2 delivery on food intake and body weight (BW) and on ghrelin-stimulated feeding. Both 4V and NTS ghrelin blocked the intake inhibitory effect of CCK and 4V ghrelin blocked CCK-induced NTS neural activation. Although 4V ghrelin increased low-demand FR-5 responding, it did not increase high-demand PR or reinstatement of operant responding. Fourth ventricle LEAP2 reduced chow intake and BW and blocked hindbrain ghrelin-stimulated feeding. Data support a role for hindbrain GHSR in bidirectional control of food intake through mechanisms that include interacting with the NTS neural processing of GI satiation signals but not food motivation and food seeking.


Asunto(s)
Hepcidinas , Receptores de Ghrelina , Receptores de Ghrelina/metabolismo , Ghrelina/farmacología , Ingestión de Alimentos , Núcleo Solitario/metabolismo , Colecistoquinina/farmacología
3.
Neuropsychopharmacology ; 48(2): 351-361, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36114285

RESUMEN

Hindbrain NTS neurons are highly attuned to internal physiological and external environmental factors that contribute to the control of food intake but the relevant neural phenotypes and pathways remain elusive. Here, we investigated the role of NTS A2 neurons and their projections in the control of feeding behaviors. In male TH Cre rats, we first confirmed selective targeting of NTS A2 neurons and showed that chemogenetic stimulation of these neurons significantly suppressed dark cycle food intake, deprivation re-feed and high fat diet intake. Despite reducing intake, activation of NTS A2 neurons had no effect on food approach, anxiety-like behaviors, locomotor activity, blood glucose levels nor did it induce nausea/malaise, thus revealing a selective role for these neurons in the consummatory aspect of food intake control. Pathway-specific mapping and manipulation of NTS A2 neurons showed that these effects were mediated by NTS A2 neurons projecting to the paraventricular nucleus of the hypothalamus (PVH) because chemogenetic activation of these projections, but not projections to bed nucleus of the stria terminalis (BNST), reduced food intake. Cell-type specific analyses demonstrated that activation of NTS A2 neurons recruited both PVH oxytocin (OT)- and corticotropin-releasing factor (CRF)-expressing neurons, and plasma analyses showed increased plasma corticosterone following NTS A2 stimulation. While we also showed that chemogenetic inhibition of NTS A2 neurons attenuated the intake inhibitory effects of CCK, the specificity of transgene expression was low. Together, these findings showed that NTS A2 neurons are sufficient to control the consummatory aspects of feeding, regardless of energy status or food palatability and identified their projections to PVH, but not BNST, in food intake control.


Asunto(s)
Hipotálamo , Núcleo Solitario , Masculino , Ratas , Animales , Núcleo Solitario/metabolismo , Hipotálamo/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Neuronas , Conducta Alimentaria
4.
Mol Metab ; 58: 101444, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35091058

RESUMEN

OBJECTIVE: The behavioral mechanisms and the neuronal pathways mediated by amylin and its long-acting analog sCT (salmon calcitonin) are not fully understood and it is unclear to what extent sCT and amylin engage overlapping or distinct neuronal subpopulations to reduce food intake. We here hypothesize that amylin and sCT recruit different neuronal population to mediate their anorectic effects. METHODS: Viral approaches were used to inhibit calcitonin gene-related peptide (CGRP) lateral parabrachial nucleus (LPBN) neurons and assess their role in amylin's and sCT's ability to decrease food intake in mice. In addition, to test the involvement of LPBN CGRP neuropeptidergic signaling in the mediation of amylin and sCT's effects, a LPBN site-specific knockdown was performed in rats. To deeper investigate whether the greater anorectic effect of sCT compared to amylin is due do the recruitment of additional neuronal pathways related to malaise multiple and distinct animal models tested whether amylin and sCT induce conditioned avoidance, nausea, emesis, and conditioned affective taste aversion. RESULTS: Our results indicate that permanent or transient inhibition of CGRP neurons in LPBN blunts sCT-, but not amylin-induced anorexia and neuronal activation. Importantly, sCT but not amylin induces behaviors indicative of malaise including conditioned affective aversion, nausea, emesis, and conditioned avoidance; the latter mediated by CGRPLPBN neurons. CONCLUSIONS: Together, the present study highlights that although amylin and sCT comparably decrease food intake, sCT is distinctive from amylin in the activation of anorectic neuronal pathways associated with malaise.


Asunto(s)
Depresores del Apetito , Polipéptido Amiloide de los Islotes Pancreáticos , Animales , Anorexia/inducido químicamente , Depresores del Apetito/efectos adversos , Depresores del Apetito/metabolismo , Calcitonina , Péptido Relacionado con Gen de Calcitonina/metabolismo , Polipéptido Amiloide de los Islotes Pancreáticos/metabolismo , Ratones , Náusea/metabolismo , Neuronas/metabolismo , Ratas , Vómitos
5.
Diabetes ; 70(11): 2545-2553, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34380697

RESUMEN

Glucagon-like peptide 1 receptor (GLP-1R) agonists decrease body weight and improve glycemic control in obesity and diabetes. Patient compliance and maximal efficacy of GLP-1 therapeutics are limited by adverse side effects, including nausea and emesis. In three different species (i.e., mice, rats, and musk shrews), we show that glucose-dependent insulinotropic polypeptide receptor (GIPR) signaling blocks emesis and attenuates illness behaviors elicited by GLP-1R activation, while maintaining reduced food intake, body weight loss, and improved glucose tolerance. The area postrema and nucleus tractus solitarius (AP/NTS) of the hindbrain are required for food intake and body weight suppression by GLP-1R ligands and processing of emetic stimuli. Using single-nuclei RNA sequencing, we identified the cellular phenotypes of AP/NTS cells expressing GIPR and GLP-1R on distinct populations of inhibitory and excitatory neurons, with the greatest expression of GIPR in γ-aminobutyric acid-ergic neurons. This work suggests that combinatorial pharmaceutical targeting of GLP-1R and GIPR will increase efficacy in treating obesity and diabetes by reducing nausea and vomiting.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/agonistas , Náusea/inducido químicamente , Náusea/tratamiento farmacológico , Receptores de la Hormona Gastrointestinal/agonistas , Animales , Peso Corporal/efectos de los fármacos , Conducta Alimentaria , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Musarañas , Vómitos
6.
Nutrients ; 13(5)2021 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-34068091

RESUMEN

The mesencephalic trigeminal nucleus (Mes5) processes oral sensory-motor information, but its role in the control of energy balance remains unexplored. Here, using fluorescent in situ hybridization, we show that the Mes5 expresses the melanocortin-4 receptor. Consistent with MC4R activation in other areas of the brain, we found that Mes5 microinjection of the MC4R agonist melanotan-II (MTII) suppresses food intake and body weight in the mouse. Furthermore, NTS POMC-projecting neurons to the Mes5 can be chemogenetically activated to drive a suppression in food intake. Taken together, these findings highlight the Mes5 as a novel target of melanocortinergic control of food intake and body weight regulation, although elucidating the endogenous role of this circuit requires future study. While we observed the sufficiency of Mes5 MC4Rs for food intake and body weight suppression, these receptors do not appear to be necessary for food intake or body weight control. Collectively, the data presented here support the functional relevance of the NTS POMC to Mes5 projection pathway as a novel circuit that can be targeted to modulate food intake and body weight.


Asunto(s)
Regulación del Apetito/fisiología , Peso Corporal/fisiología , Proopiomelanocortina/fisiología , Rombencéfalo/fisiología , Tegmento Mesencefálico/fisiología , Animales , Ingestión de Alimentos/fisiología , Femenino , Hibridación Fluorescente in Situ , Masculino , Ratones , Ratones Noqueados , Neuronas/fisiología , Rombencéfalo/anatomía & histología , Técnicas Estereotáxicas
7.
Am J Physiol Regul Integr Comp Physiol ; 319(6): R673-R683, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33026822

RESUMEN

Oxytocin (OT) is a neuropeptide whose central receptor-mediated actions include reducing food intake. One mechanism of its behavioral action is the amplification of the feeding inhibitory effects of gastrointestinal (GI) satiation signals processed by hindbrain neurons. OT treatment also reduces carbohydrate intake in humans and rodents, and correspondingly, deficits in central OT receptor (OT-R) signaling increase sucrose self-administration. This suggests that additional processes contribute to central OT effects on feeding. This study investigated the hypothesis that central OT reduces food intake by decreasing food seeking and food motivation. As central OT-Rs are expressed widely, a related focus was to assess the role of one or more OT-R-expressing nuclei in food motivation and food-seeking behavior. OT was delivered to the lateral ventricle (LV), nucleus tractus solitarius (NTS), or ventral tegmental area (VTA), and a progressive ratio (PR) schedule of operant reinforcement and an operant reinstatement paradigm were used to measure motivated feeding behavior and food-seeking behavior, respectively. OT delivered to the LV, NTS, or VTA reduced 1) motivation to work for food and 2) reinstatement of food-seeking behavior. Results provide a novel and additional interpretation for central OT-driven food intake inhibition to include the reduction of food motivation and food seeking.


Asunto(s)
Depresores del Apetito/administración & dosificación , Regulación del Apetito/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Ventrículos Laterales/efectos de los fármacos , Motivación/efectos de los fármacos , Oxitocina/administración & dosificación , Núcleo Solitario/efectos de los fármacos , Área Tegmental Ventral/efectos de los fármacos , Animales , Infusiones Intraventriculares , Ventrículos Laterales/fisiología , Masculino , Ratas Sprague-Dawley , Núcleo Solitario/fisiología , Área Tegmental Ventral/fisiología
8.
Curr Biol ; 30(22): 4510-4518.e6, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-32946754

RESUMEN

Vagal afferent neuron (VAN) signaling sends information from the gut to the brain and is fundamental in the control of feeding behavior and metabolism [1]. Recent findings reveal that VAN signaling also plays a critical role in cognitive processes, including affective motivational behaviors and hippocampus (HPC)-dependent memory [2-5]. VANs, located in nodose ganglia, express receptors for various gut-derived peptide signals; however, the function of these receptors with regard to feeding behavior, metabolism, and memory control is poorly understood. We hypothesized that VAN-mediated processes are influenced by ghrelin, a stomach-derived orexigenic hormone, via communication to its receptor (GHSR) expressed on gut-innervating VANs. To examine this hypothesis, rats received nodose ganglia injections of an adeno-associated virus (AAV) expressing short hairpin RNAs targeting GHSR (or a control AAV) for RNAi-mediated VAN-specific GHSR knockdown. Results reveal that VAN GHSR knockdown induced various feeding and metabolic disturbances, including increased meal frequency, impaired glucose tolerance, delayed gastric emptying, and increased body weight compared to controls. Additionally, VAN-specific GHSR knockdown impaired HPC-dependent contextual episodic memory and reduced HPC brain-derived neurotrophic factor expression, but did not affect anxiety-like behavior or general activity levels. A functional role for endogenous VAN GHSR signaling was further confirmed by results revealing that VAN signaling is required for the hyperphagic effects of ghrelin administered at dark onset, and that gut-restricted ghrelin-induced increases in VAN firing rate require intact VAN GHSR expression. Collective results reveal that VAN GHSR signaling is required for both normal feeding and metabolic function as well as HPC-dependent memory.


Asunto(s)
Ghrelina/metabolismo , Hipocampo/fisiología , Ganglio Nudoso/metabolismo , Receptores de Ghrelina/metabolismo , Vías Aferentes/fisiología , Animales , Peso Corporal/fisiología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Conducta Alimentaria/fisiología , Vaciamiento Gástrico/fisiología , Técnicas de Silenciamiento del Gen , Glucosa/metabolismo , Hambre/fisiología , Masculino , Memoria Episódica , Ratones , Modelos Animales , Neuronas/metabolismo , Ganglio Nudoso/citología , Ganglio Nudoso/cirugía , Ratas , Ratas Transgénicas , Receptores de Ghrelina/genética , Vagotomía
9.
Endocrinology ; 161(8)2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32516384

RESUMEN

Obesity is a chronic recurring disease whose prevalence has almost tripled over the past 40 years. In individuals with obesity, there is significant increased risk of morbidity and mortality, along with decreased quality of life. Increased obesity prevalence results, at least partly, from the increased global food supply that provides ubiquitous access to tasty, energy-dense foods. These hedonic foods and the nonfood cues that through association become reward predictive cues activate brain appetitive control circuits that drive hyperphagia and weight gain by enhancing food-seeking, motivation, and reward. Behavioral therapy (diet and lifestyle modifications) is the recommended initial treatment for obesity, yet it often fails to achieve meaningful weight loss. Furthermore, those who lose weight regain it over time through biological regulation. The need to effectively treat the pathophysiology of obesity thus centers on biologically based approaches such as bariatric surgery and more recently developed drug therapies. This review highlights neurobiological aspects relevant to obesity causation and treatment by emphasizing the common aspects of the feeding-inhibitory effects of multiple signals. We focus on glucagon like peptide-1 receptor (GLP-1R) signaling as a promising obesity treatment target by discussing the activation of intestinal- and brain-derived GLP-1 and GLP-1R expressing central nervous system circuits resulting from normal eating, bariatric surgery, and GLP-1R agonist drug therapy. Given the increased availability of energy-dense foods and frequent encounters with cues that drive hyperphagia, this review also describes how bariatric surgery and GLP-1R agonist therapies influence food reward and the motivational drive to overeat.


Asunto(s)
Péptido 1 Similar al Glucagón/uso terapéutico , Hiperfagia/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Animales , Cirugía Bariátrica , Terapia Conductista , Ingestión de Alimentos/fisiología , Péptido 1 Similar al Glucagón/fisiología , Receptor del Péptido 1 Similar al Glucagón/agonistas , Humanos , Hiperfagia/complicaciones , Hiperfagia/metabolismo , Hiperfagia/terapia , Obesidad/etiología , Obesidad/metabolismo , Obesidad/terapia , Receptores de Glucagón/metabolismo , Pérdida de Peso/fisiología
10.
Cell Rep ; 31(3): 107543, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32320650

RESUMEN

The anorectic and weight-suppressive effects of growth differentiation factor-15 (GDF15) are attracting considerable attention for treating obesity. Current experiments in rats investigate whether GDF15 induces an aversive visceral malaise-based state that mediates its acute anorectic effect and, through aversion conditioning, exerts longer-term anorexia. Visceral malaise, conditioned affective food responses (taste reactivity), gastric emptying (GE), food intake, and body weight are evaluated after acute and chronic systemic dosing of GDF15 or long-acting Fc-GDF15. Pica, a marker of visceral malaise, is present at all anorectic GDF15 doses. Moreover, malaise induced by GDF15 does not decline over time, suggesting the lack of an improved tolerance after prolonged exposure. One association between GDF15 and novel food conditions a disgust/aversive response that persists beyond GDF15 acute action. Delayed GE is not a requirement for GDF15-induced anorexia. Clinical studies are required to evaluate whether GDF15's aversive-state-based anorexia will be contraindicated as an obesity treatment.


Asunto(s)
Anorexia/inducido químicamente , Factor 15 de Diferenciación de Crecimiento/administración & dosificación , Obesidad/tratamiento farmacológico , Pérdida de Peso/efectos de los fármacos , Animales , Anorexia/metabolismo , Anorexia/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Obesidad/metabolismo , Obesidad/patología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación
11.
Transl Psychiatry ; 10(1): 90, 2020 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-32152264

RESUMEN

Previous studies identify a role for hypothalamic glia in energy balance regulation; however, a narrow hypothalamic focus provides an incomplete understanding of how glia throughout the brain respond to and regulate energy homeostasis. We examined the responses of glia in the dorsal vagal complex (DVC) to the adipokine leptin and high fat diet-induced obesity. DVC astrocytes functionally express the leptin receptor; in vivo pharmacological studies suggest that DVC astrocytes partly mediate the anorectic effects of leptin in lean but not diet-induced obese rats. Ex vivo calcium imaging indicated that these changes were related to a lower proportion of leptin-responsive cells in the DVC of obese versus lean animals. Finally, we investigated DVC microglia and astroglia responses to leptin and energy balance dysregulation in vivo: obesity decreased DVC astrogliosis, whereas the absence of leptin signaling in Zucker rats was associated with extensive astrogliosis in the DVC and decreased hypothalamic micro- and astrogliosis. These data uncover a novel functional heterogeneity of astrocytes in different brain nuclei of relevance to leptin signaling and energy balance regulation.


Asunto(s)
Hipotálamo , Leptina , Animales , Metabolismo Energético , Hipotálamo/metabolismo , Leptina/metabolismo , Neuroglía/metabolismo , Ratas , Ratas Zucker
12.
Cell Metab ; 31(2): 351-362.e5, 2020 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-31928886

RESUMEN

Growth differentiation factor 15 (GDF15) is a cytokine that reduces food intake through activation of hindbrain GFRAL-RET receptors and has become a keen target of interest for anti-obesity therapies. Elevated endogenous GDF15 is associated with energy balance disturbances, cancer progression, chemotherapy-induced anorexia, and morning sickness. We hypothesized that GDF15 causes emesis and that its anorectic effects are related to this function. Here, we examined feeding and emesis and/or emetic-like behaviors in three different mammalian laboratory species to help elucidate the role of GDF15 in these behaviors. Data show that GDF15 causes emesis in Suncus murinus (musk shrews) and induces behaviors indicative of nausea/malaise (e.g., anorexia and pica) in non-emetic species, including mice and lean or obese rats. We also present data in mice suggesting that GDF15 contributes to chemotherapy-induced malaise. Together, these results indicate that GDF15 triggers anorexia through the induction of nausea and/or by engaging emetic neurocircuitry.


Asunto(s)
Anorexia/inducido químicamente , Peso Corporal/efectos de los fármacos , Factor 15 de Diferenciación de Crecimiento , Hipoglucemiantes , Náusea/inducido químicamente , Vómitos/inducido químicamente , Animales , Femenino , Factor 15 de Diferenciación de Crecimiento/administración & dosificación , Factor 15 de Diferenciación de Crecimiento/efectos adversos , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Musarañas
13.
Brain Res ; 1724: 146441, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31513793

RESUMEN

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.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/fisiología , Leptina/metabolismo , Afecto/fisiología , Animales , Condicionamiento Clásico/fisiología , Emociones/fisiología , Metabolismo Energético/fisiología , Conducta Alimentaria/efectos de los fármacos , Alimentos , Privación de Alimentos/fisiología , Leptina/fisiología , Masculino , Obesidad , Ratas , Ratas Sprague-Dawley , Receptores de Leptina/metabolismo , Recompensa , Rombencéfalo/metabolismo , Transducción de Señal/efectos de los fármacos , Área Tegmental Ventral/metabolismo
14.
Obesity (Silver Spring) ; 27(6): 943-949, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30998842

RESUMEN

OBJECTIVE: This study investigated whether individual differences in behavioral responses to palatable food and to the satiation signal cholecystokinin (CCK) in outbred chow-maintained Sprague-Dawley rats enabled prediction of individual differences in weight gained after subsequent high-fat/high-sugar diet (HFHSD) maintenance. METHODS: Meal size, meal number, and early dark cycle intake during initial HFHSD exposure were measured, as were early dark cycle sucrose solution and chow intake, chow meal size and meal number, the intake-suppressive effects of 0.5-µg/kg CCK injection, and CCK-induced c-Fos activation in the nucleus tractus solitarius. Subsequently, rats were maintained on an HFHSD for 5 weeks, and weight gain was determined. RESULTS: Rats that took larger and less frequent meals on the first day of HFHSD exposure, whose early dark cycle intake (HFHSD and sucrose) was larger during initial HFHSD exposure, gained more weight after HFHSD maintenance. Rats with lesser sucrose intake suppression in response to CCK gained more weight after HFHSD maintenance and displayed reduced CCK-induced c-Fos activation in the nucleus tractus solitarius. CONCLUSIONS: Together, these data identify individual differences in behavioral responses to palatable food and to CCK as novel predictors of diet-induced obesity.


Asunto(s)
Colecistoquinina/efectos adversos , Alimentos/efectos adversos , Obesidad/etiología , Animales , Ingestión de Alimentos/efectos de los fármacos , Masculino , Ratas , Ratas Sprague-Dawley
15.
Cell Rep ; 26(11): 3011-3026.e5, 2019 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-30865890

RESUMEN

Chronic low-grade inflammation and increased serum levels of the cytokine IL-6 accompany obesity. For brain-produced IL-6, the mechanisms by which it controls energy balance and its role in obesity remain unclear. Here, we show that brain-produced IL-6 is decreased in obese mice and rats in a neuroanatomically and sex-specific manner. Reduced IL-6 mRNA localized to lateral parabrachial nucleus (lPBN) astrocytes, microglia, and neurons, including paraventricular hypothalamus-innervating lPBN neurons. IL-6 microinjection into lPBN reduced food intake and increased brown adipose tissue (BAT) thermogenesis in male lean and obese rats by increasing thyroid and sympathetic outflow to BAT. Parabrachial IL-6 interacted with leptin to reduce feeding. siRNA-mediated reduction of lPBN IL-6 leads to increased weight gain and adiposity, reduced BAT thermogenesis, and increased food intake. Ambient cold exposure partly normalizes the obesity-induced suppression of lPBN IL-6. These results indicate that lPBN-produced IL-6 regulates feeding and metabolism and pinpoints (patho)physiological contexts interacting with lPBN IL-6.


Asunto(s)
Peso Corporal , Ingestión de Alimentos , Metabolismo Energético , Interleucina-6/metabolismo , Núcleos Parabraquiales/metabolismo , Termogénesis , Tejido Adiposo Pardo/metabolismo , Animales , Astrocitos/metabolismo , Femenino , Interleucina-6/genética , Leptina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Núcleos Parabraquiales/fisiología , Ratas , Ratas Sprague-Dawley , Sistema Nervioso Simpático/fisiología , Hormonas Tiroideas/metabolismo
17.
Neuropsychopharmacology ; 42(12): 2387-2397, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28811669

RESUMEN

Paraventricular thalamic nucleus (PVT) neurons receive hindbrain and hypothalamic inputs, and project to forebrain sites involved in reward and motivation function. The role of PVT in energy balance and reward control is however understudied. Given that PVT neurons express glucagon-like peptide-1 receptors (GLP-1R), which are critical to feeding and body weight control, we tested the hypothesis that PVT GLP-1R signaling contributes to food intake and reward inhibition. To assess the hypothesis, behavioral tests including chow and high-fat diet intake, meal patterns, conditioned place preference for high-fat food, cue-induced reinstatement of sucrose-seeking, and motivation to work for sucrose were employed following intra-PVT delivery of either GLP-1R agonist, exendin-4 (Ex4), or GLP-1R antagonist, exendin-9-39 (Ex9). Anatomical and electrophysiological experiments were conducted to examine the neural connections and cellular mechanisms of GLP-1R signaling on PVT-to-nucleus accumbens (NAc) projecting neurons. PVT GLP-1R agonism reduced food intake, food-motivation, and food-seeking, while blocking endogenous PVT GLP-1R signaling increased meal size and food intake. PVT neurons receive GLP-1 innervation from nucleus tractus solitarius preproglucagon neurons that were activated by food intake; these GLP-1 fibers formed close appositions to putative GLP-1R-expressing PVT cells that project to the NAc. Electrophysiological recordings of PVT-to-NAc neurons revealed that GLP-1R activation reduced their excitability, mediated in part via suppression of excitatory synaptic drive. Collectively, these behavioral, electrophysiological and anatomical data illuminate a novel function for PVT GLP-1R signaling in food intake control and suggest a role for the PVT-to-NAc pathway in mediating the effects of PVT GLP-1R activation.


Asunto(s)
Ingestión de Alimentos/fisiología , Receptor del Péptido 1 Similar al Glucagón/fisiología , Núcleos Talámicos de la Línea Media/fisiología , Recompensa , Transducción de Señal/fisiología , Animales , Dieta Alta en Grasa/psicología , Dieta Alta en Grasa/tendencias , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Cultivo de Órganos , Ratas , Ratas Sprague-Dawley
18.
Endocrinology ; 158(9): 2826-2836, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28575174

RESUMEN

Central oxytocin receptor (OT-R) signaling reduces food intake and increases energy expenditure, but the central sites and mechanisms mediating these effects are unresolved. We showed previously that pharmacological activation of OT-R in hindbrain/nucleus tractus solitarius (NTS) amplifies the intake-inhibitory effects of gastrointestinal (GI) satiation signals. Unexplored were the energetic effects of hindbrain OT-R agonism and the physiological relevance of NTS OT-R signaling on food intake and energy expenditure control. Using a virally mediated OT-R knockdown (KD) strategy and a range of behavioral paradigms, this study examined the role of endogenous NTS OT-R signaling on satiation-mediated food intake inhibition and thermogenic control. Results showed that, compared with controls, NTS OT-R KD rats consumed larger meals, were less responsive to the intake-inhibitory effects of a self-ingested preload, and consumed more chow following a 24-hour fast. These data indicate that NTS OT-R signaling is necessary for normal satiation control. Whereas both control and NTS OT-R KD rats increased core temperature following high-fat diet maintenance (relative to chow maintenance), the percent increase in core temperature was greater in control compared with NTS OT-R KD rats during the light cycle. Hindbrain oxytocin agonist delivery increased core temperature in both control and NTS OT-R KD rats and the percent increase relative to vehicle treatment was not significantly different between groups. Together, data reveal a critical role for endogenous NTS OT-R signaling in mediating the intake-inhibitory effects of endogenous GI satiation signals and in diet-induced thermogenesis.


Asunto(s)
Ingestión de Alimentos/genética , Receptores de Oxitocina/fisiología , Saciedad/fisiología , Núcleo Solitario/metabolismo , Termogénesis/genética , Animales , Regulación del Apetito/genética , Dieta Alta en Grasa , Metabolismo Energético/fisiología , Masculino , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , Receptores de Oxitocina/genética , Transducción de Señal/genética
19.
J Neurosci ; 37(2): 362-370, 2017 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-28077715

RESUMEN

Cisplatin chemotherapy is commonly used to treat cancer despite severe energy balance side effects. In rats, cisplatin activates nucleus tractus solitarius (NTS) projections to the lateral parabrachial nucleus (lPBN) and calcitonin-gene related peptide (CGRP) projections from the lPBN to the central nucleus of the amygdala (CeA). We demonstrated previously that CeA glutamate receptor signaling mediates cisplatin-induced anorexia and body weight loss. Here, we used neuroanatomical tracing, immunofluorescence, and confocal imaging to demonstrate that virtually all NTS→lPBN and lPBN→CeA CGRP projections coexpress vesicular glutamate transporter 2 (VGLUT2), providing evidence that excitatory projections mediate cisplatin-induced energy balance dysregulation. To test whether lPBN→CeA projection neurons are required for cisplatin-induced anorexia and weight loss, we inhibited these neurons chemogenetically using a retrograde Cre-recombinase-expressing canine adenovirus-2 in combination with Cre-dependent inhibitory Designer Receptors Exclusive Activated by Designer Drugs (DREADDs) before cisplatin treatment. Inhibition of lPBN→CeA neurons attenuated cisplatin-induced anorexia and body weight loss significantly. Using a similar approach, we additionally demonstrated that inhibition of NTS→lPBN neurons attenuated cisplatin-induced anorexia and body weight loss significantly. Together, our data support the view that excitatory hindbrain-forebrain projections are necessary for cisplatin's untoward effects on energy intake, elucidating a key neuroanatomical circuit driving pathological anorexia and weight loss that accompanies chemotherapy treatment. SIGNIFICANCE STATEMENT: Chemotherapy treatments are commonly used to treat cancers despite accompanying anorexia and weight loss that may limit treatment adherence and reduce patient quality of life. Strikingly, we lack a neural understanding of, and effective treatments for, chemotherapy-induced anorexia and weight loss. The current data characterize the excitatory nature of neural projections activated by cisplatin in rats and reveal the necessity of specific hindbrain-forebrain projections for cisplatin-induced anorexia and weight loss. Together, these findings help to characterize the neural mechanisms mediating cisplatin-induced anorexia, advancing opportunities to develop better-tolerated chemotherapies and adjuvant therapies to prevent anorexia and concurrent nutritional deficiencies during cancer treatment.


Asunto(s)
Amígdala del Cerebelo/fisiología , Anorexia/inducido químicamente , Cisplatino/toxicidad , Núcleos Parabraquiales/fisiología , Núcleo Solitario/fisiología , Pérdida de Peso/fisiología , Amígdala del Cerebelo/efectos de los fármacos , Animales , Anorexia/fisiopatología , Antineoplásicos/toxicidad , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Masculino , Núcleos Parabraquiales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Núcleo Solitario/efectos de los fármacos , Pérdida de Peso/efectos de los fármacos
20.
Biol Psychiatry ; 81(9): 748-756, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26555354

RESUMEN

Food intake is a complex behavior that can occur or cease to occur for a multitude of reasons. Decisions about where, when, what, and how much to eat are not merely reflexive responses to food-relevant stimuli or to changes in energy status. Rather, feeding behavior is modulated by various contextual factors and by previous experiences. The data reviewed here support the perspective that neurons in multiple hippocampal subregions constitute an important neural substrate linking the external context, the internal context, and mnemonic and cognitive information to control both appetitive and ingestive behavior. Feeding behavior is heavily influenced by hippocampal-dependent mnemonic functions, including episodic meal-related memories and conditional learned associations between food-related stimuli and postingestive consequences. These mnemonic processes are undoubtedly influenced by both external and internal factors relating to food availability, location, and physiological energy status. The afferent and efferent neuroanatomical connectivity of the subregions of the hippocampus is reviewed with regard to the integration of visuospatial and olfactory sensory information (the external context) with endocrine and gastrointestinal interoceptive stimuli (the internal context). Also discussed are recent findings demonstrating that peripherally derived endocrine signals act on receptors in hippocampal neurons to reduce (leptin, glucagon-like peptide-1) or increase (ghrelin) food intake and learned food reward-driven responding, thereby highlighting endocrine and neuropeptidergic signaling in hippocampal neurons as a novel substrate of importance in the higher-order regulation of feeding behavior.


Asunto(s)
Ingestión de Alimentos , Conducta Alimentaria/fisiología , Hipocampo/fisiología , Memoria Episódica , Neuronas/fisiología , Animales , Aprendizaje por Asociación/fisiología , Ghrelina/fisiología , Péptido 1 Similar al Glucagón/fisiología , Humanos , Interocepción/fisiología , Leptina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA