Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Metab ; 36(2): 393-407.e7, 2024 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-38242133

RESUMO

Food is a powerful natural reinforcer that guides feeding decisions. The vagus nerve conveys internal sensory information from the gut to the brain about nutritional value; however, the cellular and molecular basis of macronutrient-specific reward circuits is poorly understood. Here, we monitor in vivo calcium dynamics to provide direct evidence of independent vagal sensing pathways for the detection of dietary fats and sugars. Using activity-dependent genetic capture of vagal neurons activated in response to gut infusions of nutrients, we demonstrate the existence of separate gut-brain circuits for fat and sugar sensing that are necessary and sufficient for nutrient-specific reinforcement. Even when controlling for calories, combined activation of fat and sugar circuits increases nigrostriatal dopamine release and overeating compared with fat or sugar alone. This work provides new insights into the complex sensory circuitry that mediates motivated behavior and suggests that a subconscious internal drive to consume obesogenic diets (e.g., those high in both fat and sugar) may impede conscious dieting efforts.


Assuntos
Carboidratos , Açúcares , Humanos , Açúcares/metabolismo , Encéfalo/metabolismo , Dieta , Hiperfagia/metabolismo
2.
J Immunol ; 209(11): 2114-2132, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36261171

RESUMO

MPYS/STING (stimulator of IFN genes) senses cyclic dinucleotides (CDNs), generates type I IFNs, and plays a critical role in infection, inflammation, and cancer. In this study, analyzing genotype and haplotype data from the 1000 Genomes Project, we found that the R71H-G230A-R293Q (HAQ) MPYS allele frequency increased 57-fold in East Asians compared with sub-Saharan Africans. Meanwhile, the G230A-R293Q (AQ) allele frequency decreased by 98% in East Asians compared with sub-Saharan Africans. We propose that the HAQ and AQ alleles underwent a natural selection during the out-of-Africa migration. We used mouse models of HAQ and AQ to investigate the underlying mechanism. We found that the mice carrying the AQ allele, which disappeared in East Asians, had normal CDN-type I IFN responses. Adult AQ mice, however, had less fat mass than did HAQ or wild-type mice on a chow diet. AQ epididymal adipose tissue had increased regulatory T cells and M2 macrophages with protein expression associated with enhanced fatty acid oxidation. Conditional knockout mice and adoptive cell transfer indicate a macrophage and regulatory T cell-intrinsic role of MPYS in fatty acid metabolism. Mechanistically, AQ/IFNAR1-/- mice had a similar lean phenotype as for the AQ mice. MPYS intrinsic tryptophan fluorescence revealed that the R71H change increased MPYS hydrophilicity. Lastly, we found that the second transmembrane (TM) and the TM2-TM3 linker region of MPYS interact with activated fatty acid, fatty acyl-CoA. In summary, studying the evolution of the human MPYS gene revealed an MPYS function in modulating fatty acid metabolism that may be critical during the out-of-Africa migration.


Assuntos
Ácidos Graxos , Tolerância Imunológica , Proteínas de Membrana , Adulto , Animais , Humanos , Camundongos , Ácidos Graxos/metabolismo , Homeostase , Proteínas de Membrana/metabolismo , Camundongos Knockout , Interferon Tipo I
4.
Peptides ; 140: 170534, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33757831

RESUMO

Cocaine- and amphetamine-related transcript (CART) is a neuropeptide first discovered in the striatum of the rat brain. Later, the genetic sequence and function of CART peptide (CARTp) was found to be conserved among multiple mammalian species. Over the 25 years, since its discovery, CART mRNA (Cartpt) expression has been reported widely throughout the central and peripheral nervous systems underscoring its role in diverse physiological functions. Here, we review the localization and function of CARTp as it relates to energy homeostasis. We summarize the expression changes of central and peripheral Cartpt in response to metabolic states and make use of available large data sets to gain additional insights into the anatomy of the Cartpt expressing vagal neurons and their expression patterns in the gut. Furthermore, we provide an overview of the role of CARTp as an anorexigenic signal and its effect on energy expenditure and body weight control with insights from both pharmacological and transgenic animal studies. Subsequently, we discuss the role of CARTp in the pathophysiology of obesity and review important new developments towards identifying a candidate receptor for CARTp signalling. Altogether, the field of CARTp research has made rapid and substantial progress recently, and we review the case for considering CARTp as a potential therapeutic target for stemming the obesity epidemic.


Assuntos
Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Peptídeos/metabolismo , Nervo Vago/metabolismo , Animais , Metabolismo Energético , Homeostase , Humanos
5.
Nat Metab ; 3(2): 258-273, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33589843

RESUMO

The anorexigenic peptide glucagon-like peptide-1 (GLP-1) is secreted from gut enteroendocrine cells and brain preproglucagon (PPG) neurons, which, respectively, define the peripheral and central GLP-1 systems. PPG neurons in the nucleus tractus solitarii (NTS) are widely assumed to link the peripheral and central GLP-1 systems in a unified gut-brain satiation circuit. However, direct evidence for this hypothesis is lacking, and the necessary circuitry remains to be demonstrated. Here we show that PPGNTS neurons encode satiation in mice, consistent with vagal signalling of gastrointestinal distension. However, PPGNTS neurons predominantly receive vagal input from oxytocin-receptor-expressing vagal neurons, rather than those expressing GLP-1 receptors. PPGNTS neurons are not necessary for eating suppression by GLP-1 receptor agonists, and concurrent PPGNTS neuron activation suppresses eating more potently than semaglutide alone. We conclude that central and peripheral GLP-1 systems suppress eating via independent gut-brain circuits, providing a rationale for pharmacological activation of PPGNTS neurons in combination with GLP-1 receptor agonists as an obesity treatment strategy.


Assuntos
Sistema Nervoso Central/fisiologia , Peptídeo 1 Semelhante ao Glucagon/fisiologia , Sistema Nervoso Periférico/fisiologia , Resposta de Saciedade/fisiologia , Animais , Ingestão de Alimentos , Feminino , Trato Gastrointestinal/inervação , Trato Gastrointestinal/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Peptídeos Semelhantes ao Glucagon/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Proglucagon/metabolismo , Receptores de Ocitocina/metabolismo , Nervo Vago/fisiologia
6.
Acta Physiol (Oxf) ; 231(3): e13530, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32603548

RESUMO

AIM: The tools that have been used to assess the function of the vagus nerve lack specificity. This could explain discrepancies about the role of vagal gut-brain signalling in long-term control of energy balance. Here we use a validated approach to selectively ablate sensory vagal neurones that innervate the gut to determine the role of vagal gut-brain signalling in the control of food intake, energy expenditure and glucose homoeostasis in response to different diets. METHODS: Rat nodose ganglia were injected bilaterally with either the neurotoxin saporin conjugated to the gastrointestinal hormone cholecystokinin (CCK), or unconjugated saporin as a control. Food intake, body weight, glucose tolerance and energy expenditure were measured in both groups in response to chow or high-fat high-sugar (HFHS) diet. Willingness to work for fat or sugar was assessed by progressive ratio for orally administered solutions, while post-ingestive feedback was tested by measuring food intake after an isocaloric lipid or sucrose pre-load. RESULTS: Vagal deafferentation of the gut increases meal number in lean chow-fed rats. Switching to a HFHS diet exacerbates overeating and body weight gain. The breakpoint for sugar or fat solution did not differ between groups, suggesting that increased palatability may not drive HFHS-induced hyperphagia. Instead, decreased satiation in response to intra-gastric infusion of fat, but not sugar, promotes hyperphagia in CCK-Saporin-treated rats fed with HFHS diet. CONCLUSIONS: We conclude that intact sensory vagal neurones prevent hyperphagia and exacerbation of weight gain in response to a HFHS diet by promoting lipid-mediated satiation.


Assuntos
Hiperfagia , Açúcares , Animais , Encéfalo , Dieta Hiperlipídica/efeitos adversos , Ratos , Nervo Vago , Aumento de Peso
7.
Curr Biol ; 30(22): 4510-4518.e6, 2020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-32946754

RESUMO

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.


Assuntos
Grelina/metabolismo , Hipocampo/fisiologia , Gânglio Nodoso/metabolismo , Receptores de Grelina/metabolismo , Vias Aferentes/fisiologia , Animais , Peso Corporal/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Comportamento Alimentar/fisiologia , Esvaziamento Gástrico/fisiologia , Técnicas de Silenciamento de Genes , Glucose/metabolismo , Fome/fisiologia , Masculino , Memória Episódica , Camundongos , Modelos Animais , Neurônios/metabolismo , Gânglio Nodoso/citologia , Gânglio Nodoso/cirurgia , Ratos , Ratos Transgênicos , Receptores de Grelina/genética , Vagotomia
8.
Cell Rep ; 30(6): 2028-2039.e4, 2020 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-32049029

RESUMO

The vagus nerve conveys gastrointestinal cues to the brain to control eating behavior. In obesity, vagally mediated gut-brain signaling is disrupted. Here, we show that the cocaine- and amphetamine-regulated transcript (CART) is a neuropeptide synthesized proportional to the food consumed in vagal afferent neurons (VANs) of chow-fed rats. CART injection into the nucleus tractus solitarii (NTS), the site of vagal afferent central termination, reduces food intake. Conversely, blocking endogenous CART action in the NTS increases food intake in chow-fed rats, and this requires intact VANs. Viral-mediated Cartpt knockdown in VANs increases weight gain and daily food intake via larger meals and faster ingestion rate. In obese rats fed a high-fat, high-sugar diet, meal-induced CART synthesis in VANs is blunted and CART antibody fails to increase food intake. However, CART injection into the NTS retains its anorexigenic effect in obese rats. Restoring disrupted VAN CART signaling in obesity could be a promising therapeutic approach.


Assuntos
Hiperfagia/genética , Proteínas do Tecido Nervoso/metabolismo , Nervo Vago/efeitos dos fármacos , Aumento de Peso/genética , Animais , Humanos , Masculino , Ratos
9.
Brain Behav Immun ; 81: 444-454, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31271871

RESUMO

The gastrointestinal (GI) tract harbors commensal microorganisms as well as invasive bacteria, toxins and other pathogens and, therefore, plays a pivotal barrier and immunological role against pathogenic agents. The vagus nerve is an important regulator of the GI tract-associated immune system, having profound effects on inflammatory responses. Among GI tract organs, the liver is a key site of immune surveillance, as it has a large population of resident macrophages and receives the blood drained from the guts through the hepatic portal circulation. Although it is widely accepted that the hepatic tissue is a major target for vagus nerve fibers, the role of this neural circuit in liver immune functions is still poorly understood. Herein we used in vivo imaging techniques, including confocal microscopy and scintigraphy, to show that vagus nerve stimulation increases the phagocytosis activity by resident macrophages in the liver, even on the absence of an immune challenge. The activation of this neural circuit in a non-lethal model of sepsis optimized the removal of bacteria in the liver and resulted in the production of anti-inflammatory and pro-regenerative cytokines. Our findings provide new insights into the neural regulation of the immune system in the liver.


Assuntos
Fígado/imunologia , Fagocitose/fisiologia , Nervo Vago/fisiologia , Animais , Citocinas , Feminino , Trato Gastrointestinal , Fígado/patologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fagócitos/metabolismo , Sepse/imunologia , Nervo Vago/patologia , Estimulação do Nervo Vago/métodos
10.
Am J Physiol Endocrinol Metab ; 316(4): E568-E577, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30753113

RESUMO

Deletion of the leptin receptor from vagal afferent neurons (VAN) using a conditional deletion (Nav1.8/LepRfl/fl) results in an obese phenotype with increased food intake and lack of exogenous cholecystokinin (CCK)-induced satiation in male mice. Female mice are partially protected from weight gain and increased food intake in response to ingestion of high-fat (HF) diets. However, whether the lack of leptin signaling in VAN leads to an obese phenotype or disruption of hypothalamic-pituitary-gonadal axis function in female mice is unclear. Here, we tested the hypothesis that leptin signaling in VAN is essential to maintain estrogen signaling and control of food intake, energy expenditure, and adiposity in female mice. Female Nav1.8/LepRfl/fl mice gained more weight, had increased gonadal fat mass, increased meal number in the dark phase, and increased total food intake compared with wild-type controls. Resting energy expenditure was unaffected. The decrease in food intake produced by intraperitoneal injection of CCK (3 µg/kg body wt) was attenuated in female Nav1.8/LepRfl/fl mice compared with wild-type controls. Intraperitoneal injection of ghrelin (100 µg/kg body wt) increased food intake in Nav1.8/LepRfl/fl mice but not in wild-type controls. Ovarian steroidogenesis was suppressed, resulting in decreased plasma estradiol, which was accompanied by decreased expression of estrogen receptor-1 (Esr1) in VAN but not in the hypothalamic arcuate nucleus. These data suggest that the absence of leptin signaling in VAN is accompanied by disruption of estrogen signaling in female mice, leading to an obese phenotype possibly via altered control of feeding behavior.


Assuntos
Ingestão de Alimentos/genética , Comportamento Alimentar/fisiologia , Neurônios Aferentes/metabolismo , Obesidade/genética , Receptores para Leptina/genética , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Peso Corporal/genética , Colecistocinina/farmacologia , Dieta Hiperlipídica , Ingestão de Alimentos/efeitos dos fármacos , Metabolismo Energético , Estradiol/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Comportamento Alimentar/efeitos dos fármacos , Feminino , Grelina/farmacologia , Camundongos , Obesidade/metabolismo , Saciação , Nervo Vago/citologia , Aumento de Peso/genética
11.
Curr Opin Pharmacol ; 31: 38-43, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27591963

RESUMO

Vagal afferent neurons (VANs) play an important role in the control of food intake by signaling nutrient type and quantity to the brain. Recent findings are broadening our view of how VANs impact not only food intake but also energy homeostasis. This review focuses exclusively on studies of the vagus nerve from the past 2 years that highlight major new advancements in the field. We firstly discuss evidence that VANs can directly sense nutrients, and we consider new insights into mechanisms affecting sensing of gastric distension and signaling by gastrointestinal hormones ghrelin and GLP1. We discuss evidence that disrupting vagal afferent signaling increases long-term control of food intake and body weight management, and the importance of this gut-brain pathway in mediating beneficial effects of bariatric surgery. We conclude by highlighting novel roles for vagal afferent neurons in circadian rhythm, thermogenesis, and reward that may provide insight into mechanisms by which VAN nutrient sensing controls long-term control of energy homeostasis.


Assuntos
Metabolismo Energético/fisiologia , Neurônios Aferentes/metabolismo , Nervo Vago/fisiologia , Animais , Peso Corporal/fisiologia , Ritmo Circadiano/fisiologia , Ingestão de Alimentos/fisiologia , Grelina/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Homeostase/fisiologia , Humanos , Recompensa , Transdução de Sinais/fisiologia
12.
J Physiol ; 594(20): 5791-5815, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-26959077

RESUMO

This review highlights evidence for a role of the vagus nerve in the development of obesity and how targeting the vagus nerve with neuromodulation or pharmacology can be used as a therapeutic treatment of obesity. The vagus nerve innervating the gut plays an important role in controlling metabolism. It communicates peripheral information about the volume and type of nutrients between the gut and the brain. Depending on the nutritional status, vagal afferent neurons express two different neurochemical phenotypes that can inhibit or stimulate food intake. Chronic ingestion of calorie-rich diets reduces sensitivity of vagal afferent neurons to peripheral signals and their constitutive expression of orexigenic receptors and neuropeptides. This disruption of vagal afferent signalling is sufficient to drive hyperphagia and obesity. Furthermore neuromodulation of the vagus nerve can be used in the treatment of obesity. Although the mechanisms are poorly understood, vagal nerve stimulation prevents weight gain in response to a high-fat diet. In small clinical studies, in patients with depression or epilepsy, vagal nerve stimulation has been demonstrated to promote weight loss. Vagal blockade, which inhibits the vagus nerve, results in significant weight loss. Vagal blockade is proposed to inhibit aberrant orexigenic signals arising in obesity as a putative mechanism of vagal blockade-induced weight loss. Approaches and molecular targets to develop future pharmacotherapy targeted to the vagus nerve for the treatment of obesity are proposed. In conclusion there is strong evidence that the vagus nerve is involved in the development of obesity and it is proving to be an attractive target for the treatment of obesity.


Assuntos
Obesidade/fisiopatologia , Nervo Vago/fisiologia , Animais , Peso Corporal/fisiologia , Dieta Hiperlipídica , Humanos , Hiperfagia/fisiopatologia , Neurônios Aferentes/fisiologia
13.
Physiol Behav ; 139: 188-94, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25446227

RESUMO

Bacterially derived factors are implicated in the causation and persistence of obesity. Ingestion of a high fat diet in rodents and obesity in human subjects is associated with chronic elevation of low plasma levels of lipopolysaccharide (LPS), a breakdown product of Gram-negative bacteria. The terminals of vagal afferent neurons are positioned within the gut mucosa to convey information from the gut to the brain to regulate food intake and are responsive to LPS. We hypothesized that chronic elevation of LPS could alter vagal afferent signaling. We surgically implanted osmotic mini-pumps that delivered a constant, low-dose of LPS into the intraperitoneal cavity of rats (12.5 µg/kg/hr for 6 weeks). LPS-treated rats developed hyperphagia and showed marked changes in vagal afferent neuron function. Chronic LPS treatment reduced vagal afferent leptin signaling, characterized by a decrease in leptin-induced STAT3 phosphorylation. In addition, LPS treatment decreased cholecystokinin-induced satiety. There was no alteration in leptin signaling in the hypothalamus. These findings offer a mechanism by which a change in gut microflora can promote hyperphagia, possibly leading to obesity.


Assuntos
Ingestão de Alimentos/fisiologia , Leptina/metabolismo , Lipopolissacarídeos/toxicidade , Neurônios Aferentes/fisiologia , Gânglio Nodoso/fisiopatologia , Animais , Western Blotting , Hiperfagia/fisiopatologia , Imuno-Histoquímica , Masculino , Peroxidase/metabolismo , Ratos Wistar , Saciação/fisiologia , Sincalida/administração & dosagem , Sincalida/metabolismo , Aumento de Peso/fisiologia
14.
Physiol Behav ; 103(2): 181-7, 2011 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-21277881

RESUMO

Vagal afferent neurons (VAN) express the cholecystokinin (CCK) type 1 receptor (CCK1R) and, as predicted by the role of CCK in inducing satiation, CCK1R⁻/⁻ mice ingest larger and longer meals. However, after a short fast, CCK1R⁻/⁻ mice ingesting high fat (HF) diets initiate feeding earlier than wild-type mice. We hypothesized that the increased drive to eat in CCK1R⁻/⁻ mice eating HF diet is mediated by ghrelin, a gut peptide that stimulates food intake. The decrease in time to first meal, and the increase in meal size and duration in CCK1R⁻/⁻ compared to wild-type mice ingesting high fat (HF) diet were reversed by administration of GHSR1a antagonist D-(Lys3)-GHRP-6 (p<0.05). Administration of the GHSR1a antagonist significantly increased expression of the neuropeptide cocaine and amphetamine-regulated transcript (CART) in VAN of HF-fed CCK1R⁻/⁻ but not wild-type mice. Administration of the GHSR1a antagonist decreased neuronal activity measured by immunoreactivity for fos protein in the nucleus of the solitary tract (NTS) and the arcuate nucleus of both HF-fed wild-type and CCK1R⁻/⁻ mice. The data show that hyperphagia in CCK1R⁻/⁻ mice ingesting HF diet is reversed by blockade of the ghrelin receptor, suggesting that in the absence of the CCK1R, there is an increased ghrelin-dependent drive to feed. The site of action of ghrelin receptors is unclear, but may involve an increase in expression of CART peptide in VAN in HF-fed CCK1R⁻/⁻ mice.


Assuntos
Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Comportamento Alimentar/fisiologia , Oligopeptídeos/farmacologia , Receptor de Colecistocinina A/fisiologia , Receptores de Grelina/antagonistas & inibidores , Animais , Núcleo Arqueado do Hipotálamo/fisiologia , Comportamento Alimentar/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptor de Colecistocinina A/genética , Receptores de Grelina/fisiologia , Núcleo Solitário/fisiologia , Nervo Vago/metabolismo
15.
J Neurosci ; 28(45): 11583-92, 2008 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-18987194

RESUMO

The intestinal hormones CCK and PYY3-36 inhibit gastric emptying and food intake via vagal afferent neurons. Here we report that CCK regulates the expression of Y2R, at which PYY3-36 acts. In nodose ganglia from rats fasted up to 48 h, there was a fivefold decrease of Y2R mRNA compared with rats fed ad libitum; Y2R mRNA in fasted rats was increased by administration of CCK, and by refeeding through a mechanism sensitive to the CCK1R antagonist lorglumide. Antibodies to Y2R revealed expression in both neurons and satellite cells; most of the former (89 +/- 4%) also expressed CCK1R. With fasting there was loss of Y2R immunoreactivity in CCK1R-expressing neurons many of which projected to the stomach, but not in satellite cells or neurons projecting to the ileum or proximal colon. Expression of a Y2R promoter-luciferase reporter (Y2R-luc) in cultured vagal afferent neurons was increased in response to CCK by 12.3 +/- 0.1-fold and by phorbol ester (16.2 +/- 0.4-fold); the response to both was abolished by the protein kinase C inhibitor Ro-32,0432. PYY3-36 stimulated CREB phosphorylation in rat nodose neurons after priming with CCK; in wild-type mice PYY3-36 increased Fos labeling in brainstem neurons but in mice null for CCK1R this response was abolished. Thus Y2R is expressed by functionally distinct subsets of nodose ganglion neurons projecting to the stomach and ileum/colon; in the former expression is dependent on stimulation by CCK, and there is evidence that PYY3-36 effects on vagal afferent neurons are CCK dependent.


Assuntos
Colagogos e Coleréticos/farmacologia , Colecistocinina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Neurônios Aferentes/efeitos dos fármacos , Receptores de Neuropeptídeo Y/metabolismo , Estômago/inervação , Nervo Vago/citologia , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Células Cultivadas , Jejum/fisiologia , Antagonistas de Hormônios/farmacologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Gânglio Nodoso/citologia , Proteínas Oncogênicas v-fos/metabolismo , Proglumida/análogos & derivados , Proglumida/farmacologia , RNA Mensageiro/metabolismo , Ratos , Receptor de Colecistocinina A/deficiência , Receptores de Neuropeptídeo Y/genética , Resposta de Saciedade/efeitos dos fármacos , Resposta de Saciedade/fisiologia
16.
J Neurosci ; 27(11): 2876-82, 2007 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-17360909

RESUMO

The neuropeptide transmitter cocaine- and amphetamine-regulated transcript (CART) inhibits food intake and is expressed by both vagal afferent and hypothalamic neurons. Here we report that cholecystokinin (CCK) regulates CART expression in rat vagal afferent neurons. Thus, CART was virtually undetectable after energy restriction for 24 h, but administration of CCK to fasted rats increased CART immunoreactivity, and refeeding of fasted animals promptly increased CART by a mechanism sensitive to a CCK-1 receptor antagonist. In vagal afferent neurons incubated in serum-free medium, CART was virtually undetectable, whereas the orexigenic peptide melanin-concentrating hormone (MCH) was readily detected. The addition of CCK rapidly induced CART expression and downregulated MCH. Using a CART promoter-luciferase reporter vector transfected into cultured vagal afferent neurons, we showed that CCK stimulation of CART transcription was mediated by activation of protein kinase C and cAMP response element-binding protein (CREB). The action of CCK on CART expression was inhibited by the orexigenic peptide ghrelin, through a mechanism that involved exclusion of phosphorylated CREB from the nucleus. Thus, CCK reciprocally regulates expression of CART and MCH within the same vagal afferent neuron; ghrelin inhibits the effect of CCK at least in part through control of the nuclear localization of phosphoCREB, revealing previously unsuspected modulation of gut-brain signals implicated in control of food intake.


Assuntos
Colecistocinina/farmacologia , Regulação para Baixo/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Neurônios Aferentes/metabolismo , Hormônios Peptídicos/farmacologia , Regulação para Cima/fisiologia , Nervo Vago/metabolismo , Animais , Células Cultivadas , Colecistocinina/metabolismo , Regulação para Baixo/efeitos dos fármacos , Grelina , Masculino , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios Aferentes/efeitos dos fármacos , Gânglio Nodoso/efeitos dos fármacos , Gânglio Nodoso/metabolismo , Hormônios Peptídicos/metabolismo , Ratos , Ratos Wistar , Regulação para Cima/efeitos dos fármacos , Nervo Vago/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA