Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Endocrinol (Lausanne) ; 14: 1158287, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37234803

RESUMO

Introduction: Oleoylethanolamide (OEA), an endogenous N-acylethanolamine acting as a gut-to-brain signal to control food intake and metabolism, has been attracting attention as a target for novel therapies against obesity and eating disorders. Numerous observations suggested that the OEA effects might be peripherally mediated, although they involve central pathways including noradrenergic, histaminergic and oxytocinergic systems of the brainstem and the hypothalamus. Whether these pathways are activated directly by OEA or whether they are downstream of afferent nerves is still highly debated. Some early studies suggested vagal afferent fibers as the main route, but our previous observations have contradicted this idea and led us to consider the blood circulation as an alternative way for OEA's central actions. Methods: To test this hypothesis, we first investigated the impact of subdiaphragmatic vagal deafferentation (SDA) on the OEA-induced activation of selected brain nuclei. Then, we analyzed the pattern of OEA distribution in plasma and brain at different time points after intraperitoneal administration in addition to measuring food intake. Results: Confirming and extending our previous findings that subdiaphragmatic vagal afferents are not necessary for the eating-inhibitory effect of exogenous OEA, our present results demonstrate that vagal sensory fibers are also not necessary for the neurochemical effects of OEA. Rather, within a few minutes after intraperitoneal administration, we found an increased concentration of intact OEA in different brain areas, associated with the inhibition of food intake. Conclusion: Our results support that systemic OEA rapidly reaches the brain via the circulation and inhibits eating by acting directly on selected brain nuclei.


Assuntos
Encéfalo , Ingestão de Alimentos , Ingestão de Alimentos/fisiologia , Encéfalo/metabolismo , Endocanabinoides/farmacologia , Endocanabinoides/metabolismo , Ácidos Oleicos/farmacologia , Ácidos Oleicos/metabolismo
2.
Basic Clin Pharmacol Toxicol ; 127(3): 163-177, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32363722

RESUMO

The prevalence of obesity and associated comorbidities such as type 2 diabetes and cardiovascular disease is increasing globally. Body-weight loss reduces the risk of morbidity and mortality in obese individuals, and thus, pharmacotherapies that induce weight loss can be of great value in improving the health and well-being of people living with obesity. Treatment with amylin and calcitonin receptor agonists reduces food intake and induces weight loss in several animal models, and a number of companies have started clinical testing for peptide analogues in the treatment of obesity and/or type 2 diabetes. Studies predominantly performed in rodent models show that amylin and the dual amylin/calcitonin receptor agonist salmon calcitonin achieve their metabolic effects by engaging areas in the brain associated with regulating homeostatic energy balance. In particular, signalling via neuronal circuits in the caudal hindbrain and the hypothalamus is implicated in mediating effects on food intake and energy expenditure. We review the current literature investigating the interaction of amylin/calcitonin receptor agonists with neurocircuits that induce the observed metabolic effects. Moreover, the status of drug development of amylin and calcitonin receptor agonists for the treatment of metabolic diseases is summarized.


Assuntos
Agonistas dos Receptores da Amilina/farmacologia , Agonistas dos Receptores da Amilina/uso terapêutico , Doenças Metabólicas/tratamento farmacológico , Receptores da Calcitonina/agonistas , Receptores da Calcitonina/uso terapêutico , Animais , Metabolismo Energético , Humanos , Hipotálamo , Polipeptídeo Amiloide das Ilhotas Pancreáticas/fisiologia , Leptina , Camundongos , Ratos , Rombencéfalo
3.
Neuropharmacology ; 167: 107987, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32035146

RESUMO

The amylin receptor (AMY) and calcitonin receptor (CTR) agonists induce acute suppression of food intake in rodents by binding to receptors in the area postrema (AP) and potentially by targeting arcuate (ARC) neurons directly. Salmon calcitonin (sCT) induces more potent, longer lasting anorectic effects compared to amylin. We thus aimed to investigate whether AMY/CTR agonists target key neuronal populations in the ARC, and whether differing brain distribution patterns could mediate the observed differences in efficacy with sCT and amylin treatment. Brains were examined by whole brain 3D imaging and confocal microscopy following subcutaneous administration of fluorescently labelled peptides to mice. We found that sCT, but not amylin, internalizes into a subset of ARC NPY neurons, along with an unknown subset of ARC, AP and dorsal vagal motor nucleus cells. ARC POMC neurons were not targeted. Furthermore, amylin and sCT displayed similar distribution patterns binding to receptors in the AP, the organum vasculosum of the lamina terminalis (OVLT) and the ARC. Amylin distributed within the median eminence with only specs of sCT being present in this region, however amylin was only detectable 10 minutes after injection while sCT displayed a residence time of up to 2 hours post injection. We conclude that AMY/CTR agonists bind to receptors in a subset of ARC NPY neurons and in circumventricular organs. Furthermore, the more sustained and greater anorectic efficacy of sCT compared to rat amylin is not attributable to differences in brain distribution patterns but may more likely be explained by greater potency at both the CTR and AMY.


Assuntos
Núcleo Arqueado do Hipotálamo/metabolismo , Calcitonina/metabolismo , Hormônios e Agentes Reguladores de Cálcio/metabolismo , Neurônios/metabolismo , Neuropeptídeo Y/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Calcitonina/administração & dosagem , Hormônios e Agentes Reguladores de Cálcio/administração & dosagem , Linhagem Celular , Cricetinae , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Ligação Proteica/fisiologia , Ratos
4.
Mol Metab ; 8: 203-210, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29203236

RESUMO

BACKGROUND: Amylin is a pancreatic ß-cell hormone that produces effects in several different organ systems. One of its best-characterized effects is the reduction in eating and body weight seen in preclinical and clinical studies. Amylin activates specific receptors, a portion of which it shares with calcitonin gene-related peptide (CGRP). Amylin's role in the control of energy metabolism relates to its satiating effect, but recent data indicate that amylin may also affect hedonic aspects in the control of eating, including a reduction of the rewarding value of food. Recently, several amylin-based peptides have been characterized. Pramlintide (Symlin®) is currently the only one being used clinically to treat type 1 and type 2 diabetes. However other amylin analogs with improved pharmacokinetic properties are being considered as anti-obesity treatment strategies. Several other studies in obesity have shown that amylin agonists could also be useful for weight loss, especially in combination with other agents. SCOPE OF REVIEW: This review will briefly summarize amylin physiology and pharmacology and then focus on amylin's role in food reward and the effects of amylin analogs in pre-clinical testing for anti-obesity drugs. CONCLUSION: We propose here that the effects of amylin may be homeostatic and hedonic in nature.


Assuntos
Fármacos Antiobesidade/uso terapêutico , Hiperfagia/tratamento farmacológico , Polipeptídeo Amiloide das Ilhotas Pancreáticas/química , Obesidade/tratamento farmacológico , Fragmentos de Peptídeos/uso terapêutico , Recompensa , Animais , Fármacos Antiobesidade/farmacologia , Tronco Encefálico/efeitos dos fármacos , Humanos , Hiperfagia/metabolismo , Hiperfagia/fisiopatologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Obesidade/metabolismo , Obesidade/fisiopatologia , Fragmentos de Peptídeos/farmacologia
5.
Handb Exp Pharmacol ; (209): 231-50, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22249817

RESUMO

Amylin's best investigated function is to reduce eating via a meal size effect by promoting meal-ending satiation. This effect seems to depend on an activation of specific area postrema neurons. Brain areas that convey the neural signal to the forebrain include the nucleus of the solitary tract and the lateral parabrachial nucleus. Acute application of amylin modulates the activity of hypothalamic areas involved in the control of eating, namely, the lateral hypothalamic area and possibly the ventromedial hypothalamic nucleus. Amylin also interacts with other satiating signals, such as cholecystokinin, presumably in the brainstem. Interestingly, amylin also exhibits characteristics of adiposity signals; plasma levels of amylin are higher in obese individuals, chronic infusion of amylin into the brain reduces body weight gain and adiposity, and infusion of amylin antagonists increases adiposity. Furthermore, amylin maintains energy expenditure at higher levels than would be expected considering its body weight-lowering effect. However, much less is known (e.g., site of action, signaling pathways, differential activation of brain sites, and, most importantly, physiological relevance) with respect to its role as adiposity signal and regulator of energy expenditure than about its satiating action. Notwithstanding, and perhaps because amylin resistance does not seem to be a general and prohibitive concomitant of obesity, animal data and recent clinical data in humans indicate that amylin is a very promising candidate for the treatment of obesity. Amylin seems to be particularly effective when combined with other hormones such as leptin.


Assuntos
Adiposidade , Encéfalo/metabolismo , Ingestão de Alimentos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Transdução de Sinais , Animais , Fármacos Antiobesidade/farmacologia , Regulação do Apetite , Metabolismo Energético , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/farmacologia , Leptina/metabolismo , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Obesidade/fisiopatologia , Resposta de Saciedade
6.
Am J Physiol Regul Integr Comp Physiol ; 302(3): R340-51, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22129618

RESUMO

Peripheral amylin inhibits eating via the area postrema (AP). Because amylin activates the extracellular-signal regulated kinase 1/2 (ERK) pathway in some tissues, and because ERK1/2 phosphorylation (pERK) leads to acute neuronal responses, we postulated that it may be involved in amylin's eating inhibitory effect. Amylin-induced ERK phosphorylation (pERK) was investigated by immunohistochemistry in brain sections containing the AP. pERK-positive AP neurons were double-stained for the calcitonin 1a/b receptor, which is part of the functional amylin-receptor. AP sections were also phenotyped using dopamine-ß-hydroxylase (DBH) as a marker of noradrenergic neurons. The effect of fourth ventricular administration of the ERK cascade blocker U0126 on amylin's eating inhibitory action was tested in feeding trials. The number of pERK-positive neurons in the AP was highest ∼10-15 min after amylin treatment; the effect appeared to be dose-dependent (5-20 µg/kg amylin). A portion of pERK-positive neurons in the AP carried the amylin-receptor and 22% of the pERK-positive neurons were noradrenergic. Pretreatment of rats with U0126 decreased the number of pERK-positive neurons in the AP after amylin injection. U0126 also attenuated the ability of amylin to reduce eating, at least when the animals had been fasted 24 h prior to the feeding trial. Overall, our results suggest that amylin directly stimulates pERK in AP neurons in a time- and dose-dependent manner. Part of the AP neurons displaying pERK were noradrenergic. At least under fasting conditions, pERK was shown to be a necessary part in the signaling cascade mediating amylin's anorectic effect.


Assuntos
Anorexia/fisiopatologia , Regulação do Apetite/efeitos dos fármacos , Regulação do Apetite/fisiologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/farmacologia , Sistema de Sinalização das MAP Quinases/fisiologia , Animais , Área Postrema/efeitos dos fármacos , Área Postrema/patologia , Área Postrema/fisiopatologia , Butadienos/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Quarto Ventrículo/efeitos dos fármacos , Quarto Ventrículo/patologia , Quarto Ventrículo/fisiopatologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Nitrilas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Polipeptídeo Amiloide de Ilhotas Pancreáticas/efeitos dos fármacos , Receptores de Polipeptídeo Amiloide de Ilhotas Pancreáticas/fisiologia , Fatores de Tempo
7.
Physiol Behav ; 103(1): 25-30, 2011 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-21035478

RESUMO

Amylin is secreted by pancreatic beta-cells and seems to function as a physiological signal of satiation and possibly also as an adiposity signal. Amylin's satiating effect is mediated via a direct action at area postrema (AP) neurons. The central pathways mediating amylin's effects rely on connections from the AP to the nucleus of the solitary tract and lateral parabrachial nucleus. Amylin was shown to interact, probably at the brainstem, with other satiating signals, namely cholecystokinin, glucagon-like peptide 1 and peptide YY, and other adiposity signals, namely leptin and insulin. The interaction with leptin, which is thought to involve the hypothalamus, may have important implications for the development of new and improved hormonal anti-obesity treatments. Steve Woods has contributed to the recent literature on amylin's eating inhibitory effect by some frequently cited publications. Steve's work concentrated more on the central administration of amylin and on amylin's potential role as an adiposity signal. His work will be reviewed here and discussed in the context of other important findings on amylin's role in the control of energy homeostasis.


Assuntos
Depressores do Apetite/farmacologia , Pesquisa Biomédica/história , Comportamento Alimentar/efeitos dos fármacos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/farmacologia , Adiposidade/efeitos dos fármacos , Animais , Depressores do Apetite/história , Peso Corporal/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , História do Século XX , História do Século XXI , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas/história , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Núcleo Solitário/citologia
8.
Brain Res ; 1334: 31-44, 2010 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-20382134

RESUMO

The ability of the pancreatic hormone amylin to inhibit food intake relies on a direct activation of the area postrema (AP). This activation is synaptically transmitted to the nucleus of the solitary tract (NTS), the lateral parabrachial nucleus (LPB), the central amygdaloid nucleus (Ce) and the lateral bed nucleus of stria terminalis (BSTL). Interestingly, neurons of the rostro-dorsal lateral hypothalamic area (dLHA), which are activated during fasting, are inhibited by peripheral amylin, although they lack amylin receptors. Using the retrograde tracer cholera toxin-B (Ctb) we analyzed whether the dLHA receives neuronal projections from amylin-activated brain areas. The anterograde tracer biotinylated dextran-amine (BDA) was used to confirm the projections and to identify further neuronal pathways potentially involved in amylin signaling. We identified dense projections from the amylin activated neurons in the LPB and sparse projections from the NTS to the dLHA. LPB fiber efferents were found in close proximity to dLHA nuclei activated by 24h of fasting. The AP and the Ce showed no projections to the dLHA. Dense efferents were also observed from the LPB to other hypothalamic areas, namely to the ventromedial, dorsomedial, paraventricular and arcuate nuclei. This study provides neuroanatomical evidence that among the amylin activated areas, the LPB provides the strongest input to the dLHA, thus it may mediate the amylin-induced inhibition of the dLHA.


Assuntos
Amiloide/farmacologia , Depressores do Apetite/farmacologia , Região Hipotalâmica Lateral/citologia , Neurônios/efeitos dos fármacos , Animais , Biotina/análogos & derivados , Biotina/metabolismo , Mapeamento Encefálico , Toxina da Cólera/metabolismo , Dextranos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Masculino , Vias Neurais/fisiologia , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar
9.
Physiol Behav ; 100(5): 511-8, 2010 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-20226802

RESUMO

Amylin is secreted by pancreatic beta-cells and is believed to be a physiological signal of satiation. Amylin's effect on eating has been shown to be mediated via a direct action at the area postrema (AP) via amylin receptors that are heterodimers of the calcitonin receptor core protein with a receptor activity modifying protein. Peripheral amylin leads to accumulation of cyclic guanosine monophosphate, phosphorylated extracellular-signal regulated kinase 1/2 and c-Fos protein in AP neurons. The particular amylin-activated AP neurons mediating its anorexigenic action seem to be noradrenergic. The central pathways mediating amylin's effects have been characterized by lesioning and tracing studies, identifying important connections from the AP to the nucleus of the solitary tract and lateral parabrachial nucleus. Amylin was shown to interact, probably at the brainstem, with other signals involved in the short term control of food intake, namely cholecystokinin, glucagon-like peptide 1 and peptide YY. Amylin also interacts with the adiposity signal leptin; this interaction, which is thought to involve the hypothalamus, may have important implications for the development of new and improved hormonal obesity treatments. In conclusion, amylin actions on food intake seem to reside primarily within the brainstem, and the associated mechanisms are starting to be unraveled. The paper represents an invited review by a symposium, award winner or keynote speaker at the Society for the Study of Ingestive Behavior [SSIB] Annual Meeting in Portland, July 2009.


Assuntos
Amiloide/efeitos adversos , Anorexia/induzido quimicamente , Depressores do Apetite/efeitos adversos , Tronco Encefálico/efeitos dos fármacos , Tronco Encefálico/fisiologia , Animais , Tronco Encefálico/citologia , Ingestão de Alimentos/efeitos dos fármacos , Humanos , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Neurônios/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA