Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 450
Filtrar
1.
Neurosci Res ; 200: 41-47, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37774845

RESUMO

Recent studies has consistently demonstrated a relationship between secretin and autism-like behavior in mice. Therefore, secretin-receptor knockout (SCTR-KO) mice are used to study autism. However, with respect to humans, some studies have reported that secretin administration could improve autistic symptoms in contrast to other studies. A consistent finding revealed that several patients with autism spectrum disorders (ASD) experience comorbid sleep disorders. To examine the relationship between secretin and sleep, we recorded the core body temperature and locomotor activity of SCTR-KO (-/-) and wild-type (WT) (+/+) mice in the baseline condition and after 4 h of sleep deprivation. No significant differences were observed between the SCTR-KO and control mice in the baseline condition. However, during the first dark period following sleep deprivation, we observed an increase in non-rapid eye movement sleep in the SCTR-KO group, which demonstrated that the absence of secretin induces fragmentation making it difficult for the SCTR-KO mice to maintain sleep and wakefulness. Our results follow previous reports that a large proportion of patients with ASD complain of drowsiness and decreased focus during the day. Secretin functions as an intestinal peptide that neutralizes gastric acid and as a neuropeptide in the brain; it also affects social cognitive behavior and acts as a neurotrophic factor. We have proposed that secretin might be a contributing factor to the modulation of sleep.


Assuntos
Secretina , Privação do Sono , Animais , Humanos , Camundongos , Eletroencefalografia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Secretina/fisiologia , Sono/fisiologia
2.
Endocrinology ; 162(9)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34089599

RESUMO

The obesity pandemic requires effective preventative and therapeutic intervention strategies. Successful and sustained obesity treatment is currently limited to bariatric surgery. Modulating the release of gut hormones is considered promising to mimic bariatric surgery with its beneficial effects on food intake, body weight, and blood glucose levels. The gut peptide secretin was the first molecule to be termed a hormone; nevertheless, only recently has it been established as a legitimate anorexigenic peptide. In contrast to gut hormones that crosstalk with the brain either directly or by afferent neuronal projections, secretin mediates meal-associated brown fat thermogenesis to induce meal termination, thereby qualifying this physiological mechanism as an attractive, peripheral target for the treatment of obesity. In this perspective, it is of pivotal interest to deepen our as yet superficial knowledge on the physiological roles of secretin as well as meal-associated thermogenesis in energy balance and body weight regulation. Of note, the emerging differences between meal-associated thermogenesis and cold-induced thermogenesis must be taken into account. In fact, there is no correlation between these 2 entities. In addition, the investigation of potential effects of secretin in hedonic-driven food intake, bariatric surgery and chronic treatment using suitable application strategies to overcome pharmacokinetic limitations will provide further insight into its potential to influence energy balance. The aim of this article is to review the facts on secretin's metabolic effects, address prevailing gaps in our knowledge, and provide an overview on the opportunities and challenges of the therapeutic potential of secretin in body weight control.


Assuntos
Obesidade/prevenção & controle , Saciação/efeitos dos fármacos , Secretina/farmacologia , Animais , Regulação do Apetite/efeitos dos fármacos , Regulação do Apetite/fisiologia , Ingestão de Energia/efeitos dos fármacos , Ingestão de Energia/fisiologia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Humanos , Obesidade/etiologia , Saciação/fisiologia , Secretina/fisiologia , Secretina/uso terapêutico , Termogênese/efeitos dos fármacos
3.
Exp Physiol ; 105(8): 1206-1213, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32271980

RESUMO

NEW FINDINGS: What is the topic of this review? Brown fat's role in meal-associated thermogenesis and the related consequences for energy balance regulation with a focus on the gut hormone secretin, which has been identified as the endocrine molecular mediator of meal-associated brown fat thermogenesis. What advances does it highlight? The finding of the secretin-induced gut-brown fat-brain axis creates new opportunities to manipulate brown fat and thereby energy balance in a natural way while living in a thermoneutral environment. The role of brown fat as a mere catabolic heater organ needs to be revised and more attention should be directed towards the regulatory role of brown fat beyond energy expenditure. ABSTRACT: Brown fat research concentrates on the energy expenditure function of this heating organ, whereas previous evidence for a role of brown fat in regulating energy intake has been mostly neglected. Ingestion of a single mixed meal activates human brown fat thermogenesis to the same degree as cold. In mice, activation of brown fat thermogenesis with a ß3 -adrenergic receptor agonist inhibits food intake. Pharmacological ß-blockade, however, inhibits neither meal-associated thermogenesis nor food intake. We recently identified the gut hormone secretin as a non-adrenergic activator of brown fat. In vivo, secretin treatment acutely increases energy expenditure and inhibits food intake in wild-type, but not in uncoupling protein 1 (UCP1)-knockout (KO) mice, which lack thermogenic brown fat function. Concurrently, secretin alters gene expression of melanocortinergic peptides of hypothalamic neurons in wild-type mice, but not UCP1-KO. Blocking endogenous secretin with a neutralizing antibody attenuates brown fat thermogenesis during refeeding, increases food intake of mice, and alters ad libitum feeding behaviour. Taken together, these findings demonstrate that secretin triggers an endocrine gut-brown adipose tissue-brain axis in the control of satiation. We hypothesize that meal-associated activation of brown adipose tissue thermogenesis induced by secretin results in a rise in brain temperature and increased melanocortinergic signalling. Taken together, brown fat is not a mere heating organ dissipating excess calories but also involved in gut-brain communication in the control of food intake.


Assuntos
Tecido Adiposo Marrom/fisiologia , Encéfalo/fisiologia , Trato Gastrointestinal/fisiologia , Secretina/fisiologia , Termogênese , Animais , Ingestão de Alimentos , Ingestão de Energia , Metabolismo Energético , Camundongos , Camundongos Knockout , Proteína Desacopladora 1
4.
Endocr Dev ; 32: 1-7, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28873390

RESUMO

The physiological studies by British physiologists William Maddock Bayliss and Ernest Henry Starling, at the beginning of the last century, demonstrated the existence of specific messenger molecules (hormones) circulating in the blood that regulate the organ function and physiological mechanisms. These findings led to the concept of endocrinology. The first 2 hormones were secretin, discovered in 1902, and gastrin, discovered in 1905. Both hormones that have been described are produced in the gut. This chapter summarizes the history around the discovery of these 2 hormones, which is perceived as the birth of endocrinology. It is noteworthy that after the discovery of these 2 gastrointestinal hormones, many other hormones were detected outside the gut, and thereafter gut hormones faded from both the clinical and scientific spotlight. Only recently, the clinical importance of the gut as the body's largest endocrine organ producing a large variety of hormones has been realized. Gastrointestinal hormones are essential regulators of metabolism, growth, development and behavior and are therefore the focus of a modern pediatric endocrinologist.


Assuntos
Endocrinologia/história , Hormônios Gastrointestinais/história , Animais , França , Gastrinas/história , Gastrinas/fisiologia , Hormônios Gastrointestinais/fisiologia , Trato Gastrointestinal , Alemanha , História do Século XX , História do Século XXI , Hormônios/história , Humanos , Londres , Pâncreas , Secretina/história , Secretina/fisiologia
5.
Biol Psychiatry ; 81(3): 243-251, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26803341

RESUMO

BACKGROUND: Social recognition underlies social behavior in animals, and patients with psychiatric disorders associated with social deficits show abnormalities in social recognition. Oxytocin is implicated in social behavior and has received attention as an effective treatment for sociobehavioral deficits. Secretin receptor-deficient mice show deficits in social behavior. The relationship between oxytocin and secretin concerning social behavior remains to be determined. METHODS: Expression of c-Fos in oxytocin neurons and release of oxytocin from their dendrites after secretin application were investigated. Social recognition was examined after intracerebroventricular or local injection of secretin, oxytocin, or an oxytocin receptor antagonist in rats, oxytocin receptor-deficient mice, and secretin receptor-deficient mice. Electron and light microscopic immunohistochemical analysis was also performed to determine whether oxytocin neurons extend their dendrites into the medial amygdala. RESULTS: Supraoptic oxytocin neurons expressed the secretin receptor. Secretin activated supraoptic oxytocin neurons and facilitated oxytocin release from dendrites. Secretin increased acquisition of social recognition in an oxytocin receptor-dependent manner. Local application of secretin into the supraoptic nucleus facilitated social recognition, and this facilitation was blocked by an oxytocin receptor antagonist injected into, but not outside of, the medial amygdala. In the medial amygdala, dendrite-like thick oxytocin processes were found to extend from the supraoptic nucleus. Furthermore, oxytocin treatment restored deficits of social recognition in secretin receptor-deficient mice. CONCLUSIONS: The results of our study demonstrate that secretin-induced dendritic oxytocin release from supraoptic neurons enhances social recognition. The newly defined secretin-oxytocin system may lead to a possible treatment for social deficits.


Assuntos
Neurônios/fisiologia , Ocitocina/fisiologia , Reconhecimento Psicológico/fisiologia , Secretina/fisiologia , Comportamento Social , Núcleo Supraóptico/fisiologia , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/fisiologia , Tonsila do Cerebelo/ultraestrutura , Animais , Dendritos/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Ocitocina/administração & dosagem , Ocitocina/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Receptores dos Hormônios Gastrointestinais/genética , Receptores dos Hormônios Gastrointestinais/fisiologia , Receptores de Ocitocina/genética , Receptores de Ocitocina/fisiologia , Secretina/administração & dosagem , Núcleo Supraóptico/metabolismo
6.
FASEB J ; 28(8): 3494-505, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24769669

RESUMO

Secretin, a classical gastrointestinal hormone released from S cells in response to acid and dietary lipid, regulates pleiotropic physiological functions, such as exocrine pancreatic secretion and gastric motility. Subsequent to recently proposed revisit on secretin's metabolic effects, we have confirmed lipolytic actions of secretin during starvation and discovered a hormone-sensitive lipase-mediated mechanistic pathway behind. In this study, a 12 wk high-fat diet (HFD) feeding to secretin receptor-knockout (SCTR(-/-)) mice and their wild-type (SCTR(+/+)) littermates revealed that, despite similar food intake, SCTR(-/-) mice gained significantly less weight (SCTR(+/+): 49.6±0.9 g; SCTR(-/-): 44.7±1.4 g; P<0.05) and exhibited lower body fat content. These SCTR(-/-) mice have corresponding alleviated HFD-associated hyperleptinemia and improved glucose/insulin tolerance. Further analyses indicate that SCTR(-/-) have impaired intestinal fatty acid absorption while having similar energy expenditure and locomotor activity. Reduced fat absorption in the intestine is further supported by lowered postprandial triglyceride concentrations in circulation in SCTR(-/-) mice. In jejunal cells, transcript and protein levels of a key fat absorption regulator, cluster of differentiation 36 (CD36), was reduced in knockout mice, while transcript of Cd36 and fatty-acid uptake in isolated enterocytes was stimulated by secretin. Based on our findings, a novel positive feedback pathway involving secretin and CD36 to enhance intestinal lipid absorption is being proposed.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Gorduras na Dieta/farmacocinética , Absorção Intestinal/genética , Obesidade/genética , Receptores Acoplados a Proteínas G/fisiologia , Receptores dos Hormônios Gastrointestinais/fisiologia , Secretina/fisiologia , Adiposidade/genética , Adiposidade/fisiologia , Animais , Antígenos CD36/fisiologia , Proteínas de Transporte/fisiologia , Metabolismo Energético , Enterócitos/metabolismo , Retroalimentação Fisiológica , Feminino , Teste de Tolerância a Glucose , Resistência à Insulina , Jejuno/metabolismo , Leptina/sangue , Locomoção , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/sangue , Obesidade/etiologia , Obesidade/fisiopatologia , Receptores Acoplados a Proteínas G/deficiência , Receptores dos Hormônios Gastrointestinais/deficiência , Triglicerídeos/sangue , Aumento de Peso
7.
FASEB J ; 28(6): 2632-44, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24599969

RESUMO

Angiotensin (ANGII) and secretin (SCT) share overlapping, interdependent osmoregulatory functions in brain, where SCT peptide/receptor function is required for ANGII action, yet the molecular basis is unknown. Since receptors for these peptides (AT1aR, SCTR) are coexpressed in osmoregulatory centers, a possible mechanism is formation of a cross-class receptor heterocomplex. Here, we demonstrate such a complex and its functional importance to modulate signaling. Association of AT1aR with SCTR reduced ability of SCT to stimulate cyclic adenosine monophosphate (cAMP), with signaling augmented in presence of ANGII or constitutively active AT1aR. Several transmembrane (TM) peptides of these receptors were able to affect their conformation within complexes, reducing receptor BRET signals. AT1aR TM1 affected only formation and activity of the heterocomplex, without effect on homomers of either receptor, and reduced SCT-stimulated cAMP responses in cells expressing both receptors. This peptide was active in vivo by injection into mouse lateral ventricle, thereby suppressing water-drinking behavior after hyperosmotic shock, similar to SCTR knockouts. This supports the interpretation that active conformation of AT1aR is a key modulator of cAMP responses induced by SCT stimulation of SCTR. The SCTR/AT1aR complex is physiologically important, providing differential signaling to SCT in settings of hyperosmolality or food intake, modulated by differences in levels of ANGII.


Assuntos
Angiotensina II/fisiologia , AMP Cíclico/fisiologia , Receptor Tipo 1 de Angiotensina/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores dos Hormônios Gastrointestinais/fisiologia , Secretina/fisiologia , Transdução de Sinais/fisiologia , Animais , Células CHO , Células COS , Chlorocebus aethiops , Cricetulus , Comportamento de Ingestão de Líquido/efeitos dos fármacos , Células HEK293 , Humanos , Ligantes , Proteínas de Membrana/fisiologia , Camundongos , Pressão Osmótica/fisiologia , Multimerização Proteica , Estrutura Quaternária de Proteína/fisiologia , Receptor Tipo 1 de Angiotensina/agonistas , Receptores Acoplados a Proteínas G/agonistas , Receptores dos Hormônios Gastrointestinais/agonistas
8.
Exp Physiol ; 98(5): 973-87, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23243148

RESUMO

Secretin has been implicated in cardiovascular regulation through its specific receptors, as well as through ß-adrenoceptors and nitric oxide, although data on its direct effect on coronary blood flow and cardiac function have remained scarce. The present study aimed to determine the primary in vivo effect of secretin on cardiac function and perfusion and the mechanisms related to the autonomic nervous system, secretin receptors and NO. In addition, in coronary endothelial cells the intracellular pathways involved in the effects of secretin on NO release were also examined. In 30 pigs, intracoronary secretin infusion at 2.97 pg for each millilitre per minute of coronary blood flow at constant heart rate and aortic blood pressure increased coronary blood flow, maximal rate of change of left ventricular pressure, segmental shortening, cardiac output and coronary NO release (P<0.05). These responses were graded in a further five pigs. Moreover, while blockade of muscarinic cholinoreceptors (n=5) and of α-adrenoceptors (n=5) did not abolish the observed responses to secretin, blockade of ß1-adrenoceptors (n=5) prevented the effects of secretin on cardiac function. In addition, blockade of ß2-adrenoceptors (n=5) and NO synthase inhibition (n=5) prevented the coronary response and the effect of secretin on NO release. All these effects were abolished by a secretin receptor inhibitor (n=5). In coronary endothelial cells, the increased NO production caused by secretin was found to be related to cAMP/protein kinase A signalling activated as downstream effectors of stimulation of secretin receptors and ß2-adrenoceptors. In conclusion, in anaesthetized pigs secretin primarily increased cardiac function and perfusion through the involvement of specific receptors, ß-adrenoceptors and NO release.


Assuntos
Coração/efeitos dos fármacos , Óxido Nítrico/fisiologia , Receptores Adrenérgicos beta 1/fisiologia , Secretina/fisiologia , Animais , Células Cultivadas , Circulação Coronária/efeitos dos fármacos , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Coração/fisiologia , Hemodinâmica/efeitos dos fármacos , Contração Miocárdica/efeitos dos fármacos , Perfusão , Receptores Adrenérgicos beta 1/efeitos dos fármacos , Secretina/administração & dosagem , Secretina/farmacologia , Suínos , Vasodilatação/efeitos dos fármacos
9.
Neurosci Lett ; 532: 29-32, 2013 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-23149133

RESUMO

The paraventricular nucleus (PVN) of hypothalamus is a major integrative center in homeostatic control. Morphological studies have revealed a high level of secretin and secretin receptor expression in the PVN. To investigate the direct electrophysiological effects of secretin in the PVN, in vivo extracellular recordings were performed in the present study. In 24 out of the 46 paraventricular neurons, micro-pressure ejection of secretin increased the firing rate from 3.07±0.43 Hz to 4.86±0.70 Hz. In another 8 out of the 46 paraventricular neurons, secretin decreased the firing rate from 2.61±0.46 Hz to 1.41±0.25 Hz. In the remaining 14 paraventricular neurons, secretin did not alter the firing rate significantly. The present findings provided direct electrophysiological evidence for the possible functions of secretin in the PVN.


Assuntos
Potenciais de Ação , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Secretina/fisiologia , Animais , Masculino , Neurônios/efeitos dos fármacos , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Ratos , Ratos Wistar , Secretina/farmacologia
10.
Regul Pept ; 178(1-3): 80-5, 2012 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-22796317

RESUMO

BACKGROUND/AIM: The renin-angiotensin system (RAS) exists in the pancreas, but the role of RAS in the regulation of pancreatic exocrine secretion under physiological conditions has been little known. The present study addressed the RAS's effect on the pancreatic secretion by using valsartan, a specific angiotensin II receptor blocker, in conscious rats. METHOD: Male Wistar rats prepared with pancreatic, biliary, duodenal and jugular vein cannulas were used. To examine the role of RAS in the pancreatic secretion, valsartan at 1, 5, or 25 mg/kg was administered into the duodenum via cannula, and volume of pancreatic juice and protein concentration were determined. In addition, to examine the role of RAS in hormone-stimulated pancreatic hypersecretion, pancreatic secretion was examined in response to stimulation of secretin or cholecystokinin after intraduodenal infusion of valsartan at 25 mg/kg. Furthermore, to examine the mechanism of action of RAS on pancreatic secretion, intravenous infusion of atropine or perivagal application of capsaicin was conducted and then the pancreatic secretion was examined following intraduodenal infusion of valsartan at 25 mg/kg. RESULTS: Volume of pancreatic juice, but not protein output, significantly decreased after administration of valsartan. However, administration of valsartan did not exert significant effects on secretin- or cholesystokinin-stimulated pancreatic secretion. Treatment with atropine and perivagal application of capsaicin completely abolished the suppressive effect of valsartan on pancreatic juice secretion. CONCLUSION: Present results suggest that RAS plays a stimulatory role in pancreatic juice secretion via cholinergic afferent pathway without affecting protein secretion and hormonally stimulated pancreatic secretion under physiological conditions.


Assuntos
Vias Aferentes/efeitos dos fármacos , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Pâncreas/efeitos dos fármacos , Suco Pancreático/metabolismo , Tetrazóis/farmacologia , Nervo Vago/efeitos dos fármacos , Valina/análogos & derivados , Amilases/sangue , Animais , Atropina/farmacologia , Bicarbonatos/metabolismo , Capsaicina/farmacologia , Colecistocinina/fisiologia , Masculino , Antagonistas Muscarínicos/farmacologia , Pâncreas/metabolismo , Proteínas/metabolismo , Ratos , Ratos Wistar , Sistema Renina-Angiotensina , Secretina/fisiologia , Fármacos do Sistema Sensorial/farmacologia , Valina/farmacologia , Valsartana
11.
J Neurosci ; 32(27): 9228-37, 2012 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-22764231

RESUMO

The potassium channel Kv1.2 α-subunit is expressed in cerebellar Purkinje cell (PC) dendrites where its pharmacological inhibition increases excitability (Khavandgar et al., 2005). Kv1.2 is also expressed in cerebellar basket cell (BC) axon terminals (Sheng et al., 1994), where its blockade increases BC inhibition of PCs (Southan and Robertson, 1998a). Secretin receptors are also expressed both in PC dendrites and BC axon terminals (for review, see (Yuan et al., 2011). The effect of secretin on PC excitability is not yet known, but, like Kv1.2 inhibitors, secretin potently increases inhibitory input to PCs (Yung et al., 2001). This suggests secretin may act in part by suppressing Kv1.2. Receptor-mediated endocytosis is a mechanism of Kv1.2 suppression (Nesti et al., 2004). This process can be regulated by protein kinase A (PKA) (Connors et al., 2008). Since secretin receptors activate PKA (Wessels-Reiker et al., 1993), we tested the hypothesis that secretin regulates Kv1.2 trafficking in the cerebellum. Using cell-surface protein biotinylation of rat cerebellar slices, we found secretin decreased cell-surface Kv1.2 levels by modulating Kv1.2 endocytic trafficking. This effect was mimicked by activating adenylate cyclase (AC) with forskolin, and was blocked by pharmacological inhibitors of AC or PKA. Imaging studies identified the BC axon terminal and PC dendrites as loci of AC-dependent Kv1.2 trafficking. The physiological significance of secretin-regulated Kv1.2 endocytosis is supported by our finding that infusion into the cerebellar cortex of either the Kv1.2 inhibitor tityustoxin-Kα, or of the Kv1.2 regulator secretin, significantly enhances acquisition of eyeblink conditioning in rats.


Assuntos
Comportamento Animal/fisiologia , Córtex Cerebelar/citologia , Córtex Cerebelar/metabolismo , Canal de Potássio Kv1.2/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Córtex Cerebelar/fisiologia , Endocitose/efeitos dos fármacos , Endocitose/fisiologia , Canal de Potássio Kv1.2/antagonistas & inibidores , Masculino , Neurônios/fisiologia , Neurotoxinas/farmacologia , Técnicas de Cultura de Órgãos , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Ratos , Ratos Sprague-Dawley , Venenos de Escorpião/farmacologia , Secretina/fisiologia
12.
Mol Endocrinol ; 26(8): 1394-405, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22692904

RESUMO

The secretin receptor (SR), a G protein-coupled receptor, mediates the effects of the gastrointestinal hormone secretin on digestion and water homeostasis. Recently, high SR expression has been observed in pancreatic ductal adenocarcinomas, cholangiocellular carcinomas, gastrinomas, and bronchopulmonary carcinoid tumors. Receptor overexpression associates with enhanced secretin-mediated signaling, but whether this molecule plays an independent role in tumorigenesis is currently unknown. We recently discovered that pheochromocytomas developing in rats affected by the MENX (multiple endocrine neoplasia-like) syndrome express at very high-level Sctr, encoding SR. We here report that SR are also highly abundant on the membranes of rat adrenal and extraadrenal pheochromocytoma, starting from early stages of tumor development, and are functional. PC12 cells, the best characterized in vitro pheochromocytoma model, also express Sctr at high level. Thus, we used them as model to study the role of SR in neoplastic transformation. Small interfering RNA-mediated knockdown of Sctr decreases PC12 cells proliferation and increases p27 levels. The proproliferative effect of SR in PC12 cells is mediated, in part, by the phosphatidylinositol 3 kinase (PI3K)/serine-threonine protein kinase (AKT) pathway. Transfection of Sctr in Y1 adrenocortical carcinoma cells, expressing low endogenous levels of Sctr, stimulates cell proliferation also, in part, via the PI3K/AKT signaling cascade. Because of the link between SR and PI3K/AKT signaling, tumor cells expressing high levels of the receptor (MENX-associated primary pheochromocytoma and NCI-H727 human bronchopulmonary carcinoid cells) respond well and in a SR-dependent manner to PI3K inhibitors, such as NVP-BEZ235. The association between SR levels and response to PI3K inhibition might open new avenues for the treatment of tumors overexpressing this receptor.


Assuntos
Neoplasias das Glândulas Suprarrenais/metabolismo , Proliferação de Células , Feocromocitoma/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Receptores dos Hormônios Gastrointestinais/fisiologia , Transdução de Sinais , Neoplasias das Glândulas Suprarrenais/patologia , Glândulas Suprarrenais/metabolismo , Glândulas Suprarrenais/patologia , Adulto , Idoso , Aminopiridinas/farmacologia , Animais , Apoptose , Sobrevivência Celular , Feminino , Fármacos Gastrointestinais/farmacologia , Técnicas de Silenciamento de Genes , Humanos , Imidazóis/farmacologia , Masculino , Morfolinas/farmacologia , Neuritos/metabolismo , Células PC12 , Feocromocitoma/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinolinas/farmacologia , Interferência de RNA , Ratos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores dos Hormônios Gastrointestinais/genética , Receptores dos Hormônios Gastrointestinais/metabolismo , Secretina/farmacologia , Secretina/fisiologia , Transcrição Gênica
13.
Trends Biochem Sci ; 36(8): 433-43, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21565514

RESUMO

Secretins form megadalton bacterial-membrane channels in at least four sophisticated multiprotein systems that are crucial for translocation of proteins and assembled fibers across the outer membrane of many species of bacteria. Secretin subunits contain multiple domains, which interact with numerous other proteins, including pilotins, secretion-system partner proteins, and exoproteins. Our understanding of the structure of secretins is rapidly progressing, and it is now recognized that features common to all secretins include a cylindrical arrangement of 12-15 subunits, a large periplasmic vestibule with a wide opening at one end and a periplasmic gate at the other. Secretins might also play a key role in the biogenesis of their cognate secretion systems.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Modelos Moleculares , Secretina/química , Secretina/fisiologia , Proteínas da Membrana Bacteriana Externa/química , Proteínas de Bactérias/química , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Bactérias Gram-Negativas/química , Bactérias Gram-Negativas/metabolismo , Proteínas de Membrana Transportadoras/química , Ligação Proteica , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transporte Proteico
14.
J Infect Dis ; 203(10): 1369-77, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21502078

RESUMO

BACKGROUND: The role of toxins secreted by the type II secretion system (T2SS) of Pseudomonas aeruginosa during lung infection has been uncertain despite decades of research. METHODS: Using a model of pneumonia in Toll-like receptor (TLR) 2,4(-/-) mice, we reexamined the role of the T2SS system. Flagellin-deficient mutants of P. aeruginosa, with mutations in the T2SS and/or T3SS, were used to infect mice. Mice were followed up for survival, with some killed at different intervals to study bacterial clearance, inflammatory responses, and lung pathology. RESULTS: Strains carrying either secretion system were lethal for mice. Double mutants were avirulent. The T3SS(+) strains killed mice within a day, and the T2SS(+) strains killed them later. Mice infected with a strain that had only the T2SS were unable to eradicate the organism from the lungs, whereas those infected with a T2SS-T3SS double deletion were able to clear this mutant. Death caused by the T2SS(+) strain was accompanied by a >50-fold increase in bacterial counts and higher numbers of viable intracellular bacteria. CONCLUSIONS: The T2SS of P. aeruginosa may play a role in death from pneumonia, but its action is delayed. These data suggest that antitoxin strategies against this organism will require measures against the toxins secreted by both T2SS and T3SS.


Assuntos
Toxinas Bacterianas/metabolismo , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/mortalidade , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/mortalidade , Pseudomonas aeruginosa/genética , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/microbiologia , Flagelina/genética , Flagelina/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Imunidade Inata , Imunocompetência , Pulmão/patologia , Camundongos , Camundongos Knockout , Mutação , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/patologia , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidade , Secretina/fisiologia , Fatores de Tempo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
15.
Neuropsychopharmacology ; 36(2): 459-71, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20927047

RESUMO

Secretin (Sct) is released into the circulation postprandially from the duodenal S-cells. The major functions of Sct originated from the gastrointestinal system are to delay gastric emptying, stimulate fluid secretion from pancreas and liver, and hence optimize the digestion process. In recent years, Sct and its receptor (Sctr) have been identified in discrete nuclei of the hypothalamus, including the paraventricular nucleus (PVN) and the arcuate nucleus (Arc). These nuclei are the primary brain sites that are engaged in regulating body energy homeostasis, thus providing anatomical evidence to support a functional role of Sct in appetite control. In this study, the effect of Sct on feeding behavior was investigated using wild-type (wt), Sct(-/-), and secretin receptor-deficient (Sctr(-/-)) mice. We found that both central and peripheral administration of Sct could induce Fos expression in the PVN and Arc, suggesting the activation of hypothalamic feeding centers by this peptide. Consistent with this notion, Sct was found to increase thyrotropin-releasing hormone and melanocortin-4 receptor (Mc4r) transcripts in the PVN, and augment proopiomelanocortin, but reduces agouti-related protein mRNA expression in the Arc. Injection of Sct was able to suppress food intake in wt mice, but not in Sctr(-/-) mice, and that this effect was abolished upon pretreatment with SHU9119, an antagonist for Mc4r. In summary, our data suggest for the first time that Sct is an anorectic peptide, and that this function is mediated by the melanocortin system.


Assuntos
Regulação do Apetite/fisiologia , Regulação para Baixo/fisiologia , Comportamento Alimentar/fisiologia , Hipotálamo/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptores Acoplados a Proteínas G/fisiologia , Receptores dos Hormônios Gastrointestinais/fisiologia , Secretina/administração & dosagem , Animais , Depressores do Apetite/química , Depressores do Apetite/metabolismo , Depressores do Apetite/farmacologia , Regulação do Apetite/efeitos dos fármacos , Regulação do Apetite/genética , Núcleo Arqueado do Hipotálamo/química , Núcleo Arqueado do Hipotálamo/fisiologia , Regulação para Baixo/genética , Comportamento Alimentar/psicologia , Hipotálamo/citologia , Injeções Intraperitoneais , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Knockout , Neurônios/química , Neurônios/metabolismo , Neurônios/fisiologia , Pró-Opiomelanocortina/fisiologia , Distribuição Aleatória , Receptores Acoplados a Proteínas G/deficiência , Receptores Acoplados a Proteínas G/genética , Receptores dos Hormônios Gastrointestinais/deficiência , Receptores dos Hormônios Gastrointestinais/genética , Secretina/deficiência , Secretina/fisiologia
16.
J Endocrinol Invest ; 33(4): 266-75, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20503484

RESUMO

Metabolic pathologies such as Type 2 Diabetes have become a major health problem for worldwide populations. Unfortunately, efforts to cure and especially to prevent these significant global problems have so far been met with disappointment. Recently, the involvement of the gut-derived hormonal dysregulation in the development of obesity-related disturbances has been intensively studied. For instance, studies of gut-derived peptides such as peptide YY 3-36, glucagon-like peptide-1, oxyntomodulin and, more recently, ghrelin have significantly improved our understanding of mechanisms underlying weight and metabolic regulation. Even though early reports of the existence of secretin, the first peptide hormone to be described, date back as far as 1825, so much and yet so little is still known about its physiological role in mammals, including humans. However, recent years have provided a better understanding of how the release of secretin is regulated by enteral secretagogues. On the other hand, most basic questions about its role in the post-prandial regulation of metabolic functions in normal and pathophysiological conditions remain to be elucidated. The present work intends to review the physiology of secretin along with its central and peripheral outcomes on metabolic functions.


Assuntos
Secretina/metabolismo , Secretina/fisiologia , Animais , Sistema Nervoso Central/anatomia & histologia , Sistema Nervoso Central/fisiologia , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Fragmentos de Peptídeos , Peptídeo YY/genética , Peptídeo YY/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores dos Hormônios Gastrointestinais/genética , Receptores dos Hormônios Gastrointestinais/metabolismo , Secretina/genética , Distribuição Tecidual
17.
Nutr Res Rev ; 23(1): 146-54, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20193096

RESUMO

The most widely used pharmacological therapies for obesity and weight management are based on inhibition of gastrointestinal lipases, resulting in a reduced energy yield of ingested foods by reducing dietary lipid absorption. Colipase-dependent pancreatic lipase is believed to be the major gastrointestinal enzyme involved in catalysis of lipid ester bonds. There is scant literature on the action of pancreatic lipase under the range of physiological conditions that occur within the human small intestine, and the literature that does exist is often contradictory. Due to the importance of pancreatic lipase activity to nutrition and weight management, the present review aims to assess the current body of knowledge with regards to the physiology behind the action of this unique gastrointestinal enzyme system. Existing data would suggest that pancreatic lipase activity is affected by intestinal pH, the presence of colipase and bile salts, but not by the physiological range of Ca ion concentration (as is commonly assumed). The control of secretion of pancreatic lipase and its associated factors appears to be driven by gastrointestinal luminal content, particularly the presence of acid or digested proteins and fats in the duodenal lumen. Secretion of colipase, bile acids and pancreatic lipase is driven by cholecystokinin and secretin release.


Assuntos
Digestão/fisiologia , Lipase/metabolismo , Metabolismo dos Lipídeos/fisiologia , Pâncreas/enzimologia , Ácidos e Sais Biliares/farmacologia , Cálcio/fisiologia , Cátions , Colecistocinina/fisiologia , Colipases/fisiologia , Inibidores Enzimáticos/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Intestino Delgado/química , Intestino Delgado/fisiologia , Intestinos/química , Lactonas/farmacologia , Lipase/antagonistas & inibidores , Lipólise , Orlistate , Secretina/fisiologia
18.
Int J Cancer ; 127(1): 43-54, 2010 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19904746

RESUMO

Secretin plays a key role in the regulation of normal cholangiocyte physiology via secretin receptor (SCTR). SCTR expression is upregulated during extrahepatic cholestasis induced by bile duct ligation and closely associated with cholangiocyte proliferative responses. Although well studied in normal cholangiocytes, the role of secretin and the expression of SCTR in the regulation of cholangiocarcinoma proliferation are unknown. In vitro, secretin (10(-7) M) displayed differential effects on normal cholangiocyte [H-69 and human intrahepatic biliary epithelial cell line (HIBEpiC)] and cholangiocarcinoma (Mz-ChA-1, HuH-28, TFK-1, SG231, CCLP1 and HuCC-T1) cell lines as such secretin is mitogenic for normal cholangiocytes and antiproliferative for cholangiocarcinoma. As expected in normal cholangiocytes (HIBEpiC), secretin increased intracellular cyclic adenosine monophosphate (cAMP) levels. However, the effect of secretin on intracellular cAMP levels was suppressed in Mz-ChA-1 cells. Secretin-stimulated intracellular cAMP levels in Mz-ChA-1 were restored by pretreatment with pertussis toxin, suggesting that the receptor coupled to Galpha(i) rather than Galpha(s). SCTR expression was found to be downregulated in 4 of the 6 cholangiocarcinoma cell lines evaluated and in human cholangiocarcinoma biopsy samples. In vivo, secretin significantly inhibited the tumor size and more than doubled tumor latency, which was associated with a decrease in proliferating cell nuclear antigen and an increase in cleaved-caspase 3 expression levels. Our results demonstrate that secretin and/or the modulation of SCTR expression might have potential as a therapeutic tool in the treatment of cholangiocarcinoma.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Colangiocarcinoma/patologia , AMP Cíclico/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo , Secretina/fisiologia , Transdução de Sinais/fisiologia , Divisão Celular/fisiologia , Linhagem Celular Tumoral , Citometria de Fluxo , Imunofluorescência , Humanos , Reação em Cadeia da Polimerase
19.
Proc Natl Acad Sci U S A ; 106(37): 15961-6, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19805236

RESUMO

Hypothalamic magnocellular neurons express either one of the neurohypophysial hormones, vasopressin or oxytocin, along with different neuropeptides or neuromodulators. Axonal terminals of these neurons are generally accepted to release solely the two hormones but not others into the circulation. Here, we show that secretin, originally isolated from upper intestinal mucosal extract, is present throughout the hypothalamo-neurohypophysial axis and that it is released from the posterior pituitary under plasma hyperosmolality conditions. In the hypothalamus, it stimulates vasopressin expression and release. Considering these findings together with our previous findings that show a direct effect of secretin on renal water reabsorption, we propose here that secretin works at multiple levels in the hypothalamus, pituitary, and kidney to regulate water homeostasis. Findings presented here challenge previous understanding regarding the neurohypophysis and could provide new concepts in treating disorders related to osmoregulation.


Assuntos
Água Corporal/fisiologia , Neuro-Hipófise/fisiologia , Secretina/fisiologia , Animais , Sequência de Bases , Primers do DNA/genética , Expressão Gênica , Homeostase/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Imuno-Histoquímica , Rim/fisiologia , Modelos Neurológicos , Sistemas Neurossecretores/fisiologia , Neurotransmissores/genética , Neurotransmissores/fisiologia , Concentração Osmolar , Ratos , Secretina/sangue , Secretina/genética , Vasopressinas/genética , Vasopressinas/fisiologia
20.
Adv Physiol Educ ; 33(3): 165-8, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19745041

RESUMO

The regulation of gastric acid secretion has been the subject of investigation for over a century. Inhibition of gastrin-induced acid secretion by the intestine-derived hormone secretin provides a classic physiological example of negative feedback in the gastrointestinal tract. A classic paper by Leonard R. Johnson and Morton I. Grossman clearly shows the ability of secretin to negatively regulate gastric acid secretion, providing students with an example of this feedback loop. In addition, this article demonstrates the step forward in gastrointestinal endocrinology that occurred when pure preparations of secretin and other gastrointestinal hormones first became available. The comparison of the effects of exogenous, purified secretin to the physiological stimulus of acid in the duodenum is an important example of how newly available reagents allow scientists such as Johnson and Grossman to clarify the mechanisms behind previously established processes. One or more figures from this classic paper can be used to give students insight into the role of secretin in the regulation of the function of the gastrointestinal tract and will also give students a clear example of how the careful experimentation and clear interest in gastrointestinal physiology led Johnson and Grossman to advance the field.


Assuntos
Ácido Gástrico/metabolismo , Secretina/fisiologia , Ensino , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...