Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 585(7826): 591-596, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32526765

RESUMEN

Recent clinical and experimental evidence has evoked the concept of the gut-brain axis to explain mutual interactions between the central nervous system and gut microbiota that are closely associated with the bidirectional effects of inflammatory bowel disease and central nervous system disorders1-4. Despite recent advances in our understanding of neuroimmune interactions, it remains unclear how the gut and brain communicate to maintain gut immune homeostasis, including in the induction and maintenance of peripheral regulatory T cells (pTreg cells), and what environmental cues prompt the host to protect itself from development of inflammatory bowel diseases. Here we report a liver-brain-gut neural arc that ensures the proper differentiation and maintenance of pTreg cells in the gut. The hepatic vagal sensory afferent nerves are responsible for indirectly sensing the gut microenvironment and relaying the sensory inputs to the nucleus tractus solitarius of the brainstem, and ultimately to the vagal parasympathetic nerves and enteric neurons. Surgical and chemical perturbation of the vagal sensory afferents at the hepatic afferent level reduced the abundance of colonic pTreg cells; this was attributed to decreased aldehyde dehydrogenase (ALDH) expression and retinoic acid synthesis by intestinal antigen-presenting cells. Activation of muscarinic acetylcholine receptors directly induced ALDH gene expression in both human and mouse colonic antigen-presenting cells, whereas genetic ablation of these receptors abolished the stimulation of antigen-presenting cells in vitro. Disruption of left vagal sensory afferents from the liver to the brainstem in mouse models of colitis reduced the colonic pTreg cell pool, resulting in increased susceptibility to colitis. These results demonstrate that the novel vago-vagal liver-brain-gut reflex arc controls the number of pTreg cells and maintains gut homeostasis. Intervention in this autonomic feedback feedforward system could help in the development of therapeutic strategies to treat or prevent immunological disorders of the gut.


Asunto(s)
Encéfalo/citología , Intestinos/citología , Intestinos/inervación , Hígado/citología , Hígado/inervación , Neuronas/fisiología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Vías Aferentes , Animales , Células Presentadoras de Antígenos/inmunología , Colitis/inmunología , Colitis/metabolismo , Colitis/patología , Homeostasis , Humanos , Intestinos/inmunología , Masculino , Ratones , Ratas , Receptores Muscarínicos/metabolismo , Bazo/citología , Bazo/inmunología , Nervio Vago/fisiología
2.
J Physiol ; 601(12): 2293-2306, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37126218

RESUMEN

The precise regulation of blood glucose levels is indispensable for maintaining physiological functions. C1 neurons determine the outflow of the autonomic nervous and endocrine systems to maintain blood glucose levels in the body. In contrast, activation of C1 neurons induces a decrease in activity, suggesting that hypoactivity also participates in maintaining blood glucose levels. To examine this, we evaluated both glycogenolysis and hypometabolism induced by the selective activation of C1 neurons. We used DbhCre/0 mice expressing receptors for chemogenetic tools in C1 neurons, resulting from microinjection of the viral vector. C1 neurons were activated by intraperitoneal injection of clozapine N-oxide (CNO). The chemogenetic activation of C1 neurons significantly decreased body temperature, oxygen consumption and carbon dioxide production. On the other hand, blood glucose levels were increased by activation of C1 neurons 2 h after CNO administration, even in the fasting state. In this situation, an increase in glucagon and corticosterone levels was observed, while hepatic glycogen content decreased significantly. Plasma insulin levels were not changed by the activation of C1 neurons despite the increase in blood glucose level. Furthermore, adrenal sympathetic nerve activity was significantly increased by the activation of C1 neurons, and plasma catecholamine levels increased significantly. In conclusion, the selective activation of C1 neurons using chemogenetic tools induced an increase in blood glucose levels, probably as a result of hepatic glycogenolysis and hypometabolism. KEY POINTS: Chemogenetic activation of C1 neurons in medulla oblongata decreased body temperature. Oxygen consumption and carbon dioxide production were decreased by chemogenetic activation of C1 neurons in medulla oblongata. Blood glucose levels were increased by chemogenetic activation of C1 neurons in medulla oblongata. Chemogenetic activation of C1 neurons in medulla oblongata increased glucagon, corticosterone and catecholamine levels in plasma. An increase in blood glucose levels by activation of C1 neurons occurred due to the combined effect of hepatic glycogenolysis and hypometabolism.


Asunto(s)
Glucemia , Glucogenólisis , Ratones , Animales , Glucagón , Corticosterona/farmacología , Dióxido de Carbono , Neuronas/fisiología , Bulbo Raquídeo/fisiología , Catecolaminas
3.
Am J Physiol Regul Integr Comp Physiol ; 324(2): R152-R160, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36534584

RESUMEN

Autonomic nerves, including the sympathetic and parasympathetic nerves, control the immune system along with their physiological functions. On the peripheral side, the interaction between the splenic sympathetic nerves and immune cells is important for the anti-inflammatory effects. However, the central mechanism underlying these anti-inflammatory effects remains unclear. C1 neurons respond to stressors and subsequently determine the outflow of the autonomic nervous system. We have previously shown that C1 neurons protect against acute kidney injury and found a signaling connection between peripheral vestibular organs and C1 neurons. Thus, we hypothesized that hypergravity load or galvanic vestibular stimulation (GVS) might protect against acute lung injury. We showed that C1 neurons are histologically and functionally activated by stimulating the peripheral vestibular organs. Protection against acute lung injury that was induced by a 2 G load disappeared due to vestibular lesions or the deletion of C1 neurons. This GVS-induced protective effect was also eliminated by the deletion of the C1 neurons. Furthermore, GVS increased splenic sympathetic nerve activity in conscious mice, and splenic sympathetic denervation abolished the GVS-induced protection against acute lung injury. Therefore, the activated pathway between C1 neurons and splenic sympathetic nerves is indispensable for GVS-induced protection against acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda , Vestíbulo del Laberinto , Ratones , Animales , Neuronas/fisiología , Bulbo Raquídeo/fisiología , Lesión Pulmonar Aguda/prevención & control , Antiinflamatorios , Estimulación Eléctrica
4.
Brain Behav Immun ; 111: 138-150, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37037362

RESUMEN

The immune system is known to be controlled by the autonomic nervous system including sympathetic and parasympathetic (vagus) nerves. C1 neurons in the medulla oblongata, which participate in the control of the autonomic nervous system, are responders to stressors and regulate the immune system. Short-term activation of C1 neurons suppresses inflammation, while the effect of a long-term activation of these neurons on the inflammatory reflex is unclear. We, herein, demonstrate that the coactivation of both the splenic sympathetic nerves and the adrenal gland adrenergic response are indispensable for the prognosis of acute lung injury. The chemogenetic activation of C1 neurons increased plasma catecholamine including adrenaline and noradrenaline levels. The deletion of catecholaminergic cells using local injections of viral vector in the adrenal gland abolished the protective effect against acute lung injury when the C1 neurons were stimulated by either chemogenetic or optogenetic tools. Furthermore, repeated activation of C1 neurons using chemogenetic tool inhibited the adrenal response without affecting the plasma noradrenaline levels, eliminated the protective effect against acute lung injury. This was rescued by the isoprenaline administration. We concluded that the maintenance of an adrenergic response via C1 neurons in the adrenal gland is a prerequisite for the delivery of an effective anti-inflammatory response.


Asunto(s)
Adrenérgicos , Neuronas , Adrenérgicos/farmacología , Bulbo Raquídeo/fisiología , Glándulas Suprarrenales , Norepinefrina/farmacología , Antiinflamatorios/farmacología
5.
Diabetes Obes Metab ; 25(6): 1534-1546, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36852745

RESUMEN

AIM: To clarify the effects of glucose-dependent insulinotropic polypeptide (GIP) receptor agonists (GIPRAs) on feeding and body weight. MATERIALS AND METHODS: Acute and subchronic effects of subcutaneous GIPFA-085, a long-acting GIPRA, on blood glucose, food intake, body weight, respiratory exchange ratio and plasma leptin levels were measured in diet-induced obese (DIO) mice and/or functional leptin-deficient ob/ob mice. The effects of GIPFA-085 on the hypothalamic arcuate nucleus (ARC) neurons from lean and DIO mice were studied by measuring cytosolic Ca2+ concentration ([Ca2+ ]i ). RESULTS: Single bolus GIPFA-085 (30, 300 nmol/kg) dose-dependently reduced blood glucose in glucose tolerance tests, elevated plasma leptin levels at 0.5-6 hours and inhibited food intake at 2-24 hours after injection in DIO mice. Daily GIPFA-085 (300 nmol/kg) inhibited food intake and increased fat utilization on day 1, and reduced body weight gain on days 3-12 of treatment in DIO, but not ob/ob, mice. GIPFA-085 increased [Ca2+ ]i in the ARC leptin-responsive and proopiomelanocortin (POMC) neurons. GIPFA-085 and leptin cooperated to increase [Ca2+ ]i in ARC neurons and inhibit food intake. CONCLUSIONS: GIPFA-085 acutely inhibits feeding and increases lipid utilization, and sustainedly lowers body weight in DIO mice via mechanisms involving rises in leptin and activation of ARC leptin-responsive and POMC neurons. This study highlights the therapeutic potential of GIPRAs for treating obesity and diabetes.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Leptina , Ratones , Animales , Leptina/metabolismo , Proopiomelanocortina/metabolismo , Proopiomelanocortina/farmacología , Proopiomelanocortina/uso terapéutico , Glucemia , Obesidad/tratamiento farmacológico , Obesidad/etiología , Dieta , Peso Corporal , Receptores Acoplados a Proteínas G , Neuronas/metabolismo , Ratones Endogámicos C57BL
6.
Biochem Biophys Res Commun ; 519(3): 553-558, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31537381

RESUMEN

Oxytocin (Oxt), a neurohormone synthesized in the neurons of hypothalamic paraventricular nucleus (PVN) and supraoptic nucleus induces milk-ejection and uterine contraction and regulates social behavior, stress responses, memory and food intake. Peripheral (intraperitoneal and subcutaneous) infusion of Oxt decreases food intake and body weight in obese animals via mechanisms involving vagal afferent nerves and in obese subjects when administered nasally. Peripherally injected and intracerebroventricularly injected Oxt inhibit food intake to similar extent and with similar time course. Thus, peripheral Oxt mimics the effects of central Oxt, however, underlying mechanisms are unclear. In the present study we explored whether intraperitoneal Oxt activates Oxt neurons in PVN via vagal afferents and whether this pathway is linked to inhibition of feeding. We here show that intraperitoneal Oxt injection induces c-Fos expression in PVN largely in Oxt neurons and inhibits food intake, and these effects are blunted by subdiaphragmatic vagotomy. The intraperitoneal Oxt-induced inhibition of food intake was blunted in Oxt KO mice, by intracerebroventricular injection of Oxt receptor antagonist, and by vagotomy. These results demonstrate that intraperitoneal Oxt injection activates PVN Oxt neurons via vagal afferent nerves, thereby inhibiting food intake. This vagal afferents-mediated Oxt's peripheral-to-central coupling may serve to promote satiety and possibly a series of neural functions of Oxt and to treat their disorders.


Asunto(s)
Ingestión de Alimentos , Neuronas/metabolismo , Oxitocina/metabolismo , Vagotomía , Animales , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxitocina/deficiencia , Receptores de Oxitocina/antagonistas & inhibidores , Receptores de Oxitocina/metabolismo
7.
Neurochem Res ; 42(9): 2658-2665, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28462451

RESUMEN

Molecular hydrogen (H2), as a new medical gas, has protective effects in neurological disorders including Parkinson's disease (PD). In our previous report, the neuroprotective effect of drinking water with saturated H2 (H2 water) in PD mice might be due to stomach-brain interaction via release of gastric hormone, ghrelin. In the present study, we assessed the effect of H2-induced ghrelin more precisely. To confirm the contribution of ghrelin in H2 water-drinking PD model mice, ghrelin-knock out (KO) mice were used. Despite the speculation, the effect of H2 water was still observed in ghrelin-KO PD model mice. To further check the involvement of ghrelin, possible contribution of ghrelin-induced vagal afferent effect was tested by performing subdiaphragmatic vagotomy before treating with H2 water and administration of MPTP (1-methyl- 4-phenyl-1,2,3,6-tetrahydropyridine). The protective effect of H2 water was still observed in the vagotomized mice in substantia nigra, suggesting that stimulation of vagal afferent nerves is not involved in H2-induced neuroprotection. Other neuroprotective substitutes in ghrelin-KO mice were speculated because H2-induced neuroprotection was not cancelled by ghrelin receptor antagonist, D-Lys3 GHRP-6, in ghrelin-KO PD model mice, unlike in wild-type PD model mice. Our results indicate that ghrelin may not be the only factor for H2-induced neuroprotection and other factors can substitute the role of ghrelin when ghrelin is absent, raising intriguing options of research for H2-responsive factors.


Asunto(s)
Encéfalo/metabolismo , Deuterio/administración & dosificación , Mucosa Gástrica/metabolismo , Ghrelina/deficiencia , Trastornos Parkinsonianos/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Ghrelina/antagonistas & inhibidores , Ghrelina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos Parkinsonianos/tratamiento farmacológico , Trastornos Parkinsonianos/patología , Estómago/efectos de los fármacos , Vagotomía/métodos , Nervio Vago/metabolismo , Nervio Vago/cirugía
8.
Am J Physiol Endocrinol Metab ; 311(5): E881-E890, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27702746

RESUMEN

Caspase-1 is a cysteine protease responsible for the processing of the proinflammatory cytokine interleukin-1ß and activated by the formation of inflammasome complexes. Although several investigations have found a link between diet-induced obesity and caspase-1, the relationship remains controversial. Here, we found that mice deficient in caspase-1 were susceptible to high-fat diet-induced obesity with increased adiposity as well as normal lipid and glucose metabolism. Caspase-1 deficiency clearly promoted the infiltration of inflammatory macrophages and increased the production of C-C motif chemokine ligand 2 (CCL2) in the adipose tissue. The dominant cellular source of CCL2 was stromal vascular fraction rather than adipocytes in the adipose tissue. These findings demonstrate a critical role of caspase-1 in macrophage-driven inflammation in the adipose tissue and the development of obesity. These data provide novel insights into the mechanisms underlying inflammation in the pathophysiology of obesity.


Asunto(s)
Tejido Adiposo/inmunología , Caspasa 1/genética , Quimiocina CCL2/inmunología , Macrófagos/inmunología , Obesidad/genética , Adipocitos/inmunología , Adipocitos/patología , Adiponectina/inmunología , Tejido Adiposo/patología , Animales , Glucemia/metabolismo , Composición Corporal , Caspasa 1/inmunología , Colesterol/metabolismo , Dieta Alta en Grasa , Citometría de Flujo , Perfilación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Insulina/metabolismo , Interferón gamma/inmunología , Interleucina-12/inmunología , Interleucina-18/inmunología , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Leptina/inmunología , Masculino , Ratones , Ratones Noqueados , Obesidad/inmunología , Obesidad/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Triglicéridos/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Microtomografía por Rayos X
9.
Biochem Biophys Res Commun ; 456(3): 727-32, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25511693

RESUMEN

Glucagon is released from the pancreatic islets postprandially and under hypoglycemic and cold conditions, and regulates glucose metabolism, feeding, energy expenditure and heat production, the functions partly controlled by the brain. Peripheral glucagon could signal to the brain via passing through the blood-brain barrier and/or acting on the vagal afferent. However, the latter remains to be determined. The present study aimed to clarify whether glucagon directly interacts with the nodose ganglion (NG) neurons of vagal afferent nerves in mice. In vivo study showed that intraperitoneal injection of glucagon induced phosphorylation of extracellular signal regulated kinase 1 and 2 (ERK1/2), cellular activation makers, in NG neurons. In fura-2 microfluorometric studies, glucagon increased cytosolic Ca(2+) concentration ([Ca(2+)]i) in single NG neurons. The glucagon-induced [Ca(2+)]i increases were suppressed by a glucagon receptor antagonist, des-His(1)-[Glu(9)]-Glucagon (1-29) amide, and the glucagon receptor mRNA was expressed in NG neurons. The majority of glucagon-responsive NG neurons exhibited [Ca(2+)]i responses to insulin and cholecystokinin-8, the hormones that are secreted postprandially and implicated in satiety. These results demonstrate that glucagon, by interacting with the glucagon receptor, directly activates vagal afferent nerves, possibly being relayed to the signaling to the brain and formation of satiety.


Asunto(s)
Señalización del Calcio/fisiología , Glucagón/fisiología , Neuronas Aferentes/fisiología , Ganglio Nudoso/fisiología , Receptores de Glucagón/fisiología , Respuesta de Saciedad/fisiología , Nervio Vago/fisiología , Animales , Señalización del Calcio/efectos de los fármacos , Glucagón/administración & dosificación , Glucagón/análogos & derivados , Glucagón/farmacología , Humanos , Inyecciones Intraperitoneales , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/enzimología , Ganglio Nudoso/efectos de los fármacos , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Glucagón/antagonistas & inhibidores , Respuesta de Saciedad/efectos de los fármacos , Nervio Vago/efectos de los fármacos
10.
Am J Physiol Regul Integr Comp Physiol ; 309(5): R561-75, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26157056

RESUMEN

d-Serine is abundant in the forebrain and physiologically important for modulating excitatory glutamatergic neurotransmission as a coagonist of synaptic N-methyl-d-aspartate (NMDA) receptor. NMDA signaling has been implicated in the control of food intake. However, the role of d-serine on appetite regulation is unknown. To clarify the effects of d-serine on appetite, we investigated the effect of oral d-serine ingestion on food intake in three different feeding paradigms (one-food access, two-food choice, and refeeding after 24-h fasting) using three different strains of male mice (C57Bl/6J, BKS, and ICR). The effect of d-serine was also tested in leptin signaling-deficient db/db mice and sensory-deafferented (capsaicin-treated) mice. The expression of orexigenic neuropeptides [neuropeptide Y (Npy) and agouti-related protein (Agrp)] in the hypothalamus was compared in fast/refed experiments. Conditioned taste aversion for high-fat diet (HFD) was tested in the d-serine-treated mice. Under the one-food-access paradigm, some of the d-serine-treated mice showed starvation, but not when fed normal chow. HFD feeding with d-serine ingestion did not cause aversion. Under the two-food-choice paradigm, d-serine suppressed the intake of high-preference food but not normal chow. d-Serine also effectively suppressed HFD intake but not normal chow in db/db mice and sensory-deafferented mice. In addition, d-serine suppressed normal chow intake after 24-h fasting despite higher orexigenic gene expression in the hypothalamus. d-Serine failed to suppress HFD intake in the presence of L-701,324, the selective and full antagonist at the glycine-binding site of the NMDA receptor. Therefore, d-serine suppresses the intake of high-preference food through coagonism toward NMDA receptors.


Asunto(s)
Depresores del Apetito/farmacología , Ingestión de Alimentos/efectos de los fármacos , Agonistas de Aminoácidos Excitadores/farmacología , Conducta Alimentaria/efectos de los fármacos , Preferencias Alimentarias/efectos de los fármacos , Receptores de N-Metil-D-Aspartato/agonistas , Serina/farmacología , Proteína Relacionada con Agouti/metabolismo , Animales , Conducta de Elección , Condicionamiento Psicológico , Dieta Alta en Grasa , Regulación hacia Abajo , Antagonistas de Aminoácidos Excitadores/farmacología , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Neuropéptido Y/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Fármacos del Sistema Sensorial , Factores de Tiempo
11.
Am J Physiol Regul Integr Comp Physiol ; 308(5): R360-9, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25540101

RESUMEN

Oxytocin (Oxt), a neuropeptide produced in the hypothalamus, is implicated in regulation of feeding. Recent studies have shown that peripheral administration of Oxt suppresses feeding and, when infused subchronically, ameliorates hyperphagic obesity. However, the route through which peripheral Oxt informs the brain is obscure. This study aimed to explore whether vagal afferents mediate the sensing and anorexigenic effect of peripherally injected Oxt in mice. Intraperitoneal Oxt injection suppressed food intake and increased c-Fos expression in nucleus tractus solitarius to which vagal afferents project. The Oxt-induced feeding suppression and c-Fos expression in nucleus tractus solitarius were blunted in mice whose vagal afferent nerves were blocked by subdiaphragmatic vagotomy or capsaicin treatment. Oxt induced membrane depolarization and increases in cytosolic Ca(2+) concentration ([Ca(2+)]i) in single vagal afferent neurons. The Oxt-induced [Ca(2+)]i increases were markedly suppressed by Oxt receptor antagonist. These Oxt-responsive neurons also responded to cholecystokinin-8 and contained cocaine- and amphetamine-regulated transcript. In obese diabetic db/db mice, leptin failed to increase, but Oxt increased [Ca(2+)]i in vagal afferent neurons, and single or subchronic infusion of Oxt decreased food intake and body weight gain. These results demonstrate that peripheral Oxt injection suppresses food intake by activating vagal afferent neurons and thereby ameliorates obesity in leptin-resistant db/db mice. The peripheral Oxt-regulated vagal afferent neuron provides a novel target for treating hyperphagia and obesity.


Asunto(s)
Fármacos Antiobesidad/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Hiperfagia/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Oxitocina/administración & dosificación , Nervio Vago/efectos de los fármacos , Potenciales de Acción , Animales , Depresores del Apetito/administración & dosificación , Calcio/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Hiperfagia/fisiopatología , Inyecciones Intraperitoneales , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/metabolismo , Obesidad/fisiopatología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Fármacos del Sistema Sensorial/farmacología , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/metabolismo , Núcleo Solitario/fisiopatología , Factores de Tiempo , Vagotomía , Nervio Vago/metabolismo , Nervio Vago/fisiopatología , Aumento de Peso/efectos de los fármacos
12.
Biochem Biophys Res Commun ; 451(2): 276-81, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-25089000

RESUMEN

Glucagon-like peptide-1 (GLP-1) receptor agonists have been used to treat type 2 diabetic patients and shown to reduce food intake and body weight. The anorexigenic effects of GLP-1 and GLP-1 receptor agonists are thought to be mediated primarily via the hypothalamic paraventricular nucleus (PVN). GLP-1, an intestinal hormone, is also localized in the nucleus tractus solitarius (NTS) of the brain stem. However, the role of endogenous GLP-1, particularly that in the NTS neurons, in feeding regulation remains to be established. The present study examined whether the NTS GLP-1 neurons project to PVN and whether the endogenous GLP-1 acts on PVN to restrict feeding. Intra-PVN injection of GLP-1 receptor antagonist exendin (9-39) increased food intake. Injection of retrograde tracer into PVN combined with immunohistochemistry for GLP-1 in NTS revealed direct projection of NTS GLP-1 neurons to PVN. Moreover, GLP-1 evoked Ca(2+) signaling in single neurons isolated from PVN. The majority of GLP-1-responsive neurons were immunoreactive predominantly to corticotropin-releasing hormone (CRH) and nesfatin-1, and less frequently to oxytocin. These results indicate that endogenous GLP-1 targets PVN to restrict feeding behavior, in which the projection from NTS GLP-1 neurons and activation of CRH and nesfatin-1 neurons might be implicated. This study reveals a neuronal basis for the anorexigenic effect of endogenous GLP-1 in the brain.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Hormona Liberadora de Corticotropina/fisiología , Proteínas de Unión al ADN/fisiología , Ingestión de Alimentos/fisiología , Péptido 1 Similar al Glucagón/fisiología , Proteínas del Tejido Nervioso/fisiología , Oxitocina/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Núcleo Solitario/fisiología , Animales , Señalización del Calcio , Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Receptor del Péptido 1 Similar al Glucagón , Humanos , Masculino , Microinyecciones , Vías Nerviosas/fisiología , Neuronas/fisiología , Nucleobindinas , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Fragmentos de Péptidos/administración & dosificación , Ratas , Ratas Wistar , Receptores de Glucagón/antagonistas & inhibidores
13.
J Nutr Sci Vitaminol (Tokyo) ; 70(3): 193-202, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38945884

RESUMEN

Oleuropein aglycone (OA), which is the absorbed form of oleuropein, is a major phenolic compound in extra virgin olive oil. We analyzed the anti-obesity effect of OA intake combined with mild treadmill walking (MTW, 4 m/min for 20 min/d, 5-6 d/wk, without electric shocks and slope) in rats under a high-fat diet (HF). Four-week-old male Sprague-Dawley rats (n=28) were equally divided into four groups: control (HF), 0.08% oleuropein-supplemented HF (HFO), HF with MTW (HF+W), and HFO with MTW (HFO+W) groups. After 28 d, the inguinal subcutaneous fat content and weight gain were significantly lower in the HFO+W group than in the control group. The HFO+W group also had significantly higher levels of urinary noradrenaline secretion, interscapular brown adipose tissue, uncoupling protein 1, brain transient receptor potential ankyrin subtype 1 (TRPA1), vanilloid subtype 1 (TRPV1), and brain-derived neurotrophic factor (BDNF) than the control group. Especially, the HFO+W group showed a synergistic effect on noradrenaline secretion. Therefore, OA combined with MTW may accelerate the enhancement of UCP1 and BDNF levels in rats with HF-induced obesity by increasing noradrenaline secretion after TRPA1 and TRPV1 activation.


Asunto(s)
Tejido Adiposo Pardo , Factor Neurotrófico Derivado del Encéfalo , Dieta Alta en Grasa , Glucósidos Iridoides , Iridoides , Norepinefrina , Obesidad , Ratas Sprague-Dawley , Canal Catiónico TRPA1 , Proteína Desacopladora 1 , Animales , Masculino , Proteína Desacopladora 1/metabolismo , Glucósidos Iridoides/farmacología , Obesidad/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Iridoides/farmacología , Norepinefrina/metabolismo , Canal Catiónico TRPA1/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Ratas , Fármacos Antiobesidad/farmacología , Caminata , Aumento de Peso/efectos de los fármacos , Condicionamiento Físico Animal , Canales Catiónicos TRPV
14.
Nutrients ; 16(14)2024 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-39064713

RESUMEN

(1) Background: Proglucagon-derived peptides (PDGPs) including glucagon (Gcg), GLP-1, and GLP-2 regulate lipid metabolism in the liver, adipocytes, and intestine. However, the mechanism by which PGDPs participate in alterations in lipid metabolism induced by high-fat diet (HFD) feeding has not been elucidated. (2) Methods: Mice deficient in PGDP (GCGKO) and control mice were fed HFD for 7 days and analyzed, and differences in lipid metabolism in the liver, adipose tissue, and duodenum were investigated. (3) Results: GCGKO mice under HFD showed lower expression levels of the genes involved in free fatty acid (FFA) oxidation such as Hsl, Atgl, Cpt1a, Acox1 (p < 0.05), and Pparα (p = 0.05) mRNA in the liver than in control mice, and both FFA and triglycerides content in liver and adipose tissue weight were lower in the GCGKO mice. On the other hand, phosphorylation of hormone-sensitive lipase (HSL) in white adipose tissue did not differ between the two groups. GCGKO mice under HFD exhibited lower expression levels of Pparα and Cd36 mRNA in the duodenum as well as increased fecal cholesterol contents compared to HFD-controls. (4) Conclusions: GCGKO mice fed HFD exhibit a lesser increase in hepatic FFA and triglyceride contents and adipose tissue weight, despite reduced ß-oxidation in the liver, than in control mice. Thus, the absence of PGDP prevents dietary-induced fatty liver development due to decreased lipid uptake in the intestinal tract.


Asunto(s)
Antígenos CD36 , Dieta Alta en Grasa , Absorción Intestinal , Metabolismo de los Lípidos , Hígado , Ratones Noqueados , PPAR alfa , Proglucagón , Animales , Masculino , Dieta Alta en Grasa/efectos adversos , PPAR alfa/metabolismo , PPAR alfa/genética , Hígado/metabolismo , Proglucagón/metabolismo , Proglucagón/genética , Antígenos CD36/metabolismo , Antígenos CD36/genética , Ratones , Esterol Esterasa/metabolismo , Esterol Esterasa/genética , Triglicéridos/metabolismo , Ratones Endogámicos C57BL , Ácidos Grasos no Esterificados/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Duodeno/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Tejido Adiposo/metabolismo , Grasas de la Dieta , Péptido 2 Similar al Glucagón/metabolismo , Aciltransferasas , Lipasa
15.
J Pediatr Endocrinol Metab ; 26(3-4): 343-6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23348218

RESUMEN

Recently, a case of newborn infant with transient hyperinsulinism has been reported. This infant was reported to be free from typical perinatal risk factors of hyperinsulinism except for the fact that the mother of the baby was receiving the antidepressant bupropion during her pregnancy. However, the mother did not experience hyperinsulinism and, so far, there are no reports about the pharmacological mechanism of bupropion causing hyperinsulinemia. In this study, bupropion was shown to inhibit KATP channel activity in pancreatic ß-cell membranes and induce insulin secretion in relatively high concentration. This study shows, for the first time, that bupropion has a direct electrophysiological action on pancreatic ß-cells and can cause insulin secretion and also highlights the risk of using bupropion during pregnancy.


Asunto(s)
Bupropión/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Activación del Canal Iónico/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/metabolismo , Animales , Antidepresivos de Segunda Generación/efectos adversos , Antidepresivos de Segunda Generación/farmacología , Bupropión/efectos adversos , Femenino , Humanos , Hiperinsulinismo/inducido químicamente , Hiperinsulinismo/metabolismo , Recién Nacido , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Bloqueadores de los Canales de Potasio/efectos adversos , Embarazo
16.
J Diabetes Investig ; 14(6): 746-755, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36977210

RESUMEN

AIMS/INTRODUCTION: Imeglimin is a new antidiabetic drug structurally related to metformin. Despite this structural similarity, only imeglimin augments glucose-stimulated insulin secretion (GSIS), with the mechanism underlying this effect remaining unclear. Given that glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) also enhance GSIS, we examined whether these incretin hormones might contribute to the pharmacological actions of imeglimin. MATERIALS AND METHODS: Blood glucose and plasma insulin, GIP, and GLP-1 concentrations were measured during an oral glucose tolerance test (OGTT) performed in C57BL/6JJcl (C57BL/6) or KK-Ay/TaJcl (KK-Ay) mice after administration of a single dose of imeglimin with or without the dipeptidyl peptidase-4 inhibitor sitagliptin or the GLP-1 receptor antagonist exendin-9. The effects of imeglimin, with or without GIP or GLP-1, on GSIS were examined in C57BL/6 mouse islets. RESULTS: Imeglimin lowered blood glucose and increased plasma insulin levels during an OGTT in both C57BL/6 and KK-Ay mice, whereas it also increased the plasma levels of GIP and GLP-1 in KK-Ay mice and the GLP-1 levels in C57BL/6 mice. The combination of imeglimin and sitagliptin increased plasma insulin and GLP-1 levels during the OGTT in KK-Ay mice to a markedly greater extent than did either drug alone. Imeglimin enhanced GSIS in an additive manner with GLP-1, but not with GIP, in mouse islets. Exendin-9 had only a minor inhibitory effect on the glucose-lowering action of imeglimin during the OGTT in KK-Ay mice. CONCLUSIONS: Our data suggest that the imeglimin-induced increase in plasma GLP-1 levels likely contributes at least in part to its stimulatory effect on insulin secretion.


Asunto(s)
Glucemia , Incretinas , Animales , Ratones , Incretinas/farmacología , Insulina , Ratones Endogámicos C57BL , Fosfato de Sitagliptina/farmacología , Hipoglucemiantes/farmacología , Glucosa/farmacología , Péptido 1 Similar al Glucagón , Polipéptido Inhibidor Gástrico
17.
Endocrinology ; 164(6)2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-37139968

RESUMEN

Protein intake potently increases body temperature and energy expenditure, but the underlying mechanism thereof remains incompletely understood. Simultaneously, protein intake potently stimulates glucagon-like peptide-1 (GLP-1) secretion. Here, we examined the involvement of GLP-1 in the thermic effects of dietary proteins in rodents by measuring rectal temperature and energy expenditure and modulating GLP-1 signaling. Rectal temperature of rats or mice fasted for 4 or 5 hours were measured using a thermocouple thermometer before and after an oral administration of nutrients. Oxygen consumption after oral protein administration was also measured in rats. Rectal temperature measurements in rats confirmed an increase in core body temperature after refeeding, and the thermic effect of the oral administration of protein was greater than that of a representative carbohydrate or lipid. Among the five dietary proteins examined (casein, whey, rice, egg, and soy), soy protein had the highest thermic effect. The thermic effect of soy protein was also demonstrated by increased oxygen consumption. Studies using a nonselective ß-adrenergic receptor antagonist and thermal camera suggested that brown adipose tissue did not contribute to soy protein-induced increase in rectal temperature. Furthermore, the thermic effect of soy protein was completely abolished by antagonism and knockout of the GLP-1 receptor, yet potentiated via augmentation of intact GLP-1 levels through inhibition of dipeptidyl peptidase-4 activity. These results indicate that GLP-1 signaling is essential for the thermic effects of dietary proteins in rats and mice, and extend the metabolic actions of GLP-1 ensuing from nutrient ingestion to encompass the thermic response to ingested protein.


Asunto(s)
Péptido 1 Similar al Glucagón , Roedores , Ratas , Ratones , Masculino , Animales , Péptido 1 Similar al Glucagón/metabolismo , Roedores/metabolismo , Proteínas de Soja/farmacología , Proteínas en la Dieta , Ingestión de Alimentos/fisiología , Receptor del Péptido 1 Similar al Glucagón , Péptido 2 Similar al Glucagón/farmacología
18.
J Physiol Sci ; 72(1): 18, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35915429

RESUMEN

Hypothermia has been observed during hypergravity load in mice and rats. This response is beneficial for maintaining blood glucose level, although food intake decreases. However, saving glucose is not enough to maintain blood glucose level during hypergravity load. In this study, we examined the contribution of humoral factors related to glycolysis in maintaining blood glucose level in a 2 G environment. Increased plasma corticosterone levels were observed in mice with intact peripheral vestibular organs, but not in mice with vestibular lesions. Plasma glucagon levels did not change, and decrease in plasma adrenaline levels was observed in mice with intact peripheral vestibular organs. Accordingly, it is possible that increase in plasma corticosterone level and hypothermia contribute to prevent hypoglycemia in a 2 G environment.


Asunto(s)
Hiperglucemia , Hipergravedad , Hipotermia , Animales , Glucemia , Corticosterona , Hipergravedad/efectos adversos , Ratones , Ratas
19.
Nutrients ; 14(12)2022 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-35745222

RESUMEN

Gamma-aminobutyric acid (GABA) is present in the mammalian brain as the main inhibitory neurotransmitter and in foods. It is widely used as a supplement that regulates brain function through stress-reducing and sleep-enhancing effects. However, its underlying mechanisms remain poorly understood, as it is reportedly unable to cross the blood-brain barrier. Here, we explored whether a single peroral administration of GABA affects feeding behavior as an evaluation of brain function and the involvement of vagal afferent nerves. Peroral GABA at 20 and 200 mg/kg immediately before refeeding suppressed short-term food intake without aversive behaviors in mice. However, GABA administration 30 min before refeeding demonstrated no effects. A rise in circulating GABA concentrations by the peroral administration of 200 mg/kg GABA was similar to that by the intraperitoneal injection of 20 mg/kg GABA, which did not alter feeding. The feeding suppression by peroral GABA was blunted by the denervation of vagal afferents. Unexpectedly, peroral GABA alone did not alter vagal afferent activities histologically. The coadministration of a liquid diet and GABA potentiated the postprandial activation of vagal afferents, thereby enhancing postprandial satiation. In conclusion, dietary GABA activates vagal afferents in collaboration with meals or meal-evoked factors and regulates brain function including feeding behavior.


Asunto(s)
Ingestión de Alimentos , Saciedad , Animales , Dieta , Ingestión de Alimentos/fisiología , Mamíferos , Ratones , Saciedad/fisiología , Nervio Vago/fisiología , Ácido gamma-Aminobutírico/farmacología
20.
Biosci Biotechnol Biochem ; 75(5): 904-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21597186

RESUMEN

The administration of such a transient receptor potential vanilloid 1 (TRPV1) agonist as capsaicin, which is a pungent ingredient of red pepper, promotes energy metabolism and suppresses visceral fat accumulation. We have recently identified monoacylglycerols (MGs) having an unsaturated long-chain fatty acid as the novel TRPV1 agonist in foods. We investigated in this present study the effects of dietary MGs on uncoupling protein 1 (UCP1) expression in interscapular brown adipose tissue (IBAT) and on fat accumulation in mice fed with a high-fat, high-sucrose diet. The MG30 diet that substituted 30% of all lipids for MGs (a mixture of 1-oleoylglycerol, 1-linoleoylglycerol and 1-linolenoylglycerol) significantly increased the UCP1 content of IBAT and decreased the weight of epididymal white adipose tissue, and the serum glucose, total cholesterol and free fatty acid levels. The diet containing only 1-oleoylglycerol as MG also increased UCP1 expression in IBAT. MGs that activated TRPV1 also therefore induced the expression of UCP 1 and prevented visceral fat accumulation as well as capsaicin.


Asunto(s)
Adipocitos Marrones/efectos de los fármacos , Carbohidratos de la Dieta/efectos adversos , Grasas de la Dieta/efectos adversos , Grasa Intraabdominal/efectos de los fármacos , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Monoglicéridos/farmacología , Canales Catiónicos TRPV/agonistas , Adipocitos Marrones/metabolismo , Animales , Hipercolesterolemia/etiología , Hipercolesterolemia/prevención & control , Hiperglucemia/etiología , Hiperglucemia/prevención & control , Grasa Intraabdominal/metabolismo , Masculino , Ratones , Monoglicéridos/química , Sacarosa/efectos adversos , Proteína Desacopladora 1 , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA