Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
Más filtros

Banco de datos
Tipo del documento
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.
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
3.
Int J Mol Sci ; 24(9)2023 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-37175953

RESUMEN

1,5-Anhydro-D-fructose (1,5-AF) is a bioactive monosaccharide that is produced by the glycogenolysis in mammalians and is metabolized to 1,5-anhydro-D-glucitol (1,5-AG). 1,5-AG is used as a marker of glycemic control in diabetes patients. 1,5-AF has a variety of physiological activities, but its effects on energy metabolism, including feeding behavior, are unclarified. The present study examined whether 1,5-AF possesses the effect of satiety. Peroral administration of 1,5-AF, and not of 1,5-AG, suppressed daily food intake. Intracerebroventricular (ICV) administration of 1,5-AF also suppressed feeding. To investigate the neurons targeted by 1,5-AF, we investigated c-Fos expression in the hypothalamus and brain stem. ICV injection of 1,5-AF significantly increased c-Fos positive oxytocin neurons and mRNA expression of oxytocin in the paraventricular nucleus (PVN). Moreover, 1,5-AF increased cytosolic Ca2+ concentration of oxytocin neurons in the PVN. Furthermore, the satiety effect of 1,5-AF was abolished in oxytocin knockout mice. These findings reveal that 1,5-AF activates PVN oxytocin neurons to suppress feeding, indicating its potential as the energy storage monitoring messenger to the hypothalamus for integrative regulation of energy metabolism.


Asunto(s)
Oxitocina , Núcleo Hipotalámico Paraventricular , Ratones , Animales , Núcleo Hipotalámico Paraventricular/metabolismo , Oxitocina/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Mamíferos/metabolismo
4.
Biochem Biophys Res Commun ; 613: 159-165, 2022 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-35561584

RESUMEN

A rare sugar D-Allulose has sweetness without calorie. Previous studies have shown that D-Allulose improves glucose and energy metabolism and ameliorates obesity. However, underlying mechanisms remain elusive. This study explored the effect of central injection of D-Allulose on feeding behavior in mice. We also examined direct effects of D-Allulose on the neurons in the hypothalamic arcuate nucleus (ARC) that regulate feeding, including the anorexigenic glucagon-like peptide-1 (GLP-1)-responsive neurons and proopiomelanocortin (POMC) neurons. Single neurons were isolated from ARC and cytosolic Ca2+ concentration ([Ca2+]i) was measured by fura-2 microfluorometry. Administration of D-Allulose at 5.6, 16.7 and 56 mM concentration-dependently increased [Ca2+]i in ARC neurons. The [Ca2+]i increases took place similarly when the osmolarity of superfusion solution was kept constant. The majority (40%) of the D-Allulose-responsive neurons also responded to GLP-1 with [Ca2+]i increases. D-Allulose increased [Ca2+]i in 33% of POMC neurons in ARC. D-Allulose potentiated the GLP-1 action to increase [Ca2+]i in ARC neurons including POMC neurons. Intracerebroventricular injection of D-Allulose significantly decreased food intake at 1 and 2 h after injection. These results demonstrate that D-Allulose cooperates with glucagon-like peptide-1 and activates the ARC neurons including POMC neurons. Furthermore, central injection of D-Allulose inhibits feeding. These central actions of D-Allulose may underlie the ability of D-Allulose to counteract obesity and diabetes.


Asunto(s)
Núcleo Arqueado del Hipotálamo , Proopiomelanocortina , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Fructosa , Péptido 1 Similar al Glucagón/metabolismo , Ratones , Neuronas/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Proopiomelanocortina/metabolismo
5.
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
6.
Gen Comp Endocrinol ; 271: 49-60, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30391240

RESUMEN

Recent studies have revealed nesfatin-1 as a hypothalamic neuropeptide, regulating food intake, energy expenditure and reproduction primarily by acting on the hypothalamic-pituitary-gonadal axis. Nesfatin-1 is also localized in several peripheral tissues including testes. However, functional significance of nesfatin-1 in testicular activities is not yet well documented in mammals. Therefore, this study was aimed to elucidate the direct effects of nesfatin-1 on testicular markers for steroid productions, spermatogenesis, metabolic changes and oxidative stress. The results revealed the expression of both protein and mRNA of nesfatin-1 in the testes of adult mice. The testes treated in vitro with nesfatin-1 showed significant increase in testosterone production, which correlated significantly with increased expression of steroidogenic markers and insulin receptor proteins in the testes. Furthermore, the in vitro treatment with nesfatin-1 showed stimulatory effects on spermatogenesis by promoting cell proliferation (PCNA) and survival (Bcl2), while inhibiting apoptosis (caspase-3) in the testes. The nesfatin-1 treatment in vitro further increased the expression of insulin receptor and GLUT8 proteins, in parallel with increase in the intra-testicular transport of glucose and production of lactate. This nesfatin-1 induced enhanced transport of energy substrate (glucose and lactate) may be responsible for promoting spermatogenesis and steroidogenesis. Nesfatin-1 significantly reduced oxidative stress and nitric oxide, which may also be responsible for stimulatory effects on testicular activities. The present finding suggests that nesfatin-1 acts via paracrine manner to increase sperm count and fertility, thus promoting the testicular function.


Asunto(s)
Envejecimiento/metabolismo , Proteínas de Unión al Calcio/farmacología , Proteínas de Unión al ADN/farmacología , Proteínas del Tejido Nervioso/farmacología , Neuropéptidos/farmacología , Espermatogénesis , Esteroides/biosíntesis , Testículo/metabolismo , Animales , Antioxidantes/metabolismo , Biomarcadores/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Estradiol/metabolismo , Masculino , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Nucleobindinas , Estrés Oxidativo/efectos de los fármacos , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Espermatogénesis/efectos de los fármacos , Espermatozoides/efectos de los fármacos , Espermatozoides/metabolismo , Testículo/efectos de los fármacos , Testosterona/metabolismo
7.
Am J Physiol Endocrinol Metab ; 315(4): E478-E488, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28174180

RESUMEN

Light synchronizes the body's circadian rhythms by modulating the master clock located in the suprachiasmatic nucleus (SCN) of the hypothalamus. In modern lifestyles that run counter to normal circadian rhythms, the extended and/or irregular light exposure impairs circadian rhythms and, consequently, promotes feeding and metabolic disorders. However, the neuronal pathway through which light is coupled to feeding behavior is less elucidated. The present study employed the light exposure during the dark phase of the day in rats and observed its effect on neuronal activity and feeding behavior. Light exposure acutely suppressed food intake and elevated c-Fos expression in the AVP neurons of SCN and the oxytocin (Oxt) neurons of paraventricular nucleus (PVN) in the hypothalamus. The light-induced suppression of food intake was abolished by blockade of the Oxt receptor in the brain. Retrograde tracer analysis demonstrated the projection of SCN AVP neurons to the PVN. Furthermore, intracerebroventricular injection of AVP suppressed food intake and increased c-Fos in PVN Oxt neurons. Intra-PVN injection of AVP exerted a stronger anorexigenic effect than intracerebroventriclar injection. AVP also induced intracellular Ca2+ signaling and increased firing frequency in Oxt neurons in PVN slices. These results reveal the novel neurocircuit from SCN AVP to PVN Oxt that relays light reception to inhibition of feeding behavior. This light-induced neurocircuit may serve as a pathway for forming the circadian feeding rhythm and linking irregular light exposure to arrhythmic feeding and, consequently, obesity and metabolic diseases.


Asunto(s)
Arginina Vasopresina/metabolismo , Conducta Alimentaria/fisiología , Luz , Oxitocina/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Núcleo Supraquiasmático/metabolismo , Animales , Arginina Vasopresina/farmacología , Arginina Vasopresina/fisiología , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Ritmo Circadiano/fisiología , Conducta Alimentaria/efectos de los fármacos , Hipotálamo/metabolismo , Hipotálamo/fisiología , Inhibición Psicológica , Masculino , Vías Nerviosas , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxitocina/fisiología , Núcleo Hipotalámico Paraventricular/fisiología , Proteínas Proto-Oncogénicas c-fos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Receptores de Oxitocina , Núcleo Supraquiasmático/fisiología
8.
Biochem Biophys Res Commun ; 500(4): 910-916, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29705701

RESUMEN

In the hypothalamic arcuate nucleus (ARC), orexigenic agouti-related peptide (AgRP) neurons regulate feeding behavior and energy homeostasis. The 3-phosphoinositide-dependent protein kinase-1 (PDK1) in AgRP neurons serves as a major signaling molecule for leptin and insulin, the hormones regulating feeding behavior, energy homeostasis and circulation. However, it is unclear whether PDK1 in AGRP neurons is also involved in regulation of blood pressure. This study explored it by generating and analyzing AgRP neuron-specific PDK1 knockout (Agrp-Pdk1flox/flox) mice and effect of high salt diet on blood pressure in KO and WT mice was analyzed. Under high salt diet feeding, systolic blood pressure (SBP) of Agrp-Pdk1flox/flox mice was significantly elevated compared to Agrp-Cre mice. When the high salt diet was switched to control low salt diet, SBP of Agrp-Pdk1flox/flox mice returned to the basal level observed in Agrp-Cre mice within 1 week. In Agrp-Pdk1flox/flox mice, urinary noradrenalin excretion and NUCB2 mRNA expression in hypothalamic paraventricular nucleus (PVN) were markedly upregulated. Moreover, silencing of NUCB2 in the PVN counteracted the rises in urinary noradrenalin excretions and SBP. These results demonstrate a novel role of PDK1 in AgRP neurons to counteract the high salt diet-induced hypertension by preventing hyperactivation of PVN nesfatin-1 neurons.


Asunto(s)
Proteína Relacionada con Agouti/genética , Núcleo Arqueado del Hipotálamo/metabolismo , Hipertensión/genética , Neuronas/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteína Relacionada con Agouti/deficiencia , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/fisiopatología , Presión Sanguínea , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Ingestión de Energía/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Regulación de la Expresión Génica , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Hipertensión/fisiopatología , Insulina/genética , Insulina/metabolismo , Leptina/genética , Leptina/metabolismo , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Norepinefrina/orina , Nucleobindinas , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/fisiopatología , Proteínas Serina-Treonina Quinasas/deficiencia , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Cloruro de Sodio Dietético/efectos adversos
9.
Biochem Biophys Res Commun ; 499(3): 618-625, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29601817

RESUMEN

Endogenous GLP-1 and GLP-1 receptor agonists (GLP-1RAs) regulate glucose metabolism via common and distinct mechanisms. Postprandial release of GLP-1 is modest and it is degraded by DPP-4 within 2 min, and hence it cannot enter the brain in substantial amount. In contrast, DPP-4-resistant GLP-1RAs are administered at 10 times higher concentration than endogenous GLP-1 level, which enables them to reach several brain regions including ARC and AP, the areas implicated in glucose metabolism. Hence, some of the effects of GLP-1RAs observed clinically and experimentally, including pancreatic ß-cell proliferation, are thought to involve the brain. However, the effects of centrally acting GLP-1/GLP-1RAs on glucose metabolism and underlying neural mechanism are unclear. This study aimed to establish the link of central GLP-1/GLP-1RA action to pancreatic ß-cell proliferation. Both subcutaneous (SC) and intracerebroventricular (ICV) injections of liraglutide increased the number of pancreatic ß-cells expressing Ki67 and PCNA, proliferation markers, in C57BL/6J mice. This effect was induced by single ICV administration of liraglutide at relatively low dose that was incapable of suppressing food intake. These SC and ICV liraglutide-induced effects were inhibited by 50% and 70%, respectively, by pretreatment with atropine, a muscarinic receptor blocker. ICV liraglutide induced c-Fos expression in the area postrema (AP), nucleus tractus solitaries (NTS), and dorsal motor nucleus of the vagus (DMX) of the brain stem. These results demonstrate that central action of liraglutide induces pancreatic ß-cell proliferation via the pathway involving the brain stem AP/NTS/DMX area and vagus nerve. This route is highly sensitive to GLP-1/GLP-1RA. Hence, this brain-pancreatic ß-cell pathway may operate in type 2 diabetic patients treated with GLP-RAs and serve to counteract the reduction of ß-cell mass.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/agonistas , Células Secretoras de Insulina/citología , Liraglutida/farmacología , Bulbo Raquídeo/metabolismo , Nervio Vago/metabolismo , Animales , Atropina/farmacología , Tronco Encefálico/efectos de los fármacos , Tronco Encefálico/metabolismo , Proliferación Celular/efectos de los fármacos , Conducta Alimentaria , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Inyecciones Subcutáneas , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Liraglutida/administración & dosificación , Masculino , Bulbo Raquídeo/efectos de los fármacos , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-fos/metabolismo , Nervio Vago/efectos de los fármacos
10.
J Pharmacol Sci ; 137(1): 67-75, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29716811

RESUMEN

Oxytocin (OT) is a 9-amine neuropeptide that plays an essential role in mammalian labor, lactation, maternal bonding, and social affiliation. OT has been reported to exert an analgesic effect in both humans and animals, and the results of certain animal experiments have shown that the analgesic effect of OT is partially blocked by opioid receptor antagonists. To investigate the relationship between OT and µ opioid receptor (MOR), we evaluated how OT affects MOR in vitro by performing an electrical impedance-based receptor biosensor assay (CellKey™ assay), an intracellular cAMP assay, and a competitive receptor-binding analysis by using cells stably expressing human MOR and OT receptor. In both the CellKey™ assay and the intracellular cAMP assay, OT alone exerted no direct agonistic effect on human MOR, but treatment with 10-6 M OT markedly enhanced the MOR signaling induced by 10-6 M endomorphin-1, ß-endorphin, morphine, fentanyl, and DAMGO. Moreover, in the competitive receptor-binding assay, 10-6 M OT did not alter the affinity of endomorphin-1 or morphine for MOR. These results suggest that OT could function as a positive allosteric modulator that regulates the efficacy of MOR signaling, and thus OT might represent a previously unrecognized candidate analgesic agent.


Asunto(s)
Regulación Alostérica/efectos de los fármacos , Neuropéptidos/farmacología , Oxitocina/farmacología , Receptores Opioides mu/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Analgésicos , Animales , AMP Cíclico/metabolismo , Células HEK293 , Humanos , Oxitocina/fisiología , Receptores Opioides mu/fisiología , Estimulación Química
11.
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
12.
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
13.
J Immunol ; 192(9): 4342-51, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24696236

RESUMEN

Inflammation plays a key role in the pathophysiology of hepatic ischemia-reperfusion (I/R) injury. However, the mechanism by which hepatic I/R induces inflammatory responses remains unclear. Recent evidence indicates that a sterile inflammatory response triggered by I/R is mediated through a multiple-protein complex called the inflammasome. Therefore, we investigated the role of the inflammasome in hepatic I/R injury and found that hepatic I/R stimuli upregulated the inflammasome-component molecule, nucleotide-binding oligomerization domain-like receptor (NLR) family pyrin domain-containing 3 (NLRP3), but not apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC). NLRP3(-/-) mice, but not ASC(-/-) and caspase-1(-/-) mice, had significantly less liver injury after hepatic I/R. NLRP3(-/-) mice showed reduced inflammatory responses, reactive oxygen species production, and apoptosis in I/R liver. Notably, infiltration of neutrophils, but not macrophages, was markedly inhibited in the I/R liver of NLRP3(-/-) mice. Bone marrow transplantation experiments showed that NLRP3 not only in bone marrow-derived cells, but also in non-bone marrow-derived cells contributed to liver injury after I/R. In vitro experiments revealed that keratinocyte-derived chemokine-induced activation of heterotrimeric G proteins was markedly diminished. Furthermore, NLRP3(-/-) neutrophils decreased keratinocyte-derived chemokine-induced concentrations of intracellular calcium elevation, Rac activation, and actin assembly formation, thereby resulting in impaired migration activity. Taken together, NLRP3 regulates chemokine-mediated functions and recruitment of neutrophils, and thereby contributes to hepatic I/R injury independently of inflammasomes. These findings identify a novel role of NLRP3 in the pathophysiology of hepatic I/R injury.


Asunto(s)
Proteínas Portadoras/inmunología , Hígado/inmunología , Neutrófilos/inmunología , Daño por Reperfusión/inmunología , Animales , Apoptosis/inmunología , Western Blotting , Proteínas Portadoras/metabolismo , Quimiotaxis de Leucocito , Citometría de Flujo , Inmunohistoquímica , Inflamasomas/inmunología , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Neutrófilos/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Daño por Reperfusión/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Endocr J ; 63(7): 611-9, 2016 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-27097546

RESUMEN

Regeneration of pancreatic ß-cell mass benefits both type 1 and type 2 diabetic patients. A recent study identified betatrophin as a ß-cell proliferation factor. However, the expressional regulation of betatrophin remains less defined. In this study, we aimed to clarify the regulation of betatrophin expression in obese type 2 vs. type 1 diabetes model animals. We experimented type 2 diabetes models, diet-induced-obesity (DIO) mice and db/db mice, and type 1 diabetes models, C57B6 mice receiving streptozotocin (STZ) or 70% pancreatectomy to destroy or remove ß-cells. Serum betatrophin levels and betatrophin mRNA expressions in the liver, white adipose tissue (WAT) and brown adipose tissue (BAT) were measured. In DIO mice and db/db mice, serum betatrophin and betatrophin mRNA expressions in the liver, WAT and BAT were elevated in parallel with increases in body weight and plasma insulin. These elevated betatrophin mRNA expressions were not altered by treatment with SGLT2 inhibitor that ameliorated hyperglycemia. In pancreatectomized mice, betatrophin expression in WAT decreased in parallel with reductions in weight and insulin. In STZ-treated mice, betatrophin expressions in the liver, WAT and BAT were reduced. However, when the mouse liver slices were cultured with STZ, betatrophin expression was significantly reduced, indicating a direct action of STZ on the liver. These results indicate that the expression of betatrophin is upregulated in the liver, WAT and BAT in obese hyperinsulinemic type 2 diabetes but decreased in WAT in hypoinsulinemic type 1 diabetes, suggesting its positive correlation with body weight and plasma insulin but not blood glucose.


Asunto(s)
Diabetes Mellitus Experimental/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 2/genética , Hiperinsulinismo/genética , Obesidad/genética , Hormonas Peptídicas/genética , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Proteína 8 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Animales , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/patología , Hiperinsulinismo/complicaciones , Hiperinsulinismo/patología , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/complicaciones , Obesidad/patología , Hormonas Peptídicas/metabolismo
15.
Endocr J ; 63(10): 867-876, 2016 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-27321586

RESUMEN

In pancreatic ß-cells, glucose-induced closure of the ATP-sensitive K+ (KATP) channel is an initial process triggering glucose-stimulated insulin secretion (GSIS). This KATP-channel dependent pathway has been believed to be a central mechanism for GSIS. However, since the resting membrane potential of cells is determined by the balance of the net result of current amplitudes in outward and inward directions, it must be taken into consideration that not only KATP channel inhibition but also inward current via the basal opening of non-selective cation channels (NSCCs) plays a crucial role in membrane potential regulation. The basal activity of NSCCs is essential to effectively evoke depolarization in concert with KATP channel closure that is dependent on glucose metabolism. The present study summarizes recent findings regarding the roles of NSCCs in GSIS and GTP-binding protein coupled receptor-(GPCR) operated potentiation of GSIS.


Asunto(s)
Proteínas de Unión al GTP/fisiología , Glucosa/fisiología , Insulina/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Canales de Potencial de Receptor Transitorio/metabolismo , Animales , Proteínas de Unión al GTP/metabolismo , Glucosa/metabolismo , Humanos , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Transducción de Señal , Canales de Potencial de Receptor Transitorio/fisiología
16.
Am J Physiol Endocrinol Metab ; 309(4): E320-33, 2015 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-26081283

RESUMEN

Feeding is regulated by perception in the hypothalamus, particularly the first-order arcuate nucleus (ARC) neurons, of the body's energy state. However, the cellular device for converting energy states to the activity of critical neurons in ARC is less defined. We here show that Na(+),K(+)-ATPase (NKA) in ARC senses energy states to regulate feeding. Fasting-induced systemic ghrelin rise and glucose lowering reduced ATP-hydrolyzing activity of NKA and its substrate ATP level, respectively, preferentially in ARC. Lowering glucose concentration (LG), which mimics fasting, decreased intracellular NAD(P)H and increased Na(+) concentration in single ARC neurons that subsequently exhibited [Ca(2+)]i responses to LG, showing that they were glucose-inhibited (GI) neurons. Third ventricular injection of the NKA inhibitor ouabain induced c-Fos expression in agouti-related protein (AgRP) neurons in ARC and evoked neuropeptide Y (NPY)-dependent feeding. When injected focally into ARC, ouabain stimulated feeding and mRNA expressions for NPY and AgRP. Ouabain increased [Ca(2+)]i in single NPY/AgRP neurons with greater amplitude than in proopiomelanocortin neurons in ARC. Conversely, the specific NKA activator SSA412 suppressed fasting-induced feeding and LG-induced [Ca(2+)]i increases in ARC GI neurons. NPY/AgRP neurons highly expressed NKAα3, whose knockdown impaired feeding behavior. These results demonstrate that fasting, via ghrelin rise and LG, suppresses NKA enzyme/pump activity in ARC and thereby promotes the activation of GI neurons and NPY/AgRP-dependent feeding. This study identifies ARC NKA as a hypothalamic sensor and converter of metabolic states to key neuronal activity and feeding behaviour, providing a new target to treat hyperphagic obesity and diabetes.


Asunto(s)
Núcleo Arqueado del Hipotálamo/metabolismo , Metabolismo Energético/genética , Conducta Alimentaria/fisiología , Glucosa/farmacología , Neuronas/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/fisiología , Adenosina Trifosfato/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Conducta Animal/fisiología , Masculino , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Ratas , Ratas Sprague-Dawley , Ratas Transgénicas , ATPasa Intercambiadora de Sodio-Potasio/genética
17.
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
18.
Biochem Biophys Res Commun ; 456(4): 913-8, 2015 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-25534851

RESUMEN

An adipokine leptin plays a central role in the regulation of feeding and energy homeostasis via acting on the hypothalamus. However, its downstream neuronal mechanism is not thoroughly understood. The neurons expressing nucleobindin-2 (NUCB2)-derived nesfatin-1 in the hypothalamic paraventricular nucleus (PVN) have been implicated in feeding and energy homeostasis. The present study aimed to explore the role of PVN NUCB2/nesfatin-1 in the leptin action, by using adeno-associated virus (AAV) vectors encoding shRNA targeting NUCB2 (AAV-NUCB2-shRNA). Leptin directly interacted and increased cytosolic Ca(2+) in single neurons isolated from the PVN, predominantly in NUCB2/nesftin-1-immunoreactive neurons. Treatment with leptin in vivo and in vitro markedly increased NUCB2 mRNA expression in the PVN. Peripheral and central injections of leptin failed to significantly inhibit food intake in mice receiving AAV-NUCB2. These results indicate that PVN NUCB2/nesfatin-1 is directly targeted by leptin, and mediates its anorexigenic effect.


Asunto(s)
Anorexia/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al ADN/metabolismo , Leptina/farmacología , Proteínas del Tejido Nervioso/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Animales , Anorexia/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al ADN/genética , Dependovirus/metabolismo , Metabolismo Energético/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Homeostasis/efectos de los fármacos , Leptina/administración & dosificación , Masculino , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Nucleobindinas , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA