Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Nature ; 585(7826): 591-596, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32526765

RESUMO

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.


Assuntos
Encéfalo/citologia , Intestinos/citologia , Intestinos/inervação , Fígado/citologia , Fígado/inervação , Neurônios/fisiologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Vias Aferentes , Animais , Células Apresentadoras de Antígenos/imunologia , Colite/imunologia , Colite/metabolismo , Colite/patologia , Homeostase , Humanos , Intestinos/imunologia , Masculino , Camundongos , Ratos , Receptores Muscarínicos/metabolismo , Baço/citologia , Baço/imunologia , Nervo Vago/fisiologia
2.
J Physiol ; 601(12): 2293-2306, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37126218

RESUMO

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.


Assuntos
Glicemia , Glicogenólise , Camundongos , Animais , Glucagon , Corticosterona/farmacologia , Dióxido de Carbono , Neurônios/fisiologia , Bulbo/fisiologia , Catecolaminas
3.
Am J Physiol Regul Integr Comp Physiol ; 324(2): R152-R160, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36534584

RESUMO

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.


Assuntos
Lesão Pulmonar Aguda , Vestíbulo do Labirinto , Camundongos , Animais , Neurônios/fisiologia , Bulbo/fisiologia , Lesão Pulmonar Aguda/prevenção & controle , Anti-Inflamatórios , Estimulação Elétrica
4.
Brain Behav Immun ; 111: 138-150, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37037362

RESUMO

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.


Assuntos
Adrenérgicos , Neurônios , Adrenérgicos/farmacologia , Bulbo/fisiologia , Glândulas Suprarrenais , Norepinefrina/farmacologia , Anti-Inflamatórios/farmacologia
5.
Diabetes Obes Metab ; 25(6): 1534-1546, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36852745

RESUMO

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.


Assuntos
Núcleo Arqueado do Hipotálamo , Leptina , Camundongos , Animais , Leptina/metabolismo , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Pró-Opiomelanocortina/uso terapêutico , Glicemia , Obesidade/tratamento farmacológico , Obesidade/etiologia , Dieta , Peso Corporal , Receptores Acoplados a Proteínas G , Neurônios/metabolismo , Camundongos Endogâmicos C57BL
6.
Biochem Biophys Res Commun ; 519(3): 553-558, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31537381

RESUMO

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.


Assuntos
Ingestão de Alimentos , Neurônios/metabolismo , Ocitocina/metabolismo , Vagotomia , Animais , Masculino , Hormônios Estimuladores de Melanócitos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ocitocina/deficiência , Receptores de Ocitocina/antagonistas & inibidores , Receptores de Ocitocina/metabolismo
7.
Neurochem Res ; 42(9): 2658-2665, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28462451

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Deutério/administração & dosagem , Mucosa Gástrica/metabolismo , Grelina/deficiência , Transtornos Parkinsonianos/metabolismo , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/patologia , Grelina/antagonistas & inibidores , Grelina/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/patologia , Estômago/efeitos dos fármacos , Vagotomia/métodos , Nervo Vago/metabolismo , Nervo Vago/cirurgia
8.
Am J Physiol Endocrinol Metab ; 311(5): E881-E890, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27702746

RESUMO

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.


Assuntos
Tecido Adiposo/imunologia , Caspase 1/genética , Quimiocina CCL2/imunologia , Macrófagos/imunologia , Obesidade/genética , Adipócitos/imunologia , Adipócitos/patologia , Adiponectina/imunologia , Tecido Adiposo/patologia , Animais , Glicemia/metabolismo , Composição Corporal , Caspase 1/imunologia , Colesterol/metabolismo , Dieta Hiperlipídica , Citometria de Fluxo , Perfilação da Expressão Gênica , Teste de Tolerância a Glucose , Insulina/metabolismo , Interferon gama/imunologia , Interleucina-12/imunologia , Interleucina-18/imunologia , Interleucina-1beta/imunologia , Interleucina-6/imunologia , Leptina/imunologia , Masculino , Camundongos , Camundongos Knockout , Obesidade/imunologia , Obesidade/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Triglicerídeos/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Microtomografia por Raio-X
9.
Biochem Biophys Res Commun ; 456(3): 727-32, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25511693

RESUMO

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.


Assuntos
Sinalização do Cálcio/fisiologia , Glucagon/fisiologia , Neurônios Aferentes/fisiologia , Gânglio Nodoso/fisiologia , Receptores de Glucagon/fisiologia , Resposta de Saciedade/fisiologia , Nervo Vago/fisiologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Glucagon/administração & dosagem , Glucagon/análogos & derivados , Glucagon/farmacologia , Humanos , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/enzimologia , Gânglio Nodoso/efeitos dos fármacos , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Glucagon/antagonistas & inibidores , Resposta de Saciedade/efeitos dos fármacos , Nervo Vago/efeitos dos fármacos
10.
Am J Physiol Regul Integr Comp Physiol ; 308(5): R360-9, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25540101

RESUMO

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.


Assuntos
Fármacos Antiobesidade/administração & dosagem , Ingestão de Alimentos/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Hiperfagia/tratamento farmacológico , Obesidade/tratamento farmacológico , Ocitocina/administração & dosagem , Nervo Vago/efeitos dos fármacos , Potenciais de Ação , Animais , Depressores do Apetite/administração & dosagem , Cálcio/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação para Baixo , Hiperfagia/fisiopatologia , Injeções Intraperitoneais , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/metabolismo , Obesidade/fisiopatologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fármacos do Sistema Sensorial/farmacologia , Núcleo Solitário/efeitos dos fármacos , Núcleo Solitário/metabolismo , Núcleo Solitário/fisiopatologia , Fatores de Tempo , Vagotomia , Nervo Vago/metabolismo , Nervo Vago/fisiopatologia , Aumento de Peso/efeitos dos fármacos
11.
Am J Physiol Regul Integr Comp Physiol ; 309(5): R561-75, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26157056

RESUMO

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.


Assuntos
Depressores do Apetite/farmacologia , Ingestão de Alimentos/efeitos dos fármacos , Agonistas de Aminoácidos Excitatórios/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Preferências Alimentares/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/agonistas , Serina/farmacologia , Proteína Relacionada com Agouti/metabolismo , Animais , Comportamento de Escolha , Condicionamento Psicológico , Dieta Hiperlipídica , Regulação para Baixo , Antagonistas de Aminoácidos Excitatórios/farmacologia , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Neuropeptídeo Y/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Fármacos do Sistema Sensorial , Fatores de Tempo
12.
Biochem Biophys Res Commun ; 451(2): 276-81, 2014 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-25089000

RESUMO

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.


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Hormônio Liberador da Corticotropina/fisiologia , Proteínas de Ligação a DNA/fisiologia , Ingestão de Alimentos/fisiologia , Peptídeo 1 Semelhante ao Glucagon/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Ocitocina/fisiologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Núcleo Solitário/fisiologia , Animais , Sinalização do Cálcio , Ingestão de Alimentos/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Comportamento Alimentar/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Masculino , Microinjeções , Vias Neurais/fisiologia , Neurônios/fisiologia , Nucleobindinas , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Ratos , Ratos Wistar , Receptores de Glucagon/antagonistas & inibidores
13.
J Nutr Sci Vitaminol (Tokyo) ; 70(3): 193-202, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38945884

RESUMO

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.


Assuntos
Tecido Adiposo Marrom , Fator Neurotrófico Derivado do Encéfalo , Dieta Hiperlipídica , Glucosídeos Iridoides , Iridoides , Norepinefrina , Obesidade , Ratos Sprague-Dawley , Canal de Cátion TRPA1 , Proteína Desacopladora 1 , Animais , Masculino , Proteína Desacopladora 1/metabolismo , Glucosídeos Iridoides/farmacologia , Obesidade/metabolismo , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Iridoides/farmacologia , Norepinefrina/metabolismo , Canal de Cátion TRPA1/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Ratos , Fármacos Antiobesidade/farmacologia , Caminhada , Aumento de Peso/efeitos dos fármacos , Condicionamento Físico Animal , Canais de Cátion TRPV
14.
Nutrients ; 16(14)2024 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-39064713

RESUMO

(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.


Assuntos
Antígenos CD36 , Dieta Hiperlipídica , Absorção Intestinal , Metabolismo dos Lipídeos , Fígado , Camundongos Knockout , PPAR alfa , Proglucagon , Animais , Masculino , Dieta Hiperlipídica/efeitos adversos , PPAR alfa/metabolismo , PPAR alfa/genética , Fígado/metabolismo , Proglucagon/metabolismo , Proglucagon/genética , Antígenos CD36/metabolismo , Antígenos CD36/genética , Camundongos , Esterol Esterase/metabolismo , Esterol Esterase/genética , Triglicerídeos/metabolismo , Camundongos Endogâmicos C57BL , Ácidos Graxos não Esterificados/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Duodeno/metabolismo , Carnitina O-Palmitoiltransferase/metabolismo , Carnitina O-Palmitoiltransferase/genética , Tecido Adiposo/metabolismo , Gorduras na Dieta , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Aciltransferases , Lipase
15.
J Pediatr Endocrinol Metab ; 26(3-4): 343-6, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23348218

RESUMO

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.


Assuntos
Bupropiona/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/metabolismo , Animais , Antidepressivos de Segunda Geração/efeitos adversos , Antidepressivos de Segunda Geração/farmacologia , Bupropiona/efeitos adversos , Feminino , Humanos , Hiperinsulinismo/induzido quimicamente , Hiperinsulinismo/metabolismo , Recém-Nascido , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Bloqueadores dos Canais de Potássio/efeitos adversos , Gravidez
16.
J Diabetes Investig ; 14(6): 746-755, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36977210

RESUMO

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.


Assuntos
Glicemia , Incretinas , Animais , Camundongos , Incretinas/farmacologia , Insulina , Camundongos Endogâmicos C57BL , Fosfato de Sitagliptina/farmacologia , Hipoglicemiantes/farmacologia , Glucose/farmacologia , Peptídeo 1 Semelhante ao Glucagon , Polipeptídeo Inibidor Gástrico
17.
Endocrinology ; 164(6)2023 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-37139968

RESUMO

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.


Assuntos
Peptídeo 1 Semelhante ao Glucagon , Roedores , Ratos , Camundongos , Masculino , Animais , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Roedores/metabolismo , Proteínas de Soja/farmacologia , Proteínas Alimentares , Ingestão de Alimentos/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Peptídeo 2 Semelhante ao Glucagon/farmacologia
18.
J Physiol Sci ; 72(1): 18, 2022 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-35915429

RESUMO

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.


Assuntos
Hiperglicemia , Hipergravidade , Hipotermia , Animais , Glicemia , Corticosterona , Hipergravidade/efeitos adversos , Camundongos , Ratos
19.
Nutrients ; 14(12)2022 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-35745222

RESUMO

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.


Assuntos
Ingestão de Alimentos , Saciação , Animais , Dieta , Ingestão de Alimentos/fisiologia , Mamíferos , Camundongos , Saciação/fisiologia , Nervo Vago/fisiologia , Ácido gama-Aminobutírico/farmacologia
20.
Biosci Biotechnol Biochem ; 75(5): 904-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21597186

RESUMO

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.


Assuntos
Adipócitos Marrons/efeitos dos fármacos , Carboidratos da Dieta/efeitos adversos , Gorduras na Dieta/efeitos adversos , Gordura Intra-Abdominal/efeitos dos fármacos , Canais Iônicos/metabolismo , Proteínas Mitocondriais/metabolismo , Monoglicerídeos/farmacologia , Canais de Cátion TRPV/agonistas , Adipócitos Marrons/metabolismo , Animais , Hipercolesterolemia/etiologia , Hipercolesterolemia/prevenção & controle , Hiperglicemia/etiologia , Hiperglicemia/prevenção & controle , Gordura Intra-Abdominal/metabolismo , Masculino , Camundongos , Monoglicerídeos/química , Sacarose/efeitos adversos , Proteína Desacopladora 1 , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA