Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Tipo del documento
Intervalo de año de publicación
1.
Cell Rep ; 41(13): 111894, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-36577374

RESUMEN

Paradoxically, glucose, the primary driver of satiety, activates a small population of anorexigenic pro-opiomelanocortin (POMC) neurons. Here, we show that lactate levels in the circulation and in the cerebrospinal fluid are elevated in the fed state and the addition of lactate to glucose activates the majority of POMC neurons while increasing cytosolic NADH generation, mitochondrial respiration, and extracellular pyruvate levels. Inhibition of lactate dehydrogenases diminishes mitochondrial respiration, NADH production, and POMC neuronal activity. However, inhibition of the mitochondrial pyruvate carrier has no effect. POMC-specific downregulation of Ucp2 (Ucp2PomcKO), a molecule regulated by fatty acid metabolism and shown to play a role as transporter in the malate-aspartate shuttle, abolishes lactate- and glucose-sensing of POMC neurons. Ucp2PomcKO mice have impaired glucose metabolism and are prone to obesity on a high-fat diet. Altogether, our data show that lactate through redox signaling and blocking mitochondrial glucose utilization activates POMC neurons to regulate feeding and glucose metabolism.


Asunto(s)
NAD , Proopiomelanocortina , Ratones , Animales , Proopiomelanocortina/metabolismo , NAD/metabolismo , Glucosa/metabolismo , Neuronas/metabolismo , Lactatos/metabolismo , Hipotálamo/metabolismo , Proteína Desacopladora 2/metabolismo
2.
J Chem Neuroanat ; 123: 102117, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35680104

RESUMEN

The hypothalamus is a large brain region made of nuclei and areas involved in the control of behaviors and physiological regulations. Among them, the arcuate nucleus (ARH) and the lateral hypothalamic area (LHA) contain key neuronal populations expressing the pro-opiomelanocortin (POMC), the agouti-related peptide (AgRP), and the melanin-concentrating hormone (MCH), respectively, that are involved in goal-oriented behaviors (such as feeding behavior) and glucose homeostasis. These neuronal populations are generated from distinct parts of the germinative neuroepithelium during embryonic life, and acquire their cell fate under the influence of morphogen proteins, specific transcription factors, and epigenetic modulators. POMC and MCH neuronal development continues by sending long descending axonal projections before birth under the control of axon guidance molecules such as Netrin1 and Slit2. Later, during the postnatal period, POMC and AgRP neurons develop intra-hypothalamic projections notably to the paraventricular nucleus of the hypothalamus through the influence of other axon guidance cues such as the class3 Semaphorins. Other cellular processes, such as autophagy and primary cilia function, and hormonal cues also appear critical for the proper development of POMC neurons.


Asunto(s)
Hipotálamo , Proopiomelanocortina , Proteína Relacionada con Agouti/metabolismo , Núcleo Arqueado del Hipotálamo/metabolismo , Hipotálamo/metabolismo , Neuronas/metabolismo , Proopiomelanocortina/metabolismo
3.
J Neuroendocrinol ; 34(6): e13145, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35581942

RESUMEN

Pregnancy is energetically demanding and therefore, by necessity, reproduction and energy balance are inextricably linked. With insufficient or excessive energy stores a female is liable to suffer complications during pregnancy or produce unhealthy offspring. Gonadotropin-releasing hormone neurons are responsible for initiating both the pulsatile and subsequent surge release of luteinizing hormone to control ovulation. Meticulous work has identified two hypothalamic populations of kisspeptin (Kiss1) neurons that are critical for this pattern of release. The involvement of the hypothalamus is unsurprising because its quintessential function is to couple the endocrine and nervous systems, coordinating energy balance and reproduction. Estrogens, more specifically 17ß-estradiol (E2 ), orchestrate the activity of a triumvirate of hypothalamic neurons within the arcuate nucleus (ARH) that govern the physiological underpinnings of these behavioral dynamics. Arising from a common progenitor pool, these cells differentiate into ARH kisspeptin, pro-opiomelanocortin (POMC), and agouti related peptide/neuropeptide Y (AgRP) neurons. Although the excitability of all these subpopulations is subject to genomic and rapid estrogenic regulation, Kiss1 neurons are the most sensitive, reflecting their integral function in female fertility. Based on the premise that E2 coordinates autonomic functions around reproduction, we review recent findings on how Kiss1 neurons interact with gonadotropin-releasing hormone, AgRP and POMC neurons, as well as how the rapid membrane-initiated and intracellular signaling cascades activated by E2 in these neurons are critical for control of homeostatic functions supporting reproduction. In particular, we highlight how Kiss1 and POMC neurons conspire to inhibit AgRP neurons and diminish food motivation in service of reproductive success.


Asunto(s)
Kisspeptinas , Proopiomelanocortina , Proteína Relacionada con Agouti , Núcleo Arqueado del Hipotálamo/metabolismo , Femenino , Hormona Liberadora de Gonadotropina/metabolismo , Homeostasis , Humanos , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Reproducción/fisiología
4.
Neuroendocrinology ; 112(3): 287-297, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-33906196

RESUMEN

OBJECTIVES: The control of energy balance relies on the counterbalancing release of neuropeptides encoded by the pro-opiomelanocortin (Pomc) and agouti-related protein (Agrp) genes, expressed by 2 distinct neuronal populations of the arcuate (ARC) nucleus of the hypothalamus. Although largely segregated, single-cell resolution techniques demonstrate some degree of co-expression. We studied whether challenges to the control of energy balance influence the degree of Agrp and Pomc co-expression in ARC melanocortin neurons. METHODS: We used fluorescence-activated cell sorting followed by quantitative polymerase chain reaction and fluorescent in situ hybridization to measure Pomc and Agrp gene co-expression in POMC or AGRP neurons in response to (1) acute or chronic calorie restriction, or (2) obesity due to loss of leptin receptor expression or chronic high-fat diet feeding in male mice. RESULTS: Melanocortin ARC neurons of fed mice exhibited low, yet detectable, levels of Pomc and Agrp gene co-expression. Calorie restriction significantly increased and decreased total Agrp and Pomc expression, respectively, and reduced the expression of Pomc relative to Agrp in AGRP neurons. Leptin-deficient db/db mice showed increased total Agrp levels and decreased Pomc expression, as well as significantly increased Agrp expression relative to Pomc in POMC neurons. Expression or co-expression levels did not differ between diet-induced obese mice and lean controls. CONCLUSIONS: Changes in Agrp and Pomc co-expression within POMC and AGRP neurons following chronic calorie restriction or in db/db mice suggest an additional mechanism to further suppress the melanocortin signaling during conditions of severely reduced leptin action.


Asunto(s)
Leptina , Proopiomelanocortina , Proteína Relacionada con Agouti/genética , Proteína Relacionada con Agouti/metabolismo , Animales , Hipotálamo/metabolismo , Hibridación Fluorescente in Situ , Leptina/metabolismo , Masculino , Melanocortinas , Ratones , Neuronas/metabolismo , Estado Nutricional , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo
5.
Reprod Sci ; 29(1): 173-183, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34767244

RESUMEN

Adulthood obesity, diabetes, and metabolic diseases are associated with small for gestational age (SGA) newborns. This association could be related to abnormal appetite signaling pathways in the hypothalamus. This study investigated the appetite regulation by the hypothalamus of SGA newborns by establishing an SGA rat model and culturing SGA neural progenitor cells (NPCs) in vitro. Models of SGA were established by maternal food restriction embryonic day 10 (E10). At E18, postpartum day 1 (P1), and P5, hypothalamic neural precursor cells (NPCs) of offspring were cultured in vitro. Immunofluorescence, Western blot (WB), and qRT-PCR were used to assess NPY, POMC, and FoxO1 expression levels. The effects on mRNA expression of the FoxO1-specific inhibitor AS1842856 were examined. The results indicated that compared with controls, NPY was higher, and POMC was lower at embryonic day 18 (E18), postpartum day 1 (P1), and P5. The proliferation and migration of NPCs in the third ventricle of SGA hypothalami were lower than in controls. After treatment with the FoxO1 inhibitor AS1842856, the differences in the mRNA expression of NPY and POMC between the two groups disappeared. NPY and POMC mRNA levels in the SGA group treated with AS1842856 were not significantly different compared with the control group without AS1842856 treatment. In conclusion, SGA pups showed an increase in appetite-promoting NPY and a decrease in appetite-reducing POMC, probably contributing to adulthood weight gain, obesity, and endocrine disorders.


Asunto(s)
Proteína Forkhead Box O1/metabolismo , Hipotálamo/metabolismo , Células-Madre Neurales/metabolismo , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , Animales , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Proteína Forkhead Box O1/genética , Edad Gestacional , Hipotálamo/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Neuropéptido Y/genética , Proopiomelanocortina/genética , Quinolonas/farmacología , Ratas
6.
Trends Endocrinol Metab ; 32(4): 191-193, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33485760

RESUMEN

Clinical applications of molecular hydrogen (H2) seem to favorably affect obesity-related metabolic biomarkers in peripheral tissues, yet whether H2 directly tackles obesity pathways in the brain remains elusive. I summarize here several molecular targets in the hypothalamus and beyond that could be altered by H2 gas in obesity.


Asunto(s)
Hidrógeno , Obesidad , Humanos , Hidrógeno/farmacología , Hidrógeno/uso terapéutico , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología
7.
Alcohol ; 86: 103-112, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32304714

RESUMEN

Alcohol abuse is a worldwide public health concern, yet the precise molecular targets of alcohol in the brain are still not fully understood. Alcohol may promote its euphoric and motivational effects, in part, by activating the endogenous opioid system. One particular component of this system consists of pro-opiomelanocortin (POMC) -producing neurons in the arcuate nucleus (ArcN) of the hypothalamus, which project to reward-related brain areas. To identify the physiological effects of ethanol on ArcN POMC neurons, we utilized whole cell patch-clamp recordings and bath application of ethanol (5-40 mM) to identify alterations in spontaneous baseline activity, rheobase, spiking characteristics, or intrinsic neuronal properties. We found that 10 mM ethanol increased the number of depolarization-induced spikes in the majority of recorded cells, whereas higher concentrations of ethanol (20-40 mM) decreased the number of spikes. Interestingly, we found that basal firing rates of ArcN POMC neurons may predict physiological responding to ethanol. Rheobase and spontaneous activity, measured by spontaneous excitatory post-synaptic potentials (EPSPs) at rest, were unchanged after exposure to ethanol, regardless of concentration. These results suggest that ethanol has concentration-dependent modulatory effects on ArcN POMC neuronal activity, which may be relevant to treatments for alcohol use disorders that target endogenous opioid systems.


Asunto(s)
Etanol/farmacología , Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Proopiomelanocortina/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Ratones , Ratones Transgénicos , Técnicas de Placa-Clamp
8.
Cell Rep ; 30(9): 3067-3078.e5, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32130907

RESUMEN

Mechanistic studies in rodents evidenced synaptic remodeling in neuronal circuits that control food intake. However, the physiological relevance of this process is not well defined. Here, we show that the firing activity of anorexigenic POMC neurons located in the hypothalamus is increased after a standard meal. Postprandial hyperactivity of POMC neurons relies on synaptic plasticity that engages pre-synaptic mechanisms, which does not involve structural remodeling of synapses but retraction of glial coverage. These functional and morphological neuroglial changes are triggered by postprandial hyperglycemia. Chemogenetically induced glial retraction on POMC neurons is sufficient to increase POMC activity and modify meal patterns. These findings indicate that synaptic plasticity within the melanocortin system happens at the timescale of meals and likely contributes to short-term control of food intake. Interestingly, these effects are lost with a high-fat meal, suggesting that neuroglial plasticity of POMC neurons is involved in the satietogenic properties of foods.


Asunto(s)
Hiperglucemia/fisiopatología , Hipotálamo/metabolismo , Comidas , Neuroglía/patología , Plasticidad Neuronal , Neuronas/metabolismo , Proopiomelanocortina/metabolismo , Animales , Glucemia/metabolismo , Fenómenos Electrofisiológicos , Conducta Alimentaria , Hiperglucemia/sangre , Ratones Endogámicos C57BL , Ratones Transgénicos , Periodo Posprandial , Sinapsis/metabolismo
9.
Trends Endocrinol Metab ; 31(5): 368-377, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32035736

RESUMEN

Genome-wide association studies (GWASs) have identified SNPs of the fat mass and obesity (FTO) gene as the most important risk alleles for obesity. However, how the presence of risk alleles affect phenotype is still a matter of intense investigation. In 2014, a study revealed that long-range enhancers from the intronic regions of the FTO gene regulate iroquois-class homeobox protein (IRX)3 expression. IRX3 is expressed in hypothalamic pro-opiomelanocortin (POMC) neurons and changes in its expression levels affect body adiposity by modifying food intake and energy expenditure. These findings have placed IRX3 as a potential target for the treatment of obesity. Here, we review studies that evaluated the roles of IRX3 in development, neurogenesis, and body energy homeostasis.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/metabolismo , Metabolismo Energético/fisiología , Proteínas de Homeodominio/metabolismo , Hipotálamo/metabolismo , Obesidad/metabolismo , Proopiomelanocortina/metabolismo , Factores de Transcripción/metabolismo , Dioxigenasa FTO Dependiente de Alfa-Cetoglutarato/genética , Animales , Metabolismo Energético/genética , Proteínas de Homeodominio/genética , Humanos , Obesidad/genética , Factores de Transcripción/genética
10.
Biomolecules ; 9(10)2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31635295

RESUMEN

Anorexia is common in patients with cancer, mostly as a side effect of chemotherapy. The effect of electro-acupuncture (EA) on ameliorating cancer-related symptoms have been studied in animal models and in clinical trials. The aim of this study was to determine optimal conditions for the application of EA to alleviate anorexia, followed by the study of molecular mechanisms affecting its therapeutics. Anorexia was induced in male Wistar rats by injecting cisplatin, which was then followed by EA treatment at CV12, the acupuncture point located in the center of the abdominal midline. Body weight and food intake were measured daily throughout the duration of the study. The levels of monoamine neurotransmitters in the plasma were quantitatively analyzed by HPLC-ECD. Gastrointestinal hormone concentrations were elucidated with ELISA kits. RT-qPCR was performed to evaluate the mRNA expression of ghrelin (GHRL), neuropeptide Y (NPY), and pro-opiomelanocortin. The expression of c-Fos in the nucleus tractus solitarii was detected using western blotting analysis. The optimal conditions of EA to alleviate anorexia in rats was determined to be 1 unit for intensity and 10 Hz for frequency. EA treatment at CV12 reduced the levels of plasma monoamine neurotransmitters 5-hydroxytryptamine, 5-hydroxyindoleacetic acid, dopamine, and norepinephrine; as well as stimulated the expression of GHRL and NPY to alleviate cisplatin-induced anorexia in rats. EA stimulation at CV12 could be used to treat cisplatin-induced anorexia in rats.


Asunto(s)
Terapia por Acupuntura , Aminas/metabolismo , Anorexia/inducido químicamente , Anorexia/terapia , Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Ghrelina/metabolismo , Neurotransmisores/metabolismo , Aminas/sangre , Animales , Antineoplásicos/administración & dosificación , Cisplatino/administración & dosificación , Modelos Animales de Enfermedad , Ghrelina/sangre , Inyecciones Intraperitoneales , Masculino , Neurotransmisores/sangre , Ratas , Ratas Wistar
11.
Mol Metab ; 20: 194-204, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30503832

RESUMEN

OBJECTIVE: Life-threatening hypoglycemia is a major limiting factor in the management of diabetes. While it is known that counterregulatory responses to hypoglycemia are impaired in diabetes, molecular mechanisms underlying the reduced responses remain unclear. Given the established roles of the hypothalamic proopiomelanocortin (POMC)/melanocortin 4 receptor (MC4R) circuit in regulating sympathetic nervous system (SNS) activity and the SNS in stimulating counterregulatory responses to hypoglycemia, we hypothesized that hypothalamic POMC as well as MC4R, a receptor for POMC derived melanocyte stimulating hormones, is required for normal hypoglycemia counterregulation. METHODS: To test the hypothesis, we induced hypoglycemia or glucopenia in separate cohorts of mice deficient in either POMC or MC4R in the arcuate nucleus (ARC) or the paraventricular nucleus of the hypothalamus (PVH), respectively, and measured their circulating counterregulatory hormones. In addition, we performed a hyperinsulinemic-hypoglycemic clamp study to further validate the function of MC4R in hypoglycemia counterregulation. We also measured Pomc and Mc4r mRNA levels in the ARC and PVH, respectively, in the streptozotocin-induced type 1 diabetes mouse model and non-obese diabetic (NOD) mice to delineate molecular mechanisms by which diabetes deteriorates the defense systems against hypoglycemia. Finally, we treated diabetic mice with the MC4R agonist MTII, administered stereotaxically into the PVH, to determine its potential for restoring the counterregulatory response to hypoglycemia in diabetes. RESULTS: Stimulation of epinephrine and glucagon release in response to hypoglycemia or glucopenia was diminished in both POMC- and MC4R-deficient mice, relative to their littermate controls. Similarly, the counterregulatory response was impaired in association with decreased hypothalamic Pomc and Mc4r expression in the diabetic mice, a phenotype that was not reversed by insulin treatment which normalized glycemia. In contrast, infusion of an MC4R agonist in the PVH restored the counterregulatory response in diabetic mice. CONCLUSION: In conclusion, hypothalamic Pomc as well as Mc4r, both of which are reduced in type 1 diabetic mice, are required for normal counterregulatory responses to hypoglycemia. Therefore, enhancing MC4R function may improve hypoglycemia counterregulation in diabetes.


Asunto(s)
Hipoglucemia/metabolismo , Hipotálamo/metabolismo , Proopiomelanocortina/metabolismo , Receptor de Melanocortina Tipo 4/metabolismo , Animales , Epinefrina/metabolismo , Glucagón/metabolismo , Homeostasis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Proopiomelanocortina/deficiencia , Proopiomelanocortina/genética , Receptor de Melanocortina Tipo 4/deficiencia , Receptor de Melanocortina Tipo 4/genética
12.
Epigenetics ; 13(10-11): 1127-1140, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30395773

RESUMEN

The central nervous system monitors modifications in metabolic parameters or hormone levels (leptin) and elicits adaptive responses such as food intake and glucose homeostasis regulation. Particularly, within the hypothalamus, pro-opiomelanocortin (POMC) neurons are crucial regulators of energy balance. Consistent with a pivotal role of the melanocortin system in the control of energy homeostasis, disruption of the Pomc gene causes hyperphagia and obesity. Pomc gene expression is tightly controlled by different mechanisms. Interestingly, recent studies pointed to a key role for micro ribonucleic acid (miRNAs) in the regulation of gene expression. However, the role of miRNAs in the leptin sensitivity in hypothalamic melanocortin system has never been assessed. We developed a transgenic mouse model (PDKO) with a partial deletion of the miRNA processing enzyme DICER specifically in POMC neurons. PDKO mice exhibited a normal body weight but a decrease of food intake. Interestingly, PDKO mice had decreased metabolic rate by reduction of VO2 consumption and CO2 production which could explain that PDKO mice have normal weight while eating less. Interestingly, we observed an increase of leptin sensitivity in the POMC neurons of PDKO mice which could explain the decrease of food intake in this model. We also observed an increase in the expression of genes involved in the function of brown adipose tissue that is in polysynaptic contact with the POMC neurons. In summary, these results support the hypothesis that Dicer-derived miRNAs may be involved in the effect of leptin on POMC neurons activity.


Asunto(s)
Hipotálamo/metabolismo , Leptina/metabolismo , MicroARNs/genética , Tejido Adiposo Pardo/metabolismo , Animales , Peso Corporal , Ingestión de Alimentos , Masculino , Ratones , MicroARNs/metabolismo , Neuronas/metabolismo , Consumo de Oxígeno , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Ribonucleasa III/genética
13.
Curr Biol ; 28(23): 3736-3747.e3, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30471995

RESUMEN

Eating and sleeping represent two mutually exclusive behaviors that satisfy distinct homeostatic needs. Because an animal cannot eat and sleep at the same time, brain systems that regulate energy homeostasis are likely to influence sleep/wake behavior. Indeed, previous studies indicate that animals adjust sleep cycles around periods of food need and availability. Furthermore, hormones that affect energy homeostasis also affect sleep/wake states: the orexigenic hormone ghrelin promotes wakefulness, and the anorexigenic hormones leptin and insulin increase the duration of slow-wave sleep. However, whether neural populations that regulate feeding can influence sleep/wake states is unknown. The hypothalamic arcuate nucleus contains two neuronal populations that exert opposing effects on energy homeostasis: agouti-related protein (AgRP)-expressing neurons detect caloric need and orchestrate food-seeking behavior, whereas activity in pro-opiomelanocortin (POMC)-expressing neurons induces satiety. We tested the hypotheses that AgRP neurons affect sleep homeostasis by promoting states of wakefulness, whereas POMC neurons promote states of sleep. Indeed, optogenetic or chemogenetic stimulation of AgRP neurons in mice promoted wakefulness while decreasing the quantity and integrity of sleep. Inhibition of AgRP neurons rescued sleep integrity in food-deprived mice, highlighting the physiological importance of AgRP neuron activity for the suppression of sleep by hunger. Conversely, stimulation of POMC neurons promoted sleep states and decreased sleep fragmentation in food-deprived mice. Interestingly, we also found that sleep deprivation attenuated the effects of AgRP neuron activity on food intake and wakefulness. These results indicate that homeostatic feeding neurons can hierarchically affect behavioral outcomes, depending on homeostatic need.


Asunto(s)
Ingestión de Alimentos , Hambre , Hipotálamo/fisiología , Neuronas/fisiología , Sueño/fisiología , Vigilia/fisiología , Animales , Privación de Alimentos , Homeostasis , Masculino , Ratones
14.
Cell Tissue Res ; 374(3): 587-594, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30078105

RESUMEN

Tributyltin (TBT), an antifouling agent found in boat paints, is a common contaminant of marine and freshwater ecosystems. It is rapidly absorbed by organic materials and accumulated in many aquatic animals. Human exposure may depend on ingestion of contaminated food or by indirect exposure from household items containing organotin compounds. TBT is defined as an endocrine disruptor compound (EDC) because it binds to androgen receptors. Moreover, it is also included on the list of metabolic disruptors. The brain is a known target of TBT and this compound interferes with the orexigenic system, inducing a strong decrease in NPY expression in the hypothalamus. In the present experiment, we investigated the effect of a chronic treatment with TBT on the mouse anorexigenic system in both sexes, to look at the pro-opiomelanocortin (POMC) expression in the paraventricular (PVN), dorsomedial (DMN), ventromedial (VMN), and arcuate (ARC) hypothalamic nuclei. The results show a sexually dimorphic effect of TBT on both systems. TBT induced a significant decrease of POMC-positive structures only in female mice in DMN, ARC, and in PVN for both sexes. Apparently, these results show that TBT may interfere with the anorexigenic system in hypothalamic areas involved in the control of food intake, by inhibiting POMC in a sexually dimorphic way. In conclusion, in addition to having a direct effect on fat tissue, the effects of TBT as metabolic disruptor, may be due to gender-specific actions on both orexigenic and anorexigenic hypothalamic systems.


Asunto(s)
Envejecimiento/metabolismo , Hipotálamo/metabolismo , Proopiomelanocortina/metabolismo , Caracteres Sexuales , Compuestos de Trialquiltina/farmacología , Adiposidad/efectos de los fármacos , Animales , Femenino , Masculino , Ratones , Aumento de Peso/efectos de los fármacos
15.
J Agric Food Chem ; 66(12): 3161-3168, 2018 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-29526104

RESUMEN

The objective of this study was to investigate whether valine (Val) supplementation in a reduced protein (RP) diet regulates growth performance associated with the changes in plasma amino acids (AAs) profile, metabolism, endocrine, and neural system in piglets. Piglets or piglets with a catheter in the precaval vein were randomly assigned to two treatments, including two RP diets with standardized ileal digestible (SID) Val:Lysine (Lys) ratio of 0.45 and 0.65, respectively. The results indicated that piglets in the higher Val:Lys ratio treatment had higher average daily feed intake (ADFI) ( P < 0.001), average daily gain (ADG) ( P = 0.001), feed conversion ratio (FCR) ( P = 0.004), lower plasma urea nitrogen ( P = 0.032), expression of gastric cholecystokinin (CCK), and hypothalamic pro-opiomelanocortin (POMC). Plasma AAs profiles including postprandial plasma essential AAs (EAAs) profile and in serum, muscle, and liver involved in metabolism of AAs and fatty acids were significantly different between two treatments. In conclusion, Val influenced growth performance associated with metabolism of AAs and fatty acids and both endocrine and neural system in piglets.


Asunto(s)
Aminoácidos/sangre , Alimentación Animal/análisis , Dieta con Restricción de Proteínas/veterinaria , Sistema Endocrino/metabolismo , Sistema Nervioso/metabolismo , Porcinos/crecimiento & desarrollo , Valina/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Colecistoquinina/metabolismo , Suplementos Dietéticos/análisis , Digestión , Sistema Endocrino/efectos de los fármacos , Femenino , Íleon/metabolismo , Masculino , Sistema Nervioso/efectos de los fármacos , Proopiomelanocortina/metabolismo , Porcinos/sangre , Valina/administración & dosificación
16.
Neurochem Res ; 43(4): 821-837, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29397535

RESUMEN

The pro-opiomelanocortin (POMC)-expressing neurons of the hypothalamic arcuate nucleus (ARC) are involved in the control of food intake and metabolic processes. It is assumed that, in addition to leptin, the activity of these neurons is regulated by serotonin and dopamine, but only subtype 2C serotonin receptors (5-HT2CR) was identified earlier on the POMC-neurons. The aim of this work was a comparative study of the localization and number of leptin receptors (LepR), types 1 and 2 dopamine receptors (D1R, D2R), 5-HT1BR and 5-HT2CR on the POMC-neurons and the expression of the genes encoding them in the ARC of the normal and diet-induced obese (DIO) rodents and the agouti mice (A y /a) with the melanocortin obesity. As shown by immunohistochemistry (IHC), all the studied receptors were located on the POMC-immunopositive neurons, and their IHC-content was in agreement with the expression of their genes. In DIO rats the number of D1R and D2R in the POMC-neurons and their expression in the ARC were reduced. In DIO mice the number of D1R and D2R did not change, while the number of LepR and 5-HT2CR was increased, although to a small extent. In the POMC-neurons of agouti mice the number of LepR, D2R, 5-HT1BR and 5-HT2CR was increased, and the D1R number was reduced. Thus, our data demonstrates for the first time the localization of different types of the serotonin and dopamine receptors on the POMC-neurons and a specific pattern of the changes of their number and expression in the DIO and melanocortin obesity.


Asunto(s)
Hipotálamo/metabolismo , Obesidad/metabolismo , Proopiomelanocortina/biosíntesis , Receptores Dopaminérgicos/biosíntesis , Receptores de Leptina/biosíntesis , Receptores de Serotonina/biosíntesis , Animales , Femenino , Hipotálamo/química , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/química , Neuronas/metabolismo , Proopiomelanocortina/análisis , Ratas , Ratas Wistar , Receptores Dopaminérgicos/análisis , Receptores de Leptina/análisis , Receptores de Serotonina/análisis , Roedores
17.
Aging Cell ; 17(1)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29168299

RESUMEN

Weight loss is an effective intervention for diminishing disease burden in obese older adults. Pharmacological interventions that reduce food intake and thereby promote weight loss may offer effective strategies to reduce age-related disease. We previously reported that 17α-estradiol (17α-E2) administration elicits beneficial effects on metabolism and inflammation in old male mice. These observations were associated with reduced calorie intake. Here, we demonstrate that 17α-E2 acts through pro-opiomelanocortin (Pomc) expression in the arcuate nucleus (ARC) to reduce food intake and body mass in mouse models of obesity. These results confirm that 17α-E2 modulates appetite through selective interactions within hypothalamic anorexigenic pathways. Interestingly, some peripheral markers of metabolic homeostasis were also improved in animals with near complete loss of ARC Pomc transcription. This suggests that 17α-E2 might have central and peripheral actions that can beneficially affect metabolism cooperatively or independently.


Asunto(s)
Estradiol/farmacología , Conducta Alimentaria/efectos de los fármacos , Hipotálamo/efectos de los fármacos , Neuronas/efectos de los fármacos , Proopiomelanocortina/farmacología , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Conducta Animal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Hipotálamo/metabolismo , Leptina/metabolismo , Ratones Transgénicos , Neuronas/metabolismo , Obesidad/metabolismo , Proopiomelanocortina/metabolismo
18.
Artículo en Chino | WPRIM | ID: wpr-735147

RESUMEN

Objective:To explore the central neurobiological mechanisms of pleasure effect on rats with neuralgia treated by tuina manipulations of An-pressing and Rou-kneading Huantiao (GB 30).Methods:A total of 64 male Sprague-Dawley (SD) rats were used in this study.Eighteen rats were randomly selected as a normal group,and the other 46 rats were used to duplicate the chronic constriction injury (CCI) model.Ten rats failed in modeling and 36 rats succeeded.These 36 rats were then randomly divided into a model group and a tuina group,with 18 rats in each group.The rats in the normal group and the model group did not receive any interventions,while those in the tuina group received An-pressing and Rou-kneading Huantiao (GB 30),1 min for each time,once a day,3 weeks in total.Heating tests were evaluated to observe the change of pain-sensitivity score before intervention,1 week after intervention,2 weeks after intervention,and 3 weeks after intervention.After 1 week of intervention,2 weeks of intervention,and 3 weeks of intervention,6 rats were randomly selected from each group respectively for brain extraction.The change of Nissl's body and β-endorphin in the accumbens nucleus as well as amygdaloid nucleus of pleasure circuits,and pro-opiomelanocortin (POMC) in the arcuate nucleus were analyzed by methods of histochemistry and molecular biology.Results:After modeling,the pain-sensitivity scores of the tuina group and the model group were statistically different from the score of the normal group (both P<0.05).After An-pressing and Rou-kneading Huantiao (GB 30) for one week,the pain-sensitivity score of the tuina group had statistical difference compared with that of the model group (P<0.05).At each different time point:the amounts of Nissl's body in accumbens nucleus and amygdaloid nucleus of the tuina group were significantly more than those of the model group (all P<0.01).Besides,the numbers of β-endorphin immunoreactive cells in the accumbens nucleus and amygdaloid nucleus of the rats in the tuina group were significantly higher than those in the model group (all P<0.01),and so was the expression of POMC in arcuate nucleus (all P<0.01).Conclusion:An-pressing and Rou-kneading Huantiao (GB 30),where the sciatic nerve is ligated,can reduce pain-sensitivity score and increase pain tolerance value of rats with chronic neuralgia.It can increase the activity of neurons in accumbens nucleus and amygdaloid nucleus of pleasure circuits,which indicates that the analgesia effect of tuina therapy may correlate with pleasure effect,and also reveals a part of neurobiological mechanisms of neuralgia.

19.
Neuropeptides ; 66: 25-35, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28823463

RESUMEN

Glucose counter-regulatory dysfunction correlates with impaired activation of the hypothalamic metabolic sensor adenosine 5'-monophosphate-activated protein kinase (AMPK). Hypothalamic AMPK is controlled by hindbrain energy status; we examined here whether hindbrain AMPK regulates hypothalamic AMPK and metabolic neurotransmitter maladaptation to recurring insulin-induced hypoglycemia (RIIH). Brain tissue was harvested after single versus serial insulin (I) dosing for Western blot analysis of AMPK, phospho-AMPK (pAMPK), and relevant biosynthetic enzyme/neuropeptide expression in micro-punch dissected arcuate (ARH), ventromedial (VMH), dorsomedial (DMH) nuclei and lateral hypothalamic area (LHA) tissue. The AMPK inhibitor compound c (Cc) or vehicle was administered to the caudal fourth ventricle ahead of antecedent I injections. RIIH caused site-specific elevation (ARH, VMH, LHA) or reduction (DMH) of total AMPK protein versus acute hypoglycemia; Cc respectively exacerbated or attenuated this response in the ARH and VMH. Hindbrain AMPK correspondingly inhibited or stimulated LHA and DMH pAMPK expression during RIIH. RIIH elicited Cc-reversible augmentation of VMH glutamate decarboxylase profiles, but stimulated (ARH pro-opiomelanocortin; LHA orexin-A) or decreased (VMH nitric oxide synthase) other metabolic neurotransmitters without hindbrain sensor involvement. Results demonstrate acclimated up-regulation of total AMPK protein expression in multiple hypothalamic loci during RIIH, and document hindbrain sensor contribution to amplification of this protein profile in the VMH. Concurrent lack of net change in ARH and VMH tissue pAMPK implies adaptive reductions in local sensor activity, which may/may not reflect positive gain in energy state. It remains unclear if 'glucose-excited' VMH GABAergic and/or ARH pro-opiomelanocortin neurons exhibit AMPK habituation to RIIH, and whether diminished sensor activation in these and other mediobasal hypothalamic neurotransmitter populations may contribute to HAAF.


Asunto(s)
Adenilato Quinasa/metabolismo , Hipoglucemia/metabolismo , Hipotálamo/metabolismo , Rombencéfalo/metabolismo , Animales , Glucemia/metabolismo , Hipoglucemia/inducido químicamente , Insulina , Masculino , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Norepinefrina/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley
20.
Adv Exp Med Biol ; 960: 381-397, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28585208

RESUMEN

Leptin signaling blockade by chronic overstimulation of the leptin receptor or hypothalamic pro-inflammatory responses due to elevated levels of saturated fatty acid can induce leptin resistance by activating negative feedback pathways. Although, long form leptin receptor (Ob-Rb) initiates leptin signaling through more than seven different signal transduction pathways, excessive suppressor of cytokine signaling-3 (SOCS-3) activity is a potential mechanism for the leptin resistance that characterizes human obesity. Because the leptin-responsive metabolic pathways broadly integrate with other neurons to control energy balance, the methods used to counteract the leptin resistance has extremely limited effect. In this chapter, besides the impairment of central and peripheral leptin signaling pathways, limited access of leptin to central nervous system (CNS) through blood-brain barrier, mismatch between high leptin and the amount of leptin receptor expression, contradictory effects of cellular and circulating molecules on leptin signaling, the connection between leptin signaling and endoplasmic reticulum (ER) stress and self-regulation of leptin signaling has been discussed in terms of leptin resistance.


Asunto(s)
Dieta/efectos adversos , Leptina/metabolismo , Obesidad/fisiopatología , Animales , Ingestión de Alimentos/fisiología , Conducta Alimentaria/fisiología , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Obesidad/metabolismo , Receptores de Leptina/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA