Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Endocr Rev ; 44(2): 193-221, 2023 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35930274

RESUMEN

The etiology of central precocious puberty (CPP) is multiple and heterogeneous, including congenital and acquired causes that can be associated with structural or functional brain alterations. All causes of CPP culminate in the premature pulsatile secretion of hypothalamic GnRH and, consequently, in the premature reactivation of hypothalamic-pituitary-gonadal axis. The activation of excitatory factors or suppression of inhibitory factors during childhood represent the 2 major mechanisms of CPP, revealing a delicate balance of these opposing neuronal pathways. Hypothalamic hamartoma (HH) is the most well-known congenital cause of CPP with central nervous system abnormalities. Several mechanisms by which hamartoma causes CPP have been proposed, including an anatomical connection to the anterior hypothalamus, autonomous neuroendocrine activity in GnRH neurons, trophic factors secreted by HH, and mechanical pressure applied to the hypothalamus. The importance of genetic and/or epigenetic factors in the underlying mechanisms of CPP has grown significantly in the last decade, as demonstrated by the evidence of genetic abnormalities in hypothalamic structural lesions (eg, hamartomas, gliomas), syndromic disorders associated with CPP (Temple, Prader-Willi, Silver-Russell, and Rett syndromes), and isolated CPP from monogenic defects (MKRN3 and DLK1 loss-of-function mutations). Genetic and epigenetic discoveries involving the etiology of CPP have had influence on the diagnosis and familial counseling providing bases for potential prevention of premature sexual development and new treatment targets in the future. Global preventive actions inducing healthy lifestyle habits and less exposure to endocrine-disrupting chemicals during the lifespan are desirable because they are potentially associated with CPP.


Asunto(s)
Enfermedades Hipotalámicas , Pubertad Precoz , Humanos , Pubertad Precoz/diagnóstico , Pubertad Precoz/genética , Hormona Liberadora de Gonadotropina/metabolismo , Enfermedades Hipotalámicas/complicaciones , Hipotálamo , Pubertad , Ubiquitina-Proteína Ligasas/metabolismo
2.
Nutr Neurosci ; 25(5): 931-944, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-32954972

RESUMEN

Aim: We aimed to investigate whether maternal malnutrition during gestation/lactation induces long-lasting changes on inflammation, lipid metabolism and endocannabinoid signaling in the adult offspring hypothalamus and the role of hypothalamic astrocytes in these changes.Methods: We analyzed the effects of a free-choice hypercaloric palatable diet (P) during (pre)gestation, lactation and/or post-weaning on inflammation, lipid metabolism and endogenous cannabinoid signaling in the adult offspring hypothalamus. We also evaluated the response of primary hypothalamic astrocytes to palmitic acid and anandamide.Results: Postnatal exposure to a P diet induced factors involved in hypothalamic inflammation (Tnfa and Il6) and gliosis (Gfap, vimentin and Iba1) in adult offspring, being more significant in females. In contrast, maternal P diet reduced factors involved in astrogliosis (vimentin), fatty acid oxidation (Cpt1a) and monounsaturated fatty acid synthesis (Scd1). These changes were accompanied by an increase in the expression of the genes for the cannabinoid receptor (Cnr1) and Nape-pld, an enzyme involved in endocannabinoid synthesis, in females and a decrease in the endocannabinoid degradation enzyme Faah in males. These changes suggest that the maternal P diet results in sex-specific alterations in hypothalamic endocannabinoid signaling and lipid metabolism. This hypothesis was tested in hypothalamic astrocyte cultures, where palmitic acid (PA) and the polyunsaturated fatty acid N-arachidonoylethanolamine (anandamide or AEA) were found to induce similar changes in the endocannabinoid system (ECS) and lipid metabolism.Conclusion: These results stress the importance of both maternal diet and sex in long term metabolic programming and suggest a possible role of hypothalamic astrocytes in this process.


Asunto(s)
Cannabinoides , Endocannabinoides , Hijos Adultos , Ácidos Araquidónicos , Astrocitos/metabolismo , Cannabinoides/metabolismo , Dieta , Femenino , Gliosis/metabolismo , Humanos , Hipotálamo/metabolismo , Inflamación/metabolismo , Metabolismo de los Lípidos , Masculino , Ácido Palmítico/metabolismo , Alcamidas Poliinsaturadas , Vimentina/metabolismo
3.
Cells ; 10(3)2021 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800837

RESUMEN

Central actions of leptin and insulin on hepatic lipid metabolism can be opposing and the mechanism underlying this phenomenon remains unclear. Both hormones can modulate the central somatostatinergic system that has an inhibitory effect on growth hormone (GH) expression, which plays an important role in hepatic metabolism. Using a model of chronic central leptin infusion, we evaluated whether an increase in central leptin bioavailability modifies the serum lipid pattern through changes in hepatic lipid metabolism in male rats in response to an increase in central insulin and the possible involvement of the GH axis in these effects. We found a rise in serum GH in leptin plus insulin-treated rats, due to an increase in pituitary GH mRNA levels associated with lower hypothalamic somatostatin and pituitary somatostatin receptor-2 mRNA levels. An augment in hepatic lipolysis and a reduction in serum levels of non-esterified fatty acids (NEFA) and triglycerides were found in leptin-treated rats. These rats experienced a rise in lipogenic-related factors and normalization of serum levels of NEFA and triglycerides after insulin treatment. These results suggest that an increase in insulin in leptin-treated rats can act on the hepatic lipid metabolism through activation of the GH axis.


Asunto(s)
Hipotálamo/efectos de los fármacos , Insulina/farmacología , Leptina/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hipófisis/efectos de los fármacos , Animales , Ácidos Grasos no Esterificados/sangre , Regulación de la Expresión Génica , Hormona del Crecimiento/genética , Hormona del Crecimiento/metabolismo , Hipotálamo/metabolismo , Inyecciones Intravenosas , Inyecciones Intraventriculares , Insulina/metabolismo , Leptina/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Masculino , Hipófisis/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Transducción de Señal , Triglicéridos/sangre
4.
Nutrients ; 13(3)2021 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-33804764

RESUMEN

The present study longitudinally evaluated growth, bone mineral density, body composition, and metabolic health outcome in very low birth weight (VLBW) infants whose in-hospital target nutrient intake was within recent recommendations. From six months to three years, bone mineral density (dual-energy X-ray absorptiometry, DXA), body composition, and metabolic health outcome were compared with a reference group of term infants. The aim was to test whether in-hospital achieved weight gain until 36 weeks of gestation (light or appropriate for term equivalent age; LTEA or ATEA) predicts later growth, bone mineral density (BMD), abdominal obesity, or metabolic health outcomes such as insulin resistance, relative to term infants, during the first three years of life. Target in-hospital energy and protein intake was not achieved. Growth in weight, length and head circumference, mid arm circumference, adiposity, fat free mass (FFM), and bone mineralization in VLBW infants was less than those in term infants and influenced by nutritional status at discharge. Preterm infants had poorer motor and cognitive outcomes. Post-discharge body composition patterns indicate FFM proportional to height but lower fat mass index in LTEA preterm infants than term infants, with no evidence of increased truncal fat in preterm infants. The hypothesis of early BMD catch-up in VLBW infants after discharge was not supported by the present data. The clinical significance of these findings is unclear. The data may suggest a reduced obesity risk but an increased osteoporosis risk. Since postnatal growth restriction may have permanent negative health effects, LTEA VLBW infants would especially appear to benefit from targeted preventive interventions. Further follow-up of the infants is required.


Asunto(s)
Composición Corporal/fisiología , Densidad Ósea/fisiología , Fórmulas Infantiles , Fenómenos Fisiológicos Nutricionales del Lactante/fisiología , Factor I del Crecimiento Similar a la Insulina/metabolismo , Nutrientes/administración & dosificación , Absorciometría de Fotón , Desarrollo Infantil , Preescolar , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Hospitalización , Humanos , Lactante , Recien Nacido Prematuro , Recién Nacido de muy Bajo Peso , Estudios Longitudinales , Masculino , Estudios Prospectivos , Ingesta Diaria Recomendada , Aumento de Peso
5.
Int J Mol Sci ; 22(6)2021 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33799501

RESUMEN

The growth hormone (GH)/insulin-like growth factor I (IGF-I) axis is involved in metabolic control. Malnutrition reduces IGF-I and modifies the thermogenic capacity of brown adipose tissue (BAT). Leptin has effects on the GH/IGF-I axis and the function of BAT, but its interaction with IGF-I and the mechanisms involved in the regulation of thermogenesis remains unknown. We studied the GH/IGF-I axis and activation of IGF-I-related signaling and metabolism related to BAT thermogenesis in chronic central leptin infused (L), pair-fed (PF), and control rats. Hypothalamic somatostatin mRNA levels were increased in PF and decreased in L, while pituitary GH mRNA was reduced in PF. Serum GH and IGF-I concentrations were decreased only in PF. In BAT, the association between suppressor of cytokine signaling 3 and the IGF-I receptor was reduced, and phosphorylation of the IGF-I receptor increased in the L group. Phosphorylation of Akt and cyclic AMP response element binding protein and glucose transporter 4 mRNA levels were increased in L and mRNA levels of uncoupling protein-1 (UCP-1) and enzymes involved in lipid anabolism reduced in PF. These results suggest that modifications in UCP-1 in BAT and changes in the GH/IGF-I axis induced by negative energy balance are dependent upon leptin levels.


Asunto(s)
Tejido Adiposo Pardo/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Hormona del Crecimiento/genética , Factor I del Crecimiento Similar a la Insulina/genética , Leptina/farmacología , Termogénesis/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Metabolismo Energético/genética , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Hormona del Crecimiento/metabolismo , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Inyecciones Intraventriculares , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Fosforilación/efectos de los fármacos , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptor IGF Tipo 1/genética , Receptor IGF Tipo 1/metabolismo , Somatostatina/genética , Somatostatina/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo , Termogénesis/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
6.
Front Endocrinol (Lausanne) ; 12: 796661, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975768

RESUMEN

Dietary intervention is a common tactic employed to curtail the current obesity epidemic. Changes in nutritional status alter metabolic hormones such as insulin or leptin, as well as the insulin-like growth factor (IGF) system, but little is known about restoration of these parameters after weight loss in obese subjects and if this differs between the sexes, especially regarding the IGF system. Here male and female mice received a high fat diet (HFD) or chow for 8 weeks, then half of the HFD mice were changed to chow (HFDCH) for 4 weeks. Both sexes gained weight (p < 0.001) and increased their energy intake (p < 0.001) and basal glycemia (p < 0.5) on the HFD, with these parameters normalizing after switching to chow but at different rates in males and females. In both sexes HFD decreased hypothalamic NPY and AgRP (p < 0.001) and increased POMC (p < 0.001) mRNA levels, with all normalizing in HFDCH mice, whereas the HFD-induced decrease in ObR did not normalize (p < 0.05). All HFD mice had abnormal glucose tolerance tests (p < 0.001), with males clearly more affected, that normalized when returned to chow. HFD increased insulin levels and HOMA index (p < 0.01) in both sexes, but only HFDCH males normalized this parameter. Returning to chow normalized the HFD-induced increase in circulating leptin (p < 0.001), total IGF1 (p < 0.001), IGF2 (p < 0.001, only in females) and IGFBP3 (p < 0.001), whereas free IGF1 levels remained elevated (p < 0.01). In males IGFBP2 decreased with HFD and normalized with chow (p < 0.001), with no changes in females. Although returning to a healthy diet improved of most metabolic parameters analyzed, fIGF1 levels remained elevated and hypothalamic ObR decreased in both sexes. Moreover, there was sex differences in both the response to HFD and the switch to chow including circulating levels of IGF2 and IGFBP2, factors previously reported to be involved in glucose metabolism. Indeed, glucose metabolism was also differentially modified in males and females, suggesting that these observations could be related.


Asunto(s)
Dieta Alta en Grasa , Obesidad/dietoterapia , Obesidad/metabolismo , Pérdida de Peso/fisiología , Animales , Glucemia/análisis , Glucemia/metabolismo , Ingestión de Energía , Femenino , Hipotálamo/metabolismo , Resistencia a la Insulina , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Factor I del Crecimiento Similar a la Insulina/análisis , Factor II del Crecimiento Similar a la Insulina/análisis , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Caracteres Sexuales
7.
J Neuroendocrinol ; 32(1): e12756, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31179596

RESUMEN

Obesity is one of the most important health problems facing developed countries because being overweight is associated with a higher incidence of type 2 diabetes, cardiovascular disease and cancer, as well as other comorbidities. Although increased weight gain results from a combination of poor dietary habits and decreased energy expenditure, not all individuals have equal propensities to gain weight or to develop secondary complications of obesity. This is partially a result not only of genetics, including sex, but also the time during which an individual is exposed to an obesogenic environment. In the present study, we have compared the response of male and female mice to short-term exposure to a high-fat diet (HFD) or a low-fat diet during the peripubertal period (starting at 42 days of age) because this is a stage of dramatic hormonal and metabolic modifications. After 1 week on a HFD, there was no significant increase in body weight, although females significantly increased their energy intake. Serum leptin levels increased in both sexes, even though no change in fat mass was detected. Glyceamia and homeostasis model assessment increased in males, suggesting a rapid change in glucose metabolism. Hypothalamic pro-opiomelanocortin mRNA levels were significantly higher in females on a HFD compared to all other groups, which may be an attempt to reduce their increased energy intake. Hypothalamic inflammation and gliosis have been implicated in the development of secondary complications of obesity; however, no indication of activation of inflammatory processes or gliosis was found in response to 1 week of HFD in the hypothalamus, hippocampus or cerebellum of these young mice. These results indicate that there are both sex and age effects in the response to poor dietary intake because peripubertal male and female mice respond differently to short-term dietary changes and this response is different from that reported in adult rodents.


Asunto(s)
Glucemia/metabolismo , Peso Corporal/fisiología , Dieta Alta en Grasa , Hipotálamo/metabolismo , Maduración Sexual/fisiología , Adiposidad/fisiología , Animales , Ingestión de Energía/fisiología , Femenino , Insulina/sangre , Leptina/sangre , Masculino , Ratones , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Factores Sexuales
8.
Prog Neurobiol ; 176: 18-32, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30194984

RESUMEN

The hypothalamus is the main integrating center for metabolic control. Our understanding of how hypothalamic circuits function to control appetite and energy expenditure has increased dramatically in recent years, due to the rapid rise in the incidence of obesity and the search for effective treatments. Increasing evidence indicates that these treatments will most likely differ between males and females. Indeed, sex differences in metabolism have been demonstrated at various levels, including in two of the most studied neuronal populations involved in metabolic control: the anorexigenic proopiomelanocortin neurons and the orexigenic neuropeptide Y/Agouti-related protein neurons. Here we review what is known to date regarding the sex differences in these two neuronal populations, as well as other neuronal populations involved in metabolic control and glial cells.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Caracteres Sexuales , Animales , Astrocitos/metabolismo , Femenino , Humanos , Masculino , Neuronas/metabolismo
9.
Acta Physiol (Oxf) ; 226(2): e13244, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30589509

RESUMEN

AIM: We aimed to investigate whether a dysregulated maternal diet during gestation and lactation induces long-lasting changes in the hypothalamic control of feeding behavior in the offspring and whether this effect is sex specific. METHODS: The study included an analysis of appetite-regulating metabolic hormones and hypothalamic signaling in male and female offspring in adulthood after exposure to a free-choice high-calorie palatable low-protein (P) diet or standard chow (C) during (pre)gestation/lactation (maternal) and/or postweaning (offspring). RESULTS: Maternal exposure to the P diet resulted in decreased protein intake and body weight gain in dams and decreased body weight gain in offspring during lactation. The maternal P diet (PC) specifically increased feed efficacy and decreased body weight and cholesterol levels in the female offspring in adulthood, but no changes in adiposity or leptin levels were found. In contrast, P diet exposure after weaning (CP and PP) increased caloric intake, adiposity and circulating levels of leptin in the male and female offspring in adulthood. The hypothalami of the female offspring exposed to the maternal P diet (PC and PP) expressed high levels of the phospho-leptin receptor and low levels of SOCS3, phospho-IRS1 and phospho-AMPK, regardless of the postweaning diet. The hypothalami of the female rats in the PC group also showed increased levels of STAT3 and the orexigenic neuropeptide Agrp. CONCLUSIONS: Maternal exposure to a free-choice high-calorie low-protein diet induces a long-term feed efficacy associated with changes in leptin signaling through IRS-1 and AMPK dephosphorylation in the hypothalami of female offspring in adulthood.


Asunto(s)
Conducta Animal/efectos de los fármacos , Dieta con Restricción de Proteínas/efectos adversos , Leptina/farmacología , Efectos Tardíos de la Exposición Prenatal , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Conducta Animal/fisiología , Conducta Alimentaria/efectos de los fármacos , Conducta Alimentaria/fisiología , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Leptina/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Ratas Wistar , Receptores de Leptina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
10.
Front Neuroendocrinol ; 48: 3-12, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28552663

RESUMEN

Males and females have distinct propensities to develop obesity and its related comorbidities, partially due to gonadal steroids. There are sex differences in hypothalamic neuronal circuits, as well as in astrocytes, that participate in metabolic control and the development of obesity-associated complications. Astrocytes are involved in nutrient transport and metabolism, glucose sensing, synaptic remodeling and modulation of neuronal signaling. They express receptors for metabolic hormones and mediate effects of these metabolic signals on neurons, with astrogliosis occurring in response to high fat diet and excess weight gain. However, most studies of obesity have focused on males. Recent reports indicate that male and female astrocytes respond differently to metabolic signals and this could be involved in the differential response to high fat diet and the onset of obesity-associated pathologies. Here we focus on the sex differences in response to obesogenic paradigms and the possible role of hypothalamic astrocytes in this phenomenon.


Asunto(s)
Astrocitos/metabolismo , Hormonas Esteroides Gonadales/metabolismo , Hipotálamo/metabolismo , Obesidad/metabolismo , Caracteres Sexuales , Animales , Femenino , Humanos , Masculino
11.
Endocrinology ; 159(1): 368-387, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29077836

RESUMEN

Astrocytes participate in both physiological and pathophysiological responses to metabolic and nutrient signals. Although most studies have focused on the astrocytic response to weight gain due to high-fat/high-carbohydrate intake, surplus intake of a balanced diet also induces excess weight gain. We have accessed the effects of neonatal overnutrition, which has both age- and sex-dependent effects on weight gain, on hypothalamic inflammation/gliosis. Although both male and female Wistar rats accumulate excessive fat mass as early as postnatal day (PND) 10 with neonatal overnutrition, no increase in hypothalamic cytokine levels, markers of astrocytes or microglia, or inflammatory signaling pathways were observed. At PND 50, no effect of neonatal overnutriton was found in either sex, whereas at PND 150, males again weighed significantly more than their controls, and this was coincident with an increase in markers of inflammation and astrogliosis in the hypothalamus. Circulating triglycerides and free fatty acids were also elevated in these males, but not in females or in either sex at PND 10. Thus, the effects of fatty acids and estrogens on astrocytes in vitro were analyzed. Our results indicate that changes in circulating fatty acid levels may be involved in the induction of hypothalamic inflammation/gliosis in excess weight gain, even on a normal diet, and that estrogens could participate in the protection of females from these processes. In conclusion, the interaction of developmental influences, dietary composition, age, and sex determines the central inflammatory response and the associated long-term outcomes of excess weight gain.


Asunto(s)
Astrocitos/metabolismo , Gliosis/etiología , Hiperfagia/fisiopatología , Enfermedades Hipotalámicas/etiología , Hipotálamo/metabolismo , Microglía/metabolismo , Adiposidad , Factores de Edad , Animales , Animales Recién Nacidos , Astrocitos/inmunología , Astrocitos/patología , Biomarcadores/metabolismo , Células Cultivadas , Citocinas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Gliosis/inmunología , Gliosis/metabolismo , Gliosis/patología , Enfermedades Hipotalámicas/inmunología , Enfermedades Hipotalámicas/metabolismo , Enfermedades Hipotalámicas/patología , Hipotálamo/inmunología , Hipotálamo/patología , Mediadores de Inflamación/metabolismo , Masculino , Microglía/inmunología , Microglía/patología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratas Wistar , Caracteres Sexuales , Transducción de Señal , Aumento de Peso
12.
J Mol Endocrinol ; 58(1): R59-R71, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27864453

RESUMEN

The search for new strategies and drugs to abate the current obesity epidemic has led to the intensification of research aimed at understanding the neuroendocrine control of appetite and energy expenditure. This intensified investigation of metabolic control has also included the study of how glial cells participate in this process. Glia, the most abundant cell type in the central nervous system, perform a wide spectrum of functions and are vital for the correct functioning of neurons and neuronal circuits. Current evidence indicates that hypothalamic glia, in particular astrocytes, tanycytes and microglia, are involved in both physiological and pathophysiological mechanisms of appetite and metabolic control, at least in part by regulating the signals reaching metabolic neuronal circuits. Glia transport nutrients, hormones and neurotransmitters; they secrete growth factors, hormones, cytokines and gliotransmitters and are a source of neuroprogenitor cells. These functions are regulated, as glia also respond to numerous hormones and nutrients, with the lack of specific hormonal signaling in hypothalamic astrocytes disrupting metabolic homeostasis. Here, we review some of the more recent advances in the role of glial cells in metabolic control, with a special emphasis on the differences between glial cell responses in males and females.


Asunto(s)
Metabolismo Energético , Neuroglía/metabolismo , Animales , Astrocitos/metabolismo , Células Ependimogliales/metabolismo , Femenino , Gliosis/metabolismo , Gliosis/patología , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Masculino , Microglía/metabolismo , Neuronas/metabolismo , Factores Sexuales
13.
Peptides ; 86: 63-71, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27751931

RESUMEN

The neonatal leptin surge, occurring from postnatal day (PND) 5 to 13 and peaking at PND9 in rodents, is important for the development of neuroendocrine circuits involved in metabolic control and reproductive function. We previously demonstrated that treatment with a leptin antagonist from PND 5 to 9, coincident with peak leptin levels in the neonatal surge, modified trophic factors and markers of cell turnover and neuronal maturation in the hypothalamus of peri-pubertal rats. The kisspeptin system and metabolic neuropeptide and hormone levels were also modified. Here our aim was to investigate if the timing of pubertal onset is altered by neonatal leptin antagonism and if the previously observed peripubertal modifications in hormones and neuropeptides persist into adulthood and affect male sexual behavior. To this end, male Wistar rats were treated with a pegylated super leptin antagonist (5mg/kg, s.c.) from PND 5 to 9 and killed at PND102-103. The appearance of external signs of pubertal onset was delayed. Hypothalamic kiss1 mRNA levels were decreased in adult animals, but sexual behavior was not significantly modified. Although there was no effect on body weight or food intake, circulating leptin, insulin and triglyceride levels were increased, while hypothalamic leptin receptor, POMC and AgRP mRNA levels were decreased. In conclusion, alteration of the neonatal leptin surge can modify the timing of pubertal onset and have long-term effects on hypothalamic expression of reproductive and metabolic neuropeptides.


Asunto(s)
Leptina/fisiología , Maduración Sexual , Animales , Animales Recién Nacidos , Expresión Génica , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuropéptido Y/genética , Neuropéptido Y/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo , Ratas Wistar , Receptores de Leptina/metabolismo , Conducta Sexual Animal , Transducción de Señal , Aumento de Peso
14.
Dis Model Mech ; 9(5): 573-83, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-27013528

RESUMEN

Insulin receptor substrate-2-deficient (IRS2(-/-)) mice are considered a good model to study the development of diabetes because IRS proteins mediate the pleiotropic effects of insulin-like growth factor-I (IGF-I) and insulin on metabolism, mitogenesis and cell survival. The hypothalamus might play a key role in the early onset of diabetes, owing to its involvement in the control of glucose homeostasis and energy balance. Because some inflammatory markers are elevated in the hypothalamus of diabetic IRS2(-/-) mice, our aim was to analyze whether the diabetes associated with the absence of IRS2 results in hypothalamic injury and to analyze the intracellular mechanisms involved. Only diabetic IRS2(-/-) mice showed increased cell death and activation of caspase-8 and -3 in the hypothalamus. Regulators of apoptosis such as FADD, Bcl-2, Bcl-xL and p53 were also increased, whereas p-IκB and c-FLIPL were decreased. This was accompanied by increased levels of Nox-4 and catalase, enzymes involved in oxidative stress. In summary, the hypothalamus of diabetic IRS2(-/-) mice showed an increase in oxidative stress and inflammatory markers that finally resulted in cell death via substantial activation of the extrinsic apoptotic pathway. Conversely, non-diabetic IRS2(-/-) mice did not show cell death in the hypothalamus, possibly owing to an increase in the levels of circulating IGF-I and in the enhanced hypothalamic IGF-IR phosphorylation that would lead to the stimulation of survival pathways. In conclusion, diabetes in IRS2-deficient male mice is associated with increased oxidative stress and apoptosis in the hypothalamus.


Asunto(s)
Apoptosis , Diabetes Mellitus Experimental/patología , Hipotálamo/patología , Proteínas Sustrato del Receptor de Insulina/deficiencia , Estrés Oxidativo , Animales , Biomarcadores/metabolismo , Caspasas/metabolismo , Citocinas/metabolismo , Inflamación/patología , Proteínas Sustrato del Receptor de Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptor IGF Tipo 1/metabolismo
15.
Sci Rep ; 6: 23673, 2016 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-27026049

RESUMEN

Hypothalamic astrocytes can respond to metabolic signals, such as leptin and insulin, to modulate adjacent neuronal circuits and systemic metabolism. Ghrelin regulates appetite, adiposity and glucose metabolism, but little is known regarding the response of astrocytes to this orexigenic hormone. We have used both in vivo and in vitro approaches to demonstrate that acylated ghrelin (acyl-ghrelin) rapidly stimulates glutamate transporter expression and glutamate uptake by astrocytes. Moreover, acyl-ghrelin rapidly reduces glucose transporter (GLUT) 2 levels and glucose uptake by these glial cells. Glutamine synthetase and lactate dehydrogenase decrease, while glycogen phosphorylase and lactate transporters increase in response to acyl-ghrelin, suggesting a change in glutamate and glucose metabolism, as well as glycogen storage by astrocytes. These effects are partially mediated through ghrelin receptor 1A (GHSR-1A) as astrocytes do not respond equally to desacyl-ghrelin, an isoform that does not activate GHSR-1A. Moreover, primary astrocyte cultures from GHSR-1A knock-out mice do not change glutamate transporter or GLUT2 levels in response to acyl-ghrelin. Our results indicate that acyl-ghrelin may mediate part of its metabolic actions through modulation of hypothalamic astrocytes and that this effect could involve astrocyte mediated changes in local glucose and glutamate metabolism that alter the signals/nutrients reaching neighboring neurons.


Asunto(s)
Astrocitos/metabolismo , Ghrelina/fisiología , Transportador de Glucosa de Tipo 2/metabolismo , Receptores de Ghrelina/metabolismo , Animales , Células Cultivadas , Glucosa/metabolismo , Ácido Glutámico/metabolismo , Hipotálamo/citología , Masculino , Ratones Noqueados , Ratas Wistar , Receptores de Ghrelina/genética
16.
Prog Neurobiol ; 144: 68-87, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27000556

RESUMEN

The hypothalamus is crucial in the regulation of homeostatic functions in mammals, with the disruption of hypothalamic circuits contributing to chronic conditions such as obesity, diabetes mellitus, hypertension, and infertility. Metabolic signals and hormonal inputs drive functional and morphological changes in the hypothalamus in attempt to maintain metabolic homeostasis. However, the dramatic increase in the incidence of obesity and its secondary complications, such as type 2 diabetes, have evidenced the need to better understand how this system functions and how it can go awry. Growing evidence points to a critical role of astrocytes in orchestrating the hypothalamic response to metabolic cues by participating in processes of synaptic transmission, synaptic plasticity and nutrient sensing. These glial cells express receptors for important metabolic signals, such as the anorexigenic hormone leptin, and determine the type and quantity of nutrients reaching their neighboring neurons. Understanding the mechanisms by which astrocytes participate in hypothalamic adaptations to changes in dietary and metabolic signals is fundamental for understanding the neuroendocrine control of metabolism and key in the search for adequate treatments of metabolic diseases.


Asunto(s)
Astrocitos/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Hipotálamo/metabolismo , Obesidad/metabolismo , Transducción de Señal/fisiología , Animales , Humanos
17.
Endocrinology ; 156(7): 2571-81, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25856428

RESUMEN

Leptin (Lep) is important in the development of neuroendocrine circuits involved in metabolic control. Because both Lep and metabolism influence pubertal development, we hypothesized that early changes in Lep signaling could also modulate hypothalamic (HT) systems involved in reproduction. We previously demonstrated that a single injection of a Lep antagonist (Antag) on postnatal day (PND)9, coincident with the neonatal Lep peak, induced sexually dimorphic modifications in trophic factors and markers of cell turnover and neuronal maturation in the HT on PND13. Here, our aim was to investigate whether the alterations induced by Lep antagonism persist into puberty. Accordingly, male and female rats were treated with a pegylated super Lep Antag from PND5 to PND9 and killed just before the normal appearance of external signs of puberty (PND33 in females and PND43 in males). There was no effect on body weight, but in males food intake increased, subcutaneous adipose tissue decreased and HT neuropeptide Y and Agouti-related peptide mRNA levels were reduced, with no effect in females. In both sexes, the Antag increased HT mRNA levels of the kisspeptin receptor, G protein-coupled recepter 54 (Gpr54). Expression of the Lep receptor, trophic factors, and glial markers were differently affected in the HT of peripubertal males and females. Lep production in adipose tissue was decreased in Antag-treated rats of both sexes, with production of other cytokines being differentially regulated between sexes. In conclusion, in addition to the long-term effects on metabolism, changes in neonatal Lep levels modifies factors involved in reproduction that could possibly affect sexual maturation.


Asunto(s)
Tejido Adiposo/metabolismo , Proteína Relacionada con Agouti/genética , Hipotálamo/metabolismo , Leptina/antagonistas & inhibidores , Neuropéptido Y/genética , ARN Mensajero/metabolismo , Maduración Sexual/genética , Animales , Animales Recién Nacidos , Peso Corporal/genética , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Citocinas/metabolismo , Ingestión de Alimentos/genética , Femenino , Hormona Folículo Estimulante/genética , Perfilación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/genética , Péptidos y Proteínas de Señalización Intercelular/genética , Leptina/genética , Hormona Luteinizante/genética , Masculino , Neuropéptidos/genética , Neuropéptidos/metabolismo , Ratas , Receptores de Leptina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores Sexuales , Grasa Subcutánea , Vimentina/genética
18.
Endocrinology ; 156(4): 1272-82, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25574789

RESUMEN

Pubertal onset may be advanced by obesity, with leptin potentially acting as a permissive factor. We hypothesized that having increased body weight (BW) prepubertally affects the ability of leptin to activate intracellular signaling pathways and modulate the expression of hypothalamic neuropeptides involved in reproduction and metabolism. Because being raised in small litters (SLs) tends to increase BW at weaning, female rats were raised in litters of 4 or large litters (LLs) of 12 pups. Leptin (3 µg/g BW) or vehicle (saline) was injected sc at postnatal day (PND) 21 and 30. Rats raised in SLs weighed more at both ages, but relative visceral and subcutaneous fat was increased only on PND21. Serum leptin levels were not different at PND21 or PND30. At PND21, key elements of intracellular leptin signaling (phosphorylated signal transducer and activator of transcription 3 and phosphorylated Akt [p-Akt]) were lower in SL than in LL rats. Leptin injection stimulated phosphorylated signal transducer and activator of transcription 3 in both groups, with a greater increase in LL, whereas p-Akt rose only in SL rats. At PND30, basal leptin signaling did not differ between LL and SL rats. Leptin activation of Akt was similar at 45 minutes, but at 2 hours p-AKT levels were higher in SL than in LL rats, as was the decrease in neuropeptide Y mRNA and increase in pro-opiomelanocortin mRNA levels. No change in the reproductive axis was found. Thus, being raised in SLs increases BW and visceral body fat content, fails to increase plasma leptin concentrations, and increases the leptin responsiveness of both neuropeptide Y and pro-opiomelanocortin cells in the prepubertal hypothalamus.


Asunto(s)
Peso Corporal/fisiología , Leptina/farmacología , Neuronas/metabolismo , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , Maduración Sexual/fisiología , Animales , Femenino , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Leptina/sangre , Neuronas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptores de Leptina/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
19.
Endocrinology ; 155(12): 4856-67, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25237935

RESUMEN

GH is important in metabolic control, and mice with disruption of the gene encoding the GH receptor (GHR) and GH binding protein (GHR-/- mice) are dwarf with low serum IGF-1 and insulin levels, high GH levels, and increased longevity, despite their obesity and altered lipid and metabolic profiles. Secondary complications of high-fat diet (HFD)-induced obesity are reported to be associated with hypothalamic inflammation and gliosis. Because GH and IGF-1 can modulate inflammatory processes, our objective was to evaluate the effect of HFD on hypothalamic inflammation/gliosis in the absence of GH signaling and determine how this correlates with changes in systemic metabolism. On normal chow, GHR-/- mice had a higher percentage of fat mass and increased circulating nonesterified free fatty acids levels compared with wild type (WT), and this was associated with increased hypothalamic TNF-α and phospho-JNK levels. After 7 weeks on a HFD, both WT and GHR-/- mice had increased weight gain, with GHR-/- mice having a greater rise in their percentage of body fat. In WT mice, HFD-induced weight gain was associated with increased hypothalamic levels of phospho-JNK and the microglial marker Iba-1 (ionized calcium-binding adapter molecule 1) but decreased cytokine production. Moreover, in GHR-/- mice, the HFD decreased hypothalamic inflammatory markers to WT levels with no indication of gliosis. Thus, the GH/IGF-1 axis is important in determining not only adipose tissue accrual but also the inflammatory response to HFD. However, how hypothalamic inflammation/gliosis is defined will determine whether it can be considered a common feature of HFD-induced obesity.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Gliosis/etiología , Hormona del Crecimiento/deficiencia , Enfermedades Hipotalámicas/etiología , Inflamación/etiología , Animales , Glucemia , Composición Corporal , Tamaño Corporal , Citocinas/sangre , Enfermedades Hipotalámicas/sangre , Hipotálamo/metabolismo , Inflamación/sangre , Insulina/sangre , Factor I del Crecimiento Similar a la Insulina/metabolismo , Lípidos/sangre , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/sangre
20.
Nat Neurosci ; 17(7): 908-10, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24880214

RESUMEN

We found that leptin receptors were expressed in hypothalamic astrocytes and that their conditional deletion led to altered glial morphology and synaptic inputs onto hypothalamic neurons involved in feeding control. Leptin-regulated feeding was diminished, whereas feeding after fasting or ghrelin administration was elevated in mice with astrocyte-specific leptin receptor deficiency. These data reveal an active role of glial cells in hypothalamic synaptic remodeling and control of feeding by leptin.


Asunto(s)
Astrocitos/fisiología , Ingestión de Alimentos/fisiología , Hipotálamo/fisiología , Leptina/fisiología , Red Nerviosa/fisiología , Neuronas/fisiología , Transducción de Señal/fisiología , Animales , Recuento de Células , Potenciales Postsinápticos Excitadores/fisiología , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Hipotálamo/citología , Inmunohistoquímica , Hibridación in Situ , Leptina/genética , Masculino , Melanocortinas/fisiología , Ratones , Ratones Noqueados , Microscopía Electrónica , Cultivo Primario de Células , Proopiomelanocortina/fisiología , Intercambio Gaseoso Pulmonar/fisiología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA