Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 382
Filtrar
Más filtros

Medicinas Complementárias
País/Región como asunto
Intervalo de año de publicación
1.
Biol Psychiatry ; 91(10): 856-859, 2022 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-35369984

RESUMEN

Neuronal circuits within the hypothalamus play a critical role in the homeostatic regulation of body weight. By disrupting the development or function of these circuits, human monogenic disorders cause hyperphagia (increased food intake), neuroendocrine abnormalities, impaired sympathetic nervous system activation, and obesity. Some genetic disorders also cause maladaptive behaviors such as anxiety, autism, emotional lability, and aggression, highlighting the role of the specific molecules expressed by these hypothalamic neurons in the regulation of innate behaviors that are essential to survival. These findings inform understanding of a wide range of clinical disorders and highlight the challenges associated with targeting these hypothalamic pathways for weight loss therapy.


Asunto(s)
Apetito , Hipotálamo , Peso Corporal , Humanos , Hipotálamo/metabolismo , Leptina/fisiología , Obesidad , Síndrome
2.
Front Endocrinol (Lausanne) ; 12: 694204, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34367066

RESUMEN

The 5-hydroxytryptamine 2C receptor (5-HTR2C) is a class G protein-coupled receptor (GPCR) enriched in the hypothalamus and the brain stem, where it has been shown to regulate energy homeostasis, including feeding and glucose metabolism. Accordingly, 5-HTR2C has been the target of several anti-obesity drugs, though the associated side effects greatly curbed their clinical applications. Dissecting the specific neural circuits of 5-HTR2C-expressing neurons and the detailed molecular pathways of 5-HTR2C signaling in metabolic regulation will help to develop better therapeutic strategies towards metabolic disorders. In this review, we introduced the regulatory role of 5-HTR2C in feeding behavior and glucose metabolism, with particular focus on the molecular pathways, neural network, and its interaction with other metabolic hormones, such as leptin, ghrelin, insulin, and estrogens. Moreover, the latest progress in the clinical research on 5-HTR2C agonists was also discussed.


Asunto(s)
Encéfalo/fisiología , Metabolismo Energético/genética , Receptor de Serotonina 5-HT2C/fisiología , Animales , Encéfalo/metabolismo , Estrógenos/fisiología , Ghrelina/fisiología , Homeostasis/genética , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiología , Insulina/fisiología , Leptina/fisiología , Red Nerviosa/fisiología , Receptor de Serotonina 5-HT2C/metabolismo , Transducción de Señal/genética
3.
Front Endocrinol (Lausanne) ; 12: 585887, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34084149

RESUMEN

The peptide hormone leptin regulates food intake, body mass, and reproductive function and plays a role in fetal growth, proinflammatory immune responses, angiogenesis and lipolysis. Leptin is a product of the obese (ob) gene and, following synthesis and secretion from fat cells in white adipose tissue, binds to and activates its cognate receptor, the leptin receptor (LEP-R). LEP-R distribution facilitates leptin's pleiotropic effects, playing a crucial role in regulating body mass via a negative feedback mechanism between adipose tissue and the hypothalamus. Leptin resistance is characterized by reduced satiety, over-consumption of nutrients, and increased total body mass. Often this leads to obesity, which reduces the effectiveness of using exogenous leptin as a therapeutic agent. Thus, combining leptin therapies with leptin sensitizers may help overcome such resistance and, consequently, obesity. This review examines recent data obtained from human and animal studies related to leptin, its role in obesity, and its usefulness in obesity treatment.


Asunto(s)
Leptina/fisiología , Obesidad/etiología , Animales , Metabolismo Energético/fisiología , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiopatología , Leptina/sangre , Obesidad/epidemiología , Obesidad/metabolismo , Factores de Riesgo , Respuesta de Saciedad/fisiología , Transducción de Señal
4.
Cell Metab ; 33(6): 1155-1170.e10, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33951475

RESUMEN

Pathologies of the micro- and macrovascular systems are a hallmark of the metabolic syndrome, which can lead to chronically elevated blood pressure. However, the underlying pathomechanisms involved still need to be clarified. Here, we report that an obesity-associated increase in serum leptin triggers the select expansion of the micro-angioarchitecture in pre-autonomic brain centers that regulate hemodynamic homeostasis. By using a series of cell- and region-specific loss- and gain-of-function models, we show that this pathophysiological process depends on hypothalamic astroglial hypoxia-inducible factor 1α-vascular endothelial growth factor (HIF1α-VEGF) signaling downstream of leptin signaling. Importantly, several distinct models of HIF1α-VEGF pathway disruption in astrocytes are protected not only from obesity-induced hypothalamic angiopathy but also from sympathetic hyperactivity or arterial hypertension. These results suggest that hyperleptinemia promotes obesity-induced hypertension via a HIF1α-VEGF signaling cascade in hypothalamic astrocytes while establishing a novel mechanistic link that connects hypothalamic micro-angioarchitecture with control over systemic blood pressure.


Asunto(s)
Astrocitos/metabolismo , Hipertensión/metabolismo , Hipotálamo/metabolismo , Leptina/fisiología , Obesidad/metabolismo , Animales , Astrocitos/patología , Femenino , Hipotálamo/patología , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Int J Mol Sci ; 22(9)2021 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-33924880

RESUMEN

The altered function of adipose tissue can result in obesity, insulin resistance, and its metabolic complications. Leptin, acting on the central nervous system, modifies the composition and function of adipose tissue. To date, the molecular changes that occur in epididymal white adipose tissue (eWAT) during chronic leptin treatment are not fully understood. Herein we aimed to address whether PPARß/δ could mediate the metabolic actions induced by leptin in eWAT. To this end, male 3-month-old Wistar rats, infused intracerebroventricularly (icv) with leptin (0.2 µg/day) for 7 days, were daily co-treated intraperitoneally (ip) without or with the specific PPARß/δ receptor antagonist GSK0660 (1 mg/kg/day). In parallel, we also administered GSK0660 to control rats fed ad libitum without leptin infusion. Leptin, acting at central level, prevented the starvation-induced increase in circulating levels of FGF21, while induced markedly the endogenous expression of FGF21 and browning markers of eWAT. Interestingly, GSK0660 abolished the anorectic effects induced by icv leptin leading to increased visceral fat mass and reduced browning capacity. In addition, the pharmacological inhibition of PPARß/δ alters the immunomodulatory actions of central leptin on eWAT. In summary, our results demonstrate that PPARß/δ is involved in the up-regulation of FGF21 expression induced by leptin in visceral adipose tissue.


Asunto(s)
Tejido Adiposo Blanco/fisiología , Factores de Crecimiento de Fibroblastos/metabolismo , Leptina/fisiología , PPAR gamma/metabolismo , PPAR-beta/metabolismo , Animales , Hipotálamo/metabolismo , Infusiones Intraventriculares , Proteínas Klotho , Masculino , Proteínas de la Membrana/metabolismo , PPAR gamma/antagonistas & inhibidores , PPAR-beta/antagonistas & inhibidores , Ratas Wistar , Sulfonas , Tiofenos
6.
Reprod Biol Endocrinol ; 19(1): 12, 2021 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-33472656

RESUMEN

BACKGROUND: Energy balance is closely related to reproductive function, wherein hypothalamic kisspeptin mediates regulation of the energy balance. However, the central mechanism of kisspeptin in the regulation of male reproductive function under different energy balance states is unclear. Here, high-fat diet (HFD) and exercise were used to change the energy balance to explore the role of leptin and inflammation in the regulation of kisspeptin and the hypothalamic-pituitary-testis (HPT) axis. METHODS: Four-week-old male C57BL/6 J mice were randomly assigned to a normal control group (n = 16) or an HFD (n = 49) group. After 10 weeks of HFD feeding, obese mice were randomly divided into obesity control (n = 16), obesity moderate-load exercise (n = 16), or obesity high-load exercise (n = 17) groups. The obesity moderate-load exercise and obesity high-load exercise groups performed exercise (swimming) for 120 min/day and 120 min × 2 times/day (6 h interval), 5 days/week for 8 weeks, respectively. RESULTS: Compared to the mice in the normal group, in obese mice, the mRNA and protein expression of the leptin receptor, kiss, interleukin-10 (IL-10), and gonadotropin-releasing hormone (GnRH) decreased in the hypothalamus; serum luteinizing hormone (LH), follicle-stimulating hormone (FSH), and testosterone levels and sperm quality decreased; and serum leptin, estradiol, and tumor necrosis factor-α (TNF-α) levels and sperm apoptosis increased. Moderate- and high-load exercise effectively reduced body fat and serum leptin levels but had the opposite effects on the hypothalamus and serum IL-10 and TNF-α levels. Moderate-load exercise had anti-inflammatory effects accompanied by increased mRNA and protein expression of kiss and GnRH in the hypothalamus and increased serum FSH, LH, and testosterone levels and improved sperm quality. High-load exercise also promoted inflammation, with no significant effect on the mRNA and protein expression of kiss and GnRH in the hypothalamus, serum sex hormone level, or sperm quality. Moderate-load exercise improved leptin resistance and inflammation and reduced the inhibition of kisspeptin and the HPT axis in obese mice. The inflammatory response induced by high-load exercise may counteract the positive effect of improving leptin resistance on kisspeptin and HPT. CONCLUSION: During changes in energy balance, leptin and inflammation jointly regulate kisspeptin expression on the HPT axis.


Asunto(s)
Metabolismo Energético/fisiología , Mediadores de Inflamación/fisiología , Kisspeptinas/metabolismo , Leptina/fisiología , Reproducción/fisiología , Animales , Hipogonadismo/sangre , Hipogonadismo/complicaciones , Hipotálamo/metabolismo , Infertilidad Masculina/sangre , Infertilidad Masculina/etiología , Inflamación/sangre , Inflamación/complicaciones , Mediadores de Inflamación/sangre , Kisspeptinas/fisiología , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Transducción de Señal/fisiología
7.
Metab Syndr Relat Disord ; 18(9): 399-405, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32876506

RESUMEN

The adipokine leptin is expressed at higher concentrations in obese subjects, who also incidentally have a higher prevalence of hypertension. The pathogenesis of this obesity-related hypertension is controversial and is believed to be related to many factors including increased sympathetic activity, abnormalities of the renin-angiotensin system, sodium retention, and an endotheliopathy acting independently or in concert with increased circulating leptin. This review discusses the potential mechanisms through which changes in leptin signal transduction pathways in tissues with the leptin receptor, especially the hypothalamus, mediate the pathogenetic relationships between obesity and hypertension. The hypothesis is explored that leptin effects on blood pressure (BP) are meditated by the downstream effects of hypothalamic leptin signaling and ultimately result in activation of specific melanocortin receptors located on sympathetic neurons in the spinal cord. The physiological consequences of this sympathetic activation of the heart and kidney are activation of the renin-angiotensin system, sodium retention and circulatory expansion and finally, elevated BP. This sequence of events has been elegantly demonstrated with leptin infusion and gene knockout studies in animal models but has not been convincingly reproducibly confirmed in humans. Further studies in human subjects on the specific roles of hypothalamic leptin in essential hypertension are indicated as elucidation of the signaling pathways should provide better understanding of the role of weight loss in BP control and afford an additional mechanism for pharmacologic control of BP in adults and children at risk of cardiovascular disease.


Asunto(s)
Hipertensión/metabolismo , Leptina/fisiología , Obesidad/metabolismo , Animales , Eliminación de Gen , Genotipo , Humanos , Hipertensión/fisiopatología , Hipotálamo/metabolismo , Riñón/metabolismo , Leptina/sangre , Leptina/metabolismo , Ratones , Obesidad/fisiopatología , Proopiomelanocortina/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Médula Espinal/metabolismo , Sistema Nervioso Simpático
8.
Nutrients ; 12(7)2020 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-32630697

RESUMEN

Leptin is highly expressed in the placenta, mainly by trophoblastic cells, where it has an important autocrine trophic effect. Moreover, increased leptin levels are found in the most frequent pathology of pregnancy: gestational diabetes, where leptin may mediate the increased size of the placenta and the fetus, which becomes macrosomic. In fact, leptin mediates the increased protein synthesis, as observed in trophoblasts from gestational diabetic subjects. In addition, leptin seems to facilitate nutrients transport to the fetus in gestational diabetes by increasing the expression of the glycerol transporter aquaporin-9. The high plasma leptin levels found in gestational diabetes may be potentiated by leptin resistance at a central level, and obesity-associated inflammation plays a role in this leptin resistance. Therefore, the importance of anti-inflammatory nutrients to modify the pathology of pregnancy is clear. In fact, nutritional intervention is the first-line approach for the treatment of gestational diabetes mellitus. However, more nutritional intervention studies with nutraceuticals, such as polyphenols or polyunsaturated fatty acids, or nutritional supplementation with micronutrients or probiotics in pregnant women, are needed in order to achieve a high level of evidence. In this context, the Mediterranean diet has been recently found to reduce the risk of gestational diabetes in a multicenter randomized trial. This review will focus on the impact of maternal obesity on placental inflammation and nutrients transport, considering the mechanisms by which leptin may influence maternal and fetal health in this setting, as well as its role in pregnancy pathologies.


Asunto(s)
Diabetes Gestacional/fisiopatología , Leptina/fisiología , Estado Nutricional/fisiología , Antiinflamatorios/administración & dosificación , Diabetes Gestacional/patología , Diabetes Gestacional/terapia , Dieta Mediterránea , Femenino , Macrosomía Fetal/etiología , Macrosomía Fetal/fisiopatología , Humanos , Leptina/sangre , Terapia Nutricional , Obesidad/complicaciones , Placenta/patología , Embarazo , Complicaciones del Embarazo/fisiopatología , Trofoblastos/fisiología
9.
Mol Metab ; 37: 100994, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32278654

RESUMEN

OBJECTIVE: Sirt6 is an essential regulator of energy metabolism in multiple peripheral tissues. However, the direct role of Sirt6 in the hypothalamus, specifically pro-opiomelanocortin (POMC) neurons, controlling energy balance has not been established. Here, we aimed to determine the role of Sirt6 in hypothalamic POMC neurons in the regulation of energy balance and the underlying mechanisms. METHODS: For overexpression studies, the hypothalamic arcuate nucleus (ARC) of diet-induced obese mice was targeted bilaterally and adenovirus was delivered by using stereotaxic apparatus. For knockout studies, the POMC neuron-specific Sirt6 knockout mice (PKO mice) were generated. Mice were fed with chow diet or high-fat diet, and body weight and food intake were monitored. Whole-body energy expenditure was determined by metabolic cages. Parameters of body composition and glucose/lipid metabolism were evaluated. RESULTS: Sirt6 overexpression in the ARC ameliorated diet-induced obesity. Conversely, selective Sirt6 ablation in POMC neurons predisposed mice to obesity and metabolic disturbances. PKO mice showed an increased fat mass and food intake, while the energy expenditure was decreased. Mechanistically, Sirt6 could modulate leptin signaling in hypothalamic POMC neurons, with Sirt6 deficiency impairing leptin-induced phosphorylation of signal transducer and activator of transcription 3. The effects of leptin on reducing food intake and body weight and leptin-stimulated lipolysis were also impaired. Moreover, Sirt6 inhibition diminished the leptin-induced depolarization of POMC neurons. CONCLUSIONS: Our results reveal a key role of Sirt6 in POMC neurons against energy imbalance, suggesting that Sirt6 is an important molecular regulator for POMC neurons to promote negative energy balance.


Asunto(s)
Leptina/metabolismo , Neuronas/metabolismo , Sirtuinas/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/metabolismo , Composición Corporal , Peso Corporal , Encéfalo/metabolismo , Dieta Alta en Grasa , Metabolismo Energético/fisiología , Hipotálamo/metabolismo , Leptina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Obesidad/metabolismo , Proopiomelanocortina/metabolismo , Transducción de Señal/fisiología , Sirtuinas/fisiología
10.
Curr Mol Pharmacol ; 13(1): 17-30, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31339082

RESUMEN

BACKGROUND: Protein tyrosine phosphatases are enzymes which help in the signal transduction in diabetes, obesity, cancer, liver diseases and neurodegenerative diseases. PTP1B is the main member of this enzyme from the protein extract of human placenta. In phosphate inhibitors development, significant progress has been made over the last 10 years. In early-stage clinical trials, few compounds have reached whereas in the later stage trials or registration, yet none have progressed. Many researchers investigate different ways to improve the pharmacological properties of PTP1B inhibitors. OBJECTIVE: In the present review, authors have summarized various aspects related to the involvement of PTP1B in various types of signal transduction mechanisms and its prominent role in various diseases like cancer, liver diseases and diabetes mellitus. CONCLUSION: There are still certain challenges for the selection of PTP1B as a drug target. Therefore, continuous future efforts are required to explore this target for the development of PTP inhibitors to treat the prevailing diseases associated with it.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Terapia Molecular Dirigida , Proteína Tirosina Fosfatasa no Receptora Tipo 1/antagonistas & inhibidores , Animales , Antineoplásicos/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Diabetes Mellitus/enzimología , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/farmacocinética , Inhibidores Enzimáticos/uso terapéutico , Femenino , Predicción , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/fisiología , Leptina/fisiología , Ratones , Modelos Moleculares , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/enzimología , Fármacos Neuroprotectores/uso terapéutico , Placenta/enzimología , Embarazo , Conformación Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 1/química , Proteína Tirosina Fosfatasa no Receptora Tipo 1/fisiología , Transducción de Señal/efectos de los fármacos
11.
Nat Metab ; 1(4): 475-484, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-31535083

RESUMEN

Genome wide association studies (GWAS) in obesity have identified a large number of noncoding loci located near genes expressed in the central nervous system. However, due to the difficulties in isolating and characterizing specific neuronal subpopulations, few obesity-associated SNPs have been functionally characterized. Leptin responsive neurons in the hypothalamus are essential in controlling energy homeostasis and body weight. Here, we combine FACS-sorting of leptin-responsive hypothalamic neuron nuclei with genomic and epigenomic approaches (RNA-seq, ChIP-seq, ATAC-seq) to generate a comprehensive map of leptin-response specific regulatory elements, several of which overlap obesity-associated GWAS variants. We demonstrate the usefulness of our leptin-response neuron regulome, by functionally characterizing a novel enhancer near Socs3, a leptin response-associated transcription factor. We envision our data to serve as a useful resource and a blueprint for functionally characterizing obesity-associated SNPs in the hypothalamus.


Asunto(s)
Peso Corporal/genética , Epigenómica , Genómica , Leptina/fisiología , Animales , Estudio de Asociación del Genoma Completo , Humanos , Hipotálamo/metabolismo , Hipotálamo/fisiología , Ratones , Neuronas/fisiología , Obesidad/genética , Polimorfismo de Nucleótido Simple , Transcriptoma
12.
Brain Res ; 1724: 146441, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31513793

RESUMEN

Hunger resulting from food deprivation is associated with negative affect. This is supported by recent evidence showing that hunger-sensitive neurons drive feeding through a negative valence teaching signal. However, the complementary hypothesis that hormonal signals of energy surfeit counteract this negative valence, or even transmit positive valence, has received less attention. The adipose-derived hormone leptin signals in proportion to fat mass, is an indicator of energy surplus, and reduces food intake. Here, we showed that centrally-delivered leptin reduced food intake and conditioned a place preference in food-restricted as well as ad libitum fed rats. In contrast, leptin did not reduce food intake nor condition a place preference in obese rats, likely due to leptin resistance. Despite a well-known role for hindbrain leptin receptor signaling in energy balance control, hindbrain leptin delivery did not condition a place preference in food-restricted rats, suggesting that leptin acting in midbrain or forebrain sites mediates place preference conditioning. Supporting the hypothesis that leptin signaling induces a positive affective state, leptin also decreased the threshold for ventral tegmental area brain stimulation reward. Together, these data suggest that leptin signaling is intrinsically preferred, and support the view that signals of energy surfeit are associated with positive affect. Harnessing the positive valence of signals such as leptin may attenuate the negative affect associated with hunger, providing a compelling new approach for weight loss maintenance.


Asunto(s)
Ingestión de Alimentos/efectos de los fármacos , Conducta Alimentaria/fisiología , Leptina/metabolismo , Afecto/fisiología , Animales , Condicionamiento Clásico/fisiología , Emociones/fisiología , Metabolismo Energético/fisiología , Conducta Alimentaria/efectos de los fármacos , Alimentos , Privación de Alimentos/fisiología , Leptina/fisiología , Masculino , Obesidad , Ratas , Ratas Sprague-Dawley , Receptores de Leptina/metabolismo , Recompensa , Rombencéfalo/metabolismo , Transducción de Señal/efectos de los fármacos , Área Tegmental Ventral/metabolismo
13.
FASEB J ; 33(11): 12175-12187, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31366239

RESUMEN

Synchronization between biologic clocks and metabolism is crucial for most species. Here, we examined the ability of leptin, important in the control of energy metabolism, to induce leptin signaling at the molecular as well as the behavioral level throughout the 24-h day in mice fed either a control or a high-fat diet (HFD). Furthermore, we investigated the effects of time-restricted feeding (TRF; a limitation of HFD access to 6 h each day) on energy metabolism during different periods throughout the 24-h day. In control mice, molecular leptin sensitivity was highest at zeitgeber time (ZT)0 (lights on), declining during the light phase, and increasing during the dark phase. Surprisingly, leptin resistance in HFD-fed mice was only present from the middle of the dark to the middle of the light period. Specifically, when TRF occurred from ZT21 to ZT3 (when leptin resistance in HFD-fed mice was most profound), it resulted in a disruption of the daily rhythms of locomotor activity and energy expenditure and in increased plasma insulin levels compared with other TRF periods. These data provide evidence that leptin sensitivity is controlled by the circadian rhythm and that TRF periods may be most efficient when aligned with the leptin-sensitive period.-Boucsein, A., Rizwan, M. Z., Tups, A. Hypothalamic leptin sensitivity and health benefits of time-restricted feeding are dependent on the time of day in male mice.


Asunto(s)
Ingestión de Alimentos , Metabolismo Energético , Hipotálamo/fisiología , Leptina/fisiología , Animales , Glucemia/análisis , Ritmo Circadiano , Dieta Alta en Grasa , Insulina/sangre , Leptina/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Consumo de Oxígeno , Factor de Transcripción STAT3/fisiología , Factores de Tiempo
14.
Mol Nutr Food Res ; 63(19): e1900110, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31298470

RESUMEN

SCOPE: To identify the age-dependent effect of diets containing elevated amounts of either saturated or unsaturated fatty acids on cardiac steatosis in mice. METHODS AND RESULTS: Five- and eight-week-old C57BL/6J mice cohorts are given free access to either a saturated or an unsaturated fatty-acid-enriched diet during 8 weeks. Body weight (BW) and food intake are monitored during this period. Cardiac lipid content, carnitine palmitoyltransferase-I (CPT-I) activity, and the amount of uncoupling proteins 2 and 3 (UCP2 and UCP3) are analyzed and correlated with blood leptin concentration. Leptin and PPARγ gene expression is quantified in white adipose tissue (WAT). Both diets have a similar effect on food intake, BW, and adiposity, independently of the age. Nevertheless, cardiac steatosis is specifically identified in adolescent mice consuming the saturated diet. These animals also display lower activity of cardiac CPT-I, a down-regulation of cardiac UCP2, together with lower concentration of plasma leptin. Accordingly, leptin gene expression is reduced in the visceral WAT. CONCLUSION: Consumption of diets containing elevated amounts of saturated fat during adolescence and early adult life promotes cardiac steatosis in mice. An insufficient endocrine activity of WAT, in terms of leptin production, may account for such an effect.


Asunto(s)
Envejecimiento , Enfermedades Cardiovasculares/etiología , Grasas de la Dieta/efectos adversos , Leptina/fisiología , Tejido Adiposo Blanco/química , Tejido Adiposo Blanco/metabolismo , Factores de Edad , Animales , Enfermedades Cardiovasculares/fisiopatología , Carnitina O-Palmitoiltransferasa/metabolismo , Grasas de la Dieta/administración & dosificación , Grasas Insaturadas en la Dieta/administración & dosificación , Regulación hacia Abajo/efectos de los fármacos , Ácidos Grasos/análisis , Leptina/genética , Lípidos/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Miocardio/química , Miocardio/metabolismo , PPAR gamma/genética , Aceite de Palma/administración & dosificación , Aceite de Palma/química , Proteína Desacopladora 2/genética
15.
Appl Physiol Nutr Metab ; 44(5): 507-511, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30286297

RESUMEN

Flaxseed is useful as a functional food and alternative medicine owing to its beneficial health effects. Its action on ovarian cell functions and interrelationships with the upstream hormonal regulators remain unknown. Our aim was to examine the direct influence of flaxseed extract on basal porcine ovarian functions (proliferation, apoptosis), leptin release, and response to insulin-like growth factor I (IGF-I). First, we examined the effect of flaxseed extract on the accumulation of proliferation (PCNA) and apoptosis (Bax) markers and on leptin release in cultured porcine ovarian granulosa cells. Next, granulosa cells were cultured with IGF-I with and without flaxseed extract and analyzed for PCNA and Bax accumulation by quantitative immunocytochemistry and for leptin release by radioimmunoassay. Flaxseed decreased the accumulation of PCNA and increased that of Bax at all doses and reduced leptin output at 100 µg/mL. In contrast, IGF-I promoted PCNA accumulation and suppressed Bax. Flaxseed did not modify IGF-I action on these parameters. Thus, we showed that flaxseed influences porcine reproductive processes, having a direct effect on the ovary and the ability to affect ovarian cell proliferation, apoptosis, and leptin release. Furthermore, we confirmed the pro-proliferative and antiapoptotic actions of IGF-I but showed that flaxseed action on ovarian cell proliferation and apoptosis is not due to changes in the cell response to IGF-I. The potential direct anti-reproductive action of flaxseed needs to be considered during its application in nutrition, medicine, and animal production.


Asunto(s)
Lino , Células de la Granulosa/fisiología , Ovario/citología , Extractos Vegetales/farmacología , Animales , Apoptosis , Proliferación Celular , Células Cultivadas , Femenino , Factor I del Crecimiento Similar a la Insulina/fisiología , Leptina/fisiología , Antígeno Nuclear de Célula en Proliferación/fisiología , Porcinos , Proteína X Asociada a bcl-2/fisiología
16.
Ann Endocrinol (Paris) ; 80(1): 38-46, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30243474

RESUMEN

The present review focused on the most important effects of leptin on the hypothalamus and on how leptin regulates neuropeptides associated with food intake and GnRH secretion. This review of the literature suggests that a reduction in leptin serum concentrations results from lower body energy reserves or poor energy availability, leading to hypothalamic secretion of neuropeptides such as NPY/AgRP and QRFP to stimulate food intake. Under these negative metabolic conditions, GnRH secretion is reduced, impairing reproductive functions. In contrast, when metabolic status is inversed by an increase in food availability, energy reserves or both, leptin serum concentrations increase to an action threshold reversing the pattern of secretion: i.e., reducing NPY/AgRP and QRFP and increasing POMC and Kisspeptin, and thereby reducing food intake and stimulating GnRH secretion to promote reproductive function.


Asunto(s)
Ingestión de Alimentos/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Leptina/fisiología , Neuropéptidos/fisiología , Proteína Relacionada con Agouti/metabolismo , Animales , Metabolismo Energético/fisiología , Homeostasis/fisiología , Humanos , Hipotálamo/fisiología , Kisspeptinas/metabolismo , Leptina/sangre , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , Reproducción/fisiología
17.
Horm Behav ; 106: 93-104, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30292429

RESUMEN

Leptin and insulin's hunger-suppressing and activity-promoting actions on hypothalamic neurons are well characterized, yet the mechanisms by which they modulate the midbrain dopamine system to influence energy balance remain less clear. A subset of midbrain dopamine neurons express receptors for leptin (Lepr) and insulin (Insr). Leptin-dopamine signaling reduces running reward and homecage activity. However, dopamine-specific deletion of Lepr does not affect body weight or food intake in mice. We hypothesized insulin-dopamine signaling might compensate for disrupted leptin-dopamine signaling. To investigate the degree to which insulin and leptin exert overlapping (i.e. redundant) versus discrete control over dopamine neurons, we generated transgenic male and female mice exhibiting dopamine-specific deletion of either Lepr (Lepr KO), Insr (Insr KO) or both Lepr and Insr (Dbl KO) and assessed their feeding behavior, voluntary activity, and energy expenditure compared to control mice. No differences in body weight, daily food intake, energy expenditure or hyperphagic feeding of palatable chow were observed between Lepr, Insr or Dbl KO mice and control mice. However, consistent with previous findings, Lepr KO (but not Insr or Dbl KO) male mice exhibited significantly increased running wheel activity compared to controls. These data demonstrate that insulin and leptin do not exert redundant control of dopamine neuron-mediated modulation of energy balance. Furthermore, our results indicate neither leptin nor insulin plays a critical role in the modulation of dopamine neurons regarding hedonic feeding behavior or anxiety-related behavior.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Emociones/fisiología , Metabolismo Energético/genética , Insulina/fisiología , Leptina/fisiología , Receptor de Insulina/genética , Receptores de Leptina/genética , Animales , Ansiedad/genética , Ansiedad/metabolismo , Peso Corporal/genética , Dopamina/metabolismo , Ingestión de Alimentos/genética , Conducta Alimentaria/fisiología , Femenino , Hipotálamo/metabolismo , Insulina/metabolismo , Leptina/metabolismo , Masculino , Mesencéfalo/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor de Insulina/metabolismo , Receptores de Leptina/metabolismo , Transducción de Señal/genética
18.
J Neuroendocrinol ; 30(11): e12646, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30246441

RESUMEN

We recently showed that male rats exhibit lower hypophagia and body weight loss compared to female rats following central leptin delivery, suggesting a role for oestradiol in leptin responsiveness. Accordingly, we delivered Ob (leptin) or GFP (control) gene into the brain of male rats that were simultaneously treated with oestradiol or vehicle. In a reciprocal approach, we compared oestradiol-deficient (OVX) with intact females (sham) that received leptin or control vector. Changes in food intake), body weight and body composition were examined. In males, oestradiol and leptin resulted in lower cumulative food intake (15%) and endpoint body weight (5%), although rats receiving dual treatment (oestradiol-leptin) ate 28% less and weighed 22% less than vehicle-control. Changes in food intake were unique to each treatment, with a rapid decrease in vehicle-leptin followed by gradual renormalisation. By contrast, hypophagia in oestradiol-control was of lower amplitude and sporadic. Leptin selectively targeted fat mass and endpoint abdominal fat mass was 65%-80% lower compared to their respective control groups. In females, both leptin groups had lower body weight (endpoint values 20% lower than control groups) with the highest extent in sham animals (endpoint value was 28% less in sham-leptin than in sham-control). OVX rats rapidly started regaining their lost body weight reminiscent of the pattern in males. Leptin rapidly and robustly reduced fat mass with endpoint values 30%-35% less than control treated animals. It appears that leptin and oestradiol decreased food intake and body weight via different mechanisms, with the pattern of oestradiol-leptin being reminiscent of that observed in females and the pattern of OVX-leptin reminiscent of that observed in males. Oestrogen status did not influence initial fat mass loss by leptin. It can be concluded that oestradiol modulates the long-term response to central leptin overexpression, although its actions on energy homeostasis are additive and independent of those of leptin.


Asunto(s)
Tejido Adiposo/fisiología , Ingestión de Alimentos/fisiología , Estradiol/fisiología , Hipotálamo/fisiología , Leptina/fisiología , Tejido Adiposo/efectos de los fármacos , Animales , Depresores del Apetito/administración & dosificación , Ingestión de Alimentos/efectos de los fármacos , Estradiol/administración & dosificación , Estrógenos/administración & dosificación , Estrógenos/fisiología , Femenino , Leptina/administración & dosificación , Leptina/genética , Masculino , Ovariectomía , Ratas Sprague-Dawley , Ratas Transgénicas , Caracteres Sexuales
19.
Yakugaku Zasshi ; 138(8): 1017-1024, 2018.
Artículo en Japonés | MEDLINE | ID: mdl-30068841

RESUMEN

Various neuropeptides play an essential role in the nutrient sensing mechanism and related homeostasis. Nesfatin-1 is a newly identified neuropeptide having anorectic activity, and nesfatin-1-containing neurons are widely distributed in the brain, including the hypothalamus and brain stem. Our previous study showed that dehydration-induced anorectic effects are mediated via the central nesfatin-1 pathway in rats. Our recent studies have also shown that peripheral anorectic peptides (cholecystokinin-8, glucagon-like peptide-1, and leptin) and an antineoplastic agent (cisplatin) caused inhibition of feeding via the central nesfatin-1 pathway in rats. Nesfatin-1-containing neurons in the central nervous system, in particular the hypothalamus and the brain stem, may mediate peripheral nutrient signals and regulate feeding behavior.


Asunto(s)
Anorexia/etiología , Anorexia/genética , Colecistoquinina/fisiología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/genética , Alimentos , Péptido 1 Similar al Glucagón/fisiología , Leptina/fisiología , Fenómenos Fisiológicos de la Nutrición/genética , Fenómenos Fisiológicos de la Nutrición/fisiología , Transducción de Señal/fisiología , Animales , Antineoplásicos/efectos adversos , Tronco Encefálico/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión al Calcio/fisiología , Colecistoquinina/metabolismo , Cisplatino/efectos adversos , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Péptido 1 Similar al Glucagón/metabolismo , Hipotálamo/metabolismo , Leptina/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/fisiología , Nucleobindinas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA