Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Vis Exp ; (193)2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-37010276

RESUMO

Kisspeptins are essential for the maturation of the hypothalamic-pituitary-gonadal (HPG) axis and fertility. Hypothalamic kisspeptin neurons located in the anteroventral periventricular nucleus and rostral periventricular nucleus, as well as the arcuate nucleus of the hypothalamus, project to gonadotrophin-releasing hormone (GnRH) neurons, among other cells. Previous studies have demonstrated that kisspeptin signaling occurs through the Kiss1 receptor (Kiss1r), ultimately exciting GnRH neuron activity. In humans and experimental animal models, kisspeptins are sufficient for inducing GnRH secretion and, consequently, luteinizing hormone (LH) and follicle stimulant hormone (FSH) release. Since kisspeptins play an essential role in reproductive functions, researchers are working to assess how the intrinsic activity of hypothalamic kisspeptin neurons contributes to reproduction-related actions and identify the primary neurotransmitters/neuromodulators capable of changing these properties. The whole-cell patch-clamp technique has become a valuable tool for investigating kisspeptin neuron activity in rodent cells. This experimental technique allows researchers to record and measure spontaneous excitatory and inhibitory ionic currents, resting membrane potential, action potential firing, and other electrophysiological properties of cell membranes. In the present study, crucial aspects of the whole-cell patch-clamp technique, known as electrophysiological measurements that define hypothalamic kisspeptin neurons, and a discussion of relevant issues about the technique, are reviewed.


Assuntos
Hipotálamo , Kisspeptinas , Humanos , Animais , Kisspeptinas/metabolismo , Técnicas de Patch-Clamp , Hipotálamo/metabolismo , Hormônio Liberador de Gonadotropina , Neurônios/fisiologia
2.
Int J Mol Sci ; 23(12)2022 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-35742928

RESUMO

Leptin resistance is a hallmark of obesity. Treatments aiming to improve leptin sensitivity are considered a promising therapeutical approach against obesity. However, leptin receptor (LepR) signaling also modulates several neurovegetative aspects, such as the cardiovascular system and hepatic gluconeogenesis. Thus, we investigated the long-term consequences of increased leptin sensitivity, considering the potential beneficial and deleterious effects. To generate a mouse model with increased leptin sensitivity, the suppressor of cytokine signaling 3 (SOCS3) was ablated in LepR-expressing cells (LepR∆SOCS3 mice). LepR∆SOCS3 mice displayed reduced food intake, body adiposity and weight gain, as well as improved glucose tolerance and insulin sensitivity, and were protected against aging-induced leptin resistance. Surprisingly, a very high mortality rate was observed in aging LepR∆SOCS3 mice. LepR∆SOCS3 mice showed cardiomyocyte hypertrophy, increased myocardial fibrosis and reduced cardiovascular capacity. LepR∆SOCS3 mice exhibited impaired post-ischemic cardiac functional recovery and middle-aged LepR∆SOCS3 mice showed substantial arhythmic events during the post-ischemic reperfusion period. Finally, LepR∆SOCS3 mice exhibited fasting-induced hypoglycemia and impaired counterregulatory response to glucopenia associated with reduced gluconeogenesis. In conclusion, although increased sensitivity to leptin improved the energy and glucose homeostasis of aging LepR∆SOCS3 mice, major autonomic/neurovegetative dysfunctions compromised the health and longevity of these animals. Consequently, these potentially negative aspects need to be considered in the therapies that increase leptin sensitivity chronically.


Assuntos
Cardiopatias , Receptores para Leptina , Animais , Metabolismo Energético , Glucose/metabolismo , Cardiopatias/metabolismo , Leptina/metabolismo , Camundongos , Neurônios/metabolismo , Obesidade/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/genética , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo
3.
J Endocrinol ; 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31470413

RESUMO

Growth hormone (GH) is a key factor in the regulation of body growth, as well as a variety of other cellular and metabolic processes. Neurons expressing kisspeptin and leptin receptors (LepR) have been shown to modulate the hypothalamic-pituitary-gonadal (HPG) axis and are considered GH-responsive. The presence of functional GH receptors (GHR) in these neural populations suggests that GH may regulate the HPG axis via a central mechanism. However, there have been no studies evaluating whether or not GH-induced intracellular signaling in the brain plays a role in the timing of puberty or mediates the ovulatory cycle. Towards the goal of understanding the influence of GH on the central nervous system as a mediator of reproductive functions, GHR ablation was induced in kisspeptin and LepR expressing cells or in the entire brain. The results demonstrated that GH signaling in specific neural populations can potentially modulate the hypothalamic expression of genes related to the reproductive system or indirectly contribute to the progression of puberty. GH action in kisspeptin cells or in the entire brain was not required for sexual maturation. On the other hand, GHR ablation in LepR cells delayed puberty progression, reduced serum leptin levels, decreased body weight gain and compromised the ovulatory cycle in some individuals, while the lack of GH effects in the entire brain prompted shorter estrous cycles. These findings suggest that GH can modulate brain components of the HPG axis, although central GH signaling is not required for the timing of puberty.

4.
Physiol Rep ; 7(13): e14107, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31264387

RESUMO

GABAergic inhibitory input within the paraventricular hypothalamic nucleus (PVN) plays a key role in restraining sympathetic outflow. Although experimental evidence has shown depressed GABAA receptor function plus sympathoexcitation in hypertension and augmented GABA levels with reduced sympathetic activity after exercise training (T), the mechanisms underlying T-induced effects remain unclear. Here we investigated in T and sedentary (S) SHR and WKY: (1) time-course changes of hemodynamic parameters and PVN glutamic acid decarboxylase (GAD) isoforms' expression; (2) arterial pressure (AP) and heart rate (HR) responses, sympathetic/parasympathetic modulation of heart and vessels and baroreflex sensitivity to GABAA receptor blockade within the PVN. SHR-S versus WKY-S exhibited higher AP and HR, increased sympathetic reduced parasympathetic modulation, smaller baroreflex sensitivity, and reduced PVN GAD65 immunoreactivity. SHR-T and WKY-T showed prompt maintained increase (2-8 weeks) in GAD65 expression (responsible for GABA vesicular pool synthesis), which occurred simultaneously with HR reduction in SHR-T and preceded MAP fall in SHR-T and resting bradycardia in WKY-T. There was no change in GAD67 expression (mainly involved with GABA metabolic pool). Resting HR in both groups and basal MAP in SHR were negatively correlated with PVN GAD65 expression. Normalized baroreflex sensitivity and autonomic control observed only in SHR-T were due to recovery of GABAA receptor function into the PVN since bicuculline administration abolished these effects. Data indicated that training augments in both groups the expression/activity of GABAergic neurotransmission within presympathetic PVN neurons and restores GABAA receptors' function specifically in the SHR, therefore strengthening GABAergic modulation of sympathetic outflow in hypertension.


Assuntos
Glutamato Descarboxilase/genética , Hipertensão/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Esforço Físico , Receptores de GABA-A/metabolismo , Animais , Barorreflexo , Pressão Sanguínea , Glutamato Descarboxilase/metabolismo , Hipertensão/fisiopatologia , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratos , Ratos Endogâmicos SHR , Ratos Wistar , Sistema Nervoso Simpático/fisiopatologia
5.
Brain Res ; 1714: 210-217, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30851245

RESUMO

Growth hormone (GH) and prolactin (PRL) are known as pleiotropic hormones. Accordingly, the distribution of their receptors comprises several organs and tissues, including the central nervous system. The appropriate secretion of both hormones is essential for sexual maturation and maintenance of reproductive functions, while defects in their secretion affect puberty onset and can cause infertility. Conversely, GH therapy at a prepubertal age may accelerate puberty. On the other hand, hyperprolactinemia is a frequent cause of infertility. While the action of PRL in some central components of the Hypothalamic-Pituitary-Gonadal (HPG) axis, such as the kisspeptin neurons, has been well documented, the possible effects of GH in the hypothalamus are still elusive. Thus, the present study was designed to investigate whether somatomammotropin hormones are able to modulate the activity of critical neuronal components of the HPG axis, including kisspeptin neurons and cells of the ventral premammillary nucleus (PMv). Our results revealed that GH effects in kisspeptin neurons of the anteroventral periventricular and rostral periventricular nuclei or in PMv neurons relies predominantly on the recruitment of the signal transducer and activator of transcription 5 (STAT5) rather than through acute changes in resting membrane potential. Importantly, kisspeptin neurons located at the arcuate nucleus were not directly responsive to GH. Additionally, our findings further identified PMv neurons as potential targets of PRL, since PRL induces the phosphorylation of STAT5 and depolarizes PMv neurons. Combined, our data provide evidence that GH and PRL may affect the HPG axis via specific hypothalamic neurons.


Assuntos
Hormônio do Crescimento/metabolismo , Prolactina/metabolismo , Maturidade Sexual/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Gônadas/metabolismo , Hormônio do Crescimento/fisiologia , Sistema Hipotálamo-Hipofisário/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Hormônio Luteinizante/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fosforilação , Sistema Hipófise-Suprarrenal/metabolismo , Prolactina/fisiologia , Fator de Transcrição STAT5/metabolismo
6.
Endocrinology ; 160(1): 193-204, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30462197

RESUMO

Previous studies have shown that bromocriptine mesylate (Bromo) lowers blood glucose levels in adults with type 2 diabetes mellitus; however, the mechanism of action of the antidiabetic effects of Bromo is unclear. As a dopamine receptor agonist, Bromo can alter brain dopamine activity affecting glucose control, but it also suppresses prolactin (Prl) secretion, and Prl levels modulate glucose homeostasis. Thus, the objective of the current study was to investigate whether Bromo improves insulin sensitivity via inhibition of Prl secretion. Male and female ob/ob animals (a mouse model of obesity and insulin resistance) were treated with Bromo and/or Prl. Bromo-treated ob/ob mice exhibited lower serum Prl concentration, improved glucose and insulin tolerance, and increased insulin sensitivity in the liver and skeletal muscle compared with vehicle-treated mice. Prl replacement in Bromo-treated mice normalized serum Prl concentration without inducing hyperprolactinemia. Importantly, Prl replacement partially reversed the improvements in glucose homeostasis caused by Bromo treatment. The effects of the Prl receptor antagonist G129R-hPrl on glucose homeostasis were also investigated. We found that central G129R-hPrl infusion increased insulin tolerance of male ob/ob mice. In summary, our findings indicate that part of Bromo effects on glucose homeostasis are associated with decrease in serum Prl levels. Because G129R-hPrl treatment also improved the insulin sensitivity of ob/ob mice, pharmacological compounds that inhibit Prl signaling may represent a promising therapeutic approach to control blood glucose levels in individuals with insulin resistance.


Assuntos
Bromocriptina/administração & dosagem , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/administração & dosagem , Prolactina/metabolismo , Animais , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Humanos , Insulina/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Obesos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo
7.
Physiol Rep ; 6(5)2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29536670

RESUMO

Several metabolic adaptations emerge during pregnancy and continue through lactation, including increases in food intake and body weight, as well as insulin and leptin resistance. These maternal adaptations are thought to play a role in offspring viability and success. Using a model of attenuated maternal metabolic adaptations induced by ablation of the Socs3 gene in leptin receptor expressing cells (SOCS3 KO mice), our study aimed to investigate whether maternal metabolic changes are required for normal offspring development, and if their absence causes metabolic imbalances in adulthood. The litters were subjected to a cross-fostering experimental design to distinguish the prenatal and postnatal effects caused by maternal metabolic adaptations. Males either born or raised by SOCS3 KO mice showed reduced body weight until 8 weeks of life. Both adult males and females born or raised by SOCS3 KO mice also had lower body adiposity. Despite that, no significant changes in energy expenditure, glucose tolerance or insulin resistance were observed. However, males either born or raised by SOCS3 KO mice showed reduced brain mass in adulthood. Furthermore, animals born from SOCS3 KO mice also had lower proopiomelanocortin fiber density in the paraventricular nucleus of the hypothalamus. In conclusion, these findings indicate that the commonly observed metabolic changes in pregnancy and lactation are necessary for normal offspring growth and brain development.


Assuntos
Adaptação Fisiológica , Encéfalo/crescimento & desenvolvimento , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Adiposidade , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Metabolismo Energético , Feminino , Resistência à Insulina , Masculino , Camundongos , Gravidez , Pró-Opiomelanocortina/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Fatores Sexuais , Proteína 3 Supressora da Sinalização de Citocinas/genética , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo
8.
Mol Cell Endocrinol ; 448: 55-65, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28344041

RESUMO

Previous studies have shown that kisspeptin neurons are important mediators of prolactin's effects on reproduction. However, the cellular mechanisms recruited by prolactin to affect kisspeptin neurons remain unknown. Using whole-cell patch-clamp recordings of brain slices from kisspeptin reporter mice, we observed that 20% of kisspeptin neurons in the anteroventral periventricular nucleus was indirectly depolarized by prolactin via an unknown population of prolactin responsive neurons. This effect required the phosphatidylinositol 3-kinase signaling pathway. No effects on the activity of arcuate kisspeptin neurons were observed, despite a high percentage (70%) of arcuate neurons expressing prolactin-induced STAT5 phosphorylation. To determine whether STAT5 expression in kisspeptin cells regulates reproduction, mice carrying Stat5a/b inactivation specifically in kisspeptin cells were generated. These mutants exhibited an early onset of estrous cyclicity, indicating that STAT5 transcription factors exert an inhibitory effect on the timing of puberty.


Assuntos
Kisspeptinas/metabolismo , Fator de Transcrição STAT5/metabolismo , Maturidade Sexual , Transdução de Sinais , Animais , Núcleo Arqueado do Hipotálamo/citologia , Biomarcadores/metabolismo , Ciclo Estral/efeitos dos fármacos , Feminino , Fertilidade/efeitos dos fármacos , Hipotálamo Anterior/citologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Prolactina/farmacologia , Maturidade Sexual/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
9.
Endocrinology ; 157(10): 3901-3914, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27471877

RESUMO

Weight regain frequently follows interventions that reduce body weight, leading to a failure in long-term obesity treatment. Inhibitory proteins of the leptin signaling pathway, such as the suppressor of cytokine signaling 3 (SOCS3), have been studied in conditions that predispose animals to obesity. However, whether SOCS3 modulates postrestriction hyperphagia and weight regain remains unknown. Mice lacking SOCS3 protein specifically in leptin receptor (LepR)-expressing cells (LepR SOCS3 knockout [KO]) were generated and studied in fasting and refeeding conditions. LepR SOCS3 KO mice exhibited increased leptin sensitivity in the hypothalamus. Notably, LepR SOCS3 KO males and females showed attenuated food intake and weight regain after 48 hours of fasting. Postrestriction hyperleptinemia was also prevented in LepR SOCS3 KO mice. Next, we studied possible mechanisms and neural circuits involved in the SOCS3 effects. SOCS3 deletion did not prevent fasting- or refeeding-induced c-Fos expression in the arcuate nucleus of the hypothalamus (ARH) nor fasting-induced increased excitability of ARH LepR-expressing cells. On the other hand, SOCS3 ablation reduced the mRNA levels of hypothalamic orexigenic neuropeptides during fasting (neuropeptide Y, agouti-related protein, orexin, and melanin-concentrating hormone). In summary, our findings suggest that increased leptin sensitivity contributes to the maintenance of a reduced body weight after food deprivation. In addition, the attenuated postrestriction food intake observed in mutant mice was not explained by fasting-induced changes in the activity of ARH neurons but exclusively by a lower transcription of orexigenic neuropeptides during fasting. These results indicate a partial dissociation between the regulation of neuronal activity and gene expression in ARH LepR-expressing cells.


Assuntos
Jejum , Hiperfagia/metabolismo , Leptina/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Animais , Núcleo Arqueado do Hipotálamo/metabolismo , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores para Leptina/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/genética , Aumento de Peso
10.
Sci Rep ; 6: 22421, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26926925

RESUMO

Obesity reduces breastfeeding success and lactation performance in women. However, the mechanisms involved are not entirely understood. In the present study, female C57BL/6 mice were chronically exposed to a high-fat diet to induce obesity and subsequently exhibited impaired offspring viability (only 15% survival rate), milk production (33% reduction), mammopoiesis (one-third of the glandular area compared to control animals) and postpartum maternal behaviors (higher latency to retrieving and grouping the pups). Reproductive experience attenuated these defects. Diet-induced obese mice exhibited high basal pSTAT5 levels in the mammary tissue and hypothalamus, and an acute prolactin stimulus was unable to further increase pSTAT5 levels above basal levels. In contrast, genetically obese leptin-deficient females showed normal prolactin responsiveness. Additionally, we identified the expression of leptin receptors specifically in basal/myoepithelial cells of the mouse mammary gland. Finally, high-fat diet females exhibited altered mRNA levels of ERBB4 and NRG1, suggesting that obesity may involve disturbances to mammary gland paracrine circuits that are critical in the control of luminal progenitor function and lactation. In summary, our findings indicate that high leptin levels are a possible cause of the peripheral and central prolactin resistance observed in obese mice which leads to impaired lactation performance.


Assuntos
Lactação/fisiologia , Leptina/metabolismo , Glândulas Mamárias Animais/metabolismo , Obesidade/metabolismo , Prolactina/metabolismo , Animais , Dieta Hiperlipídica , Feminino , Hipotálamo/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Neuregulina-1/genética , RNA Mensageiro/biossíntese , Receptor ErbB-4/genética , Fator de Transcrição STAT5/metabolismo
11.
Mol Cell Endocrinol ; 423: 11-21, 2016 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-26762764

RESUMO

Leptin is a permissive factor for the onset of puberty. However, changes in adiposity frequently influence leptin sensitivity. Thus, the objective of the present study was to investigate how changes in body weight, fatness, leptin levels and leptin sensitivity interact to control the timing of puberty in female mice. Pre-pubertal obesity, induced by raising C57BL/6 mice in small litters, led to an early puberty onset. Inactivation of Socs3 gene in the brain or exclusively in leptin receptor-expressing cells reduced the body weight and leptin levels at pubertal onset, and increased leptin sensitivity. Notably, these female mice exhibited significant delays in vaginal opening, first estrus and onset of estrus cyclicity. In conclusion, our findings suggest that increased leptin sensitivity did not play an important role in favoring pubertal onset in female mice. Rather, changes in pubertal body weight, fatness and/or leptin levels were more important in influencing the timing of puberty.


Assuntos
Leptina/fisiologia , Obesidade/fisiopatologia , Maturidade Sexual , Animais , Peso Corporal , Ciclo Estral/fisiologia , Feminino , Técnicas de Inativação de Genes , Hipotálamo/metabolismo , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nestina/genética , Nestina/metabolismo , Receptores para Leptina/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo
12.
Nutrients ; 7(5): 3914-37, 2015 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-26007339

RESUMO

Leucine is a well-known activator of the mammalian target of rapamycin (mTOR). Because mTOR signaling regulates several aspects of metabolism, the potential of leucine as a dietary supplement for treating obesity and diabetes mellitus has been investigated. The objective of the present review was to summarize and discuss the available evidence regarding the mechanisms and the effects of leucine supplementation on the regulation of food intake, energy balance, and glucose homeostasis. Based on the available evidence, we conclude that although central leucine injection decreases food intake, this effect is not well reproduced when leucine is provided as a dietary supplement. Consequently, no robust evidence indicates that oral leucine supplementation significantly affects food intake, although several studies have shown that leucine supplementation may help to decrease body adiposity in specific conditions. However, more studies are necessary to assess the effects of leucine supplementation in already-obese subjects. Finally, although several studies have found that leucine supplementation improves glucose homeostasis, the underlying mechanisms involved in these potential beneficial effects remain unknown and may be partially dependent on weight loss.


Assuntos
Glicemia/metabolismo , Suplementos Nutricionais , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Energia/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Leucina/farmacologia , Homeostase/efeitos dos fármacos , Humanos , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Serina-Treonina Quinases TOR/metabolismo
13.
Circ J ; 79(6): 1372-80, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25808225

RESUMO

BACKGROUND: Hyperactivity of the renin-angiotensin system (RAS) and functional deficits in hypertension are reduced after exercise training. We evaluate in arteries, kidney and plasma of hypertensive rats the sequential effects of training on vascular angiotensinogen, Ang II and Ang (1-7) content. METHODS AND RESULTS: Spontaneously hypertensive rats (SHR) and Wistar-Kyoto rats (WKY) were trained or kept sedentary (S) for 3 months. After hemodynamic measurements (weeks 0, 1, 2, 4, 8 and 12), blood, arteries and kidneys were obtained to quantify the angiotensin content (HPLC) and angiotensinogen expression (Western Blotting). SHR-S vs. WKY-S exhibited elevated pressure, increased angiotensinogen and angiotensins' content in the renal artery with a high Ang II/Ang (1-7) ratio (~5-fold higher than in the femoral artery, kidney and plasma, and 14-fold higher than in the aorta). Training promptly reduced angiotensinogen expression and downregulated the RAS in the renal SHR artery (1st-12th week), with a specific reduction of the vasoconstrictor axis; significant reduction of the AngII/Ang (1-7) ratio (36%, T4-T8) occurred simultaneously with significant pressure fall (5%). In other SHR arteries, plasma and kidneys and in all WKY tissues, T-induced AngII and Ang (1-7) reductions were proportional, maintaining the AngII/Ang (1-7) ratio. CONCLUSIONS: Vascular RAS is not equally expressed in vessels, having crucial importance in the renal artery. In the renal SHR artery, training downregulates the vasoconstrictor and preserves the vasodilator axis while in other tissues and plasma training reduces both RAS axes, thus maintaining the vasoconstriction/vasodilatation balance in a lower level.


Assuntos
Angiotensina II/biossíntese , Angiotensina I/biossíntese , Angiotensinogênio/biossíntese , Rim/metabolismo , Fragmentos de Peptídeos/biossíntese , Condicionamento Físico Animal/fisiologia , Artéria Renal/metabolismo , Sistema Renina-Angiotensina/fisiologia , Aerobiose/fisiologia , Angiotensina I/sangue , Angiotensina II/sangue , Enzima de Conversão de Angiotensina 2 , Angiotensinogênio/sangue , Angiotensinogênio/genética , Animais , Pressão Sanguínea , Artéria Femoral , Masculino , Especificidade de Órgãos , Fragmentos de Peptídeos/sangue , Peptidil Dipeptidase A/sangue , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Corrida , Vasoconstrição/fisiologia , Vasodilatação/fisiologia
14.
Mol Metab ; 4(3): 237-45, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25737950

RESUMO

OBJECTIVE: During pregnancy, women normally increase their food intake and body fat mass, and exhibit insulin resistance. However, an increasing number of women are developing metabolic imbalances during pregnancy, including excessive gestational weight gain and gestational diabetes mellitus. Despite the negative health impacts of pregnancy-induced metabolic imbalances, their molecular causes remain unclear. Therefore, the present study investigated the molecular mechanisms responsible for orchestrating the metabolic changes observed during pregnancy. METHODS: Initially, we investigated the hypothalamic expression of key genes that could influence the energy balance and glucose homeostasis during pregnancy. Based on these results, we generated a conditional knockout mouse that lacks the suppressor of cytokine signaling-3 (SOCS3) only in leptin receptor-expressing cells and studied these animals during pregnancy. RESULTS: Among several genes involved in leptin resistance, only SOCS3 was increased in the hypothalamus of pregnant mice. Remarkably, SOCS3 deletion from leptin receptor-expressing cells prevented pregnancy-induced hyperphagia, body fat accumulation as well as leptin and insulin resistance without affecting the ability of the females to carry their gestation to term. Additionally, we found that SOCS3 conditional deletion protected females against long-term postpartum fat retention and streptozotocin-induced gestational diabetes. CONCLUSIONS: Our study identified the increased hypothalamic expression of SOCS3 as a key mechanism responsible for triggering pregnancy-induced leptin resistance and metabolic adaptations. These findings not only help to explain a common phenomenon of the mammalian physiology, but it may also aid in the development of approaches to prevent and treat gestational metabolic imbalances.

15.
Endocrinology ; 155(11): 4226-36, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25144922

RESUMO

Several studies have shown that estrogens mimic leptin's effects on energy balance regulation. However, the findings regarding the consequences of reduced sex hormone levels on leptin sensitivity are divergent. In the present study, we employed different experimental paradigms to elucidate the interaction between estrogens, leptin, and energy balance regulation. We confirmed previous reports showing that ovariectomy caused a reduction in locomotor activity and energy expenditure leading mice to obesity and glucose intolerance. However, the acute and chronic anorexigenic effects of leptin were preserved in ovariectomized (OVX) mice despite their increased serum leptin levels. We studied hypothalamic gene expression at different time points after ovariectomy and observed that changes in the expression of genes involved in leptin resistance (suppressors of cytokine signaling and protein-tyrosine phosphatases) did not precede the early onset of obesity in OVX mice. On the contrary, reduced sex hormone levels caused an up-regulation of the long form of the leptin receptor (LepR), resulting in increased activation of leptin signaling pathways in OVX leptin-treated animals. The up-regulation of the LepR was observed in long-term OVX mice (30 d or 24 wk after ovariectomy) but not 7 days after the surgery. In addition, we observed a progressive decrease in the coexpression of LepR and estrogen receptor-α in the hypothalamus after the ovariectomy, resulting in a low percentage of dual-labeled cells in OVX mice. Taken together, our findings suggest that the weight gain caused by reduced sex hormone levels is not primarily caused by induction of a leptin-resistance state.


Assuntos
Resistência a Medicamentos , Hormônios Esteroides Gonadais/sangue , Leptina/farmacologia , Aumento de Peso , Animais , Depressores do Apetite/farmacologia , Metabolismo Energético/efeitos dos fármacos , Feminino , Glucose/metabolismo , Hipogonadismo/sangue , Hipogonadismo/metabolismo , Leptina/sangue , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Ovariectomia , Fatores Sexuais , Aumento de Peso/efeitos dos fármacos
16.
Nutrients ; 6(4): 1364-73, 2014 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-24699194

RESUMO

Several studies showed that l-leucine supplementation reduces adiposity when provided before the onset of obesity. We studied rats that were exposed to a high-fat diet (HFD) for 10 weeks before they started to receive l-leucine supplementation. Fat mass was increased in l-leucine-supplemented rats consuming the HFD. Accordingly, l-leucine produced a hypothalamic pattern of gene expression that favors fat accumulation. In conclusion, l-leucine supplementation worsened the adiposity of rats previously exposed to HFD possibly by central mechanisms.


Assuntos
Adiposidade/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais , Hipotálamo/metabolismo , Leucina/efeitos adversos , Obesidade/patologia , Animais , Ingestão de Energia , Expressão Gênica , Leucina/administração & dosagem , Masculino , Ratos , Ratos Wistar
17.
PLoS One ; 8(12): e84094, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24349566

RESUMO

Leucine activates the intracellular mammalian target of the rapamycin (mTOR) pathway, and hypothalamic mTOR signaling regulates food intake. Although central infusion of leucine reduces food intake, it is still uncertain whether oral leucine supplementation is able to affect the hypothalamic circuits that control energy balance. We observed increased phosphorylation of p70s6k in the mouse hypothalamus after an acute oral gavage of leucine. We then assessed whether acute oral gavage of leucine induces the activation of neurons in several hypothalamic nuclei and in the brainstem. Leucine did not induce the expression of Fos in hypothalamic nuclei, but it increased the number of Fos-immunoreactive neurons in the area postrema. In addition, oral gavage of leucine acutely increased the 24 h food intake of mice. Nonetheless, chronic leucine supplementation in the drinking water did not change the food intake and the weight gain of ob/ob mice and of wild-type mice consuming a low- or a high-fat diet. We assessed the hypothalamic gene expression and observed that leucine supplementation increased the expression of enzymes (BCAT1, BCAT2 and BCKDK) that metabolize branched-chain amino acids. Despite these effects, leucine supplementation did not induce an anorectic pattern of gene expression in the hypothalamus. In conclusion, our data show that the brain is able to sense oral leucine intake. However, the food intake is not modified by chronic oral leucine supplementation. These results question the possible efficacy of leucine supplementation as an appetite suppressant to treat obesity.


Assuntos
Anorexia , Depressores do Apetite/farmacologia , Suplementos Nutricionais , Ingestão de Alimentos/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Hipotálamo/metabolismo , Leucina/farmacologia , Proteínas do Tecido Nervoso/biossíntese , Animais , Masculino , Camundongos , Camundongos Obesos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...