Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 109
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Phytomedicine ; 126: 155297, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38342019

RESUMO

BACKGROUND: Research on the imbalance of proopiomelanocortin (POMC)/agouti-related protein (AgRP) neurons in the hypothalamus holds potential insights into the pathophysiology of diabetes. Jinkui Shenqi pills (JSP), a prevalent traditional Chinese medicine, regulate hypothalamic function and treat diabetes. PURPOSE: To investigate the hypoglycemic effect of JSP and explore the probable mechanism in treating diabetes. METHODS: A type 2 diabetes mouse model was used to investigate the pharmacodynamics of JSP. The glucose-lowering efficacy of JSP was assessed through various metrics including body weight, food consumption, fasting blood glucose (FBG), serum insulin levels, and an oral glucose tolerance test (OGTT). To elucidate the modulatory effects of JSP on hypothalamic mechanisms, we quantified the expression and activity of POMC and AgRP and assessed the insulin-mediated phosphoinositide 3-kinase (PI3K)/protein kinase A (AKT)/forkhead box O1 (FOXO1) pathway in diabetic mice via western blotting and immunohistochemistry. Additionally, primary hypothalamic neurons were exposed to high glucose and palmitic acid levels to induce insulin resistance, and the influence of JSP on POMC/AgRP protein expression and activation was evaluated by PI3K protein inhibition using western blotting and immunofluorescence. RESULTS: Medium- and high-dose JSP treatment effectively inhibited appetite, resulting in a steady declining trend in body weight, FBG, and OGTT results in diabetic mice (p < 0.05). These JSP groups also had significantly increased insulin levels (p < 0.05). Importantly, the medium-dose group exhibited notable protection of hypothalamic neuronal and synaptic structures, leading to augmentation of dendritic length and branching (p < 0.05). Furthermore, low-, medium-, and high-dose JSP groups exhibited increased phosphorylated (p) INSR, PI3K, pPI3K, AKT, and pAKT expression, as well as decreased FOXO1 and increased pFOXO1 expression, indicating improved hypothalamic insulin resistance in diabetic mice (p < 0.05). Treatment with 10% JSP-enriched serum produced a marked elevation of both expression and activation of POMC (p < 0.05), with a concurrent reduction in AgRP expression and activation within primary hypothalamic neurons (p < 0.05). Intriguingly, these effects could be attributed to the regulatory dynamics of PI3K activity. CONCLUSION: Our findings suggest that JSP can ameliorate diabetes by regulating POMC/AgRP expression and activity. The insulin-mediated PI3K/AKT/FOXO1 pathway plays an important regulatory role in this intricate process.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Medicamentos de Ervas Chinesas , Resistência à Insulina , Camundongos , Animais , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/farmacologia , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Hipotálamo/metabolismo , Insulina/metabolismo , Glucose/metabolismo , Peso Corporal
2.
Steroids ; 203: 109367, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38266463

RESUMO

While the effects of progesterone on body weight and appetite in pre-menopausal conditions have been well elucidated, its effects in post-menopausal conditions have not been clarified. On the contrary, the effects of estrogen on body weight and appetite in post-menopausal conditions have been well established. In this study, the effects of progesterone treatment on body weight, appetite, and fat mass in ovariectomized rats were evaluated. In addition, the central and/or peripheral levels of oxytocin (OT), leptin, and their receptors, which are potent anorectic factors, were examined. Female rats were ovariectomized and divided into control, progesterone-treated, and estrogen-treated groups. Body weight, food intake, and subcutaneous fat mass were lower in both the progesterone and estrogen groups than in the control group. The estrogen group exhibited higher serum OT levels than the control group, whereas the OT levels of the progesterone and control groups did not differ. The serum leptin levels of both the progesterone and estrogen groups were lower than those of the control group. Gene expression analysis of OT, leptin, and their receptors in the hypothalamus and adipose tissue found few significant differences among the groups. Hypothalamic neuropeptide Y (NPY) and pro-opiomelanocortin (POMC) mRNA levels involved in appetite regulation were slightly altered in the progesterone and estrogen groups. These findings suggest that progesterone treatment may have favorable effects on body weight, appetite, and fat mass regulation in post-menopausal conditions and that the mechanisms underlying these effects of progesterone differ from those underlying the effects of estrogen.


Assuntos
Leptina , Progesterona , Ratos , Animais , Feminino , Leptina/metabolismo , Progesterona/farmacologia , Progesterona/metabolismo , Ingestão de Alimentos , Peso Corporal , Hipotálamo , Proteínas de Transporte , Estrogênios/farmacologia , Estrogênios/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia
3.
Endocr Rev ; 44(6): 1096-1106, 2023 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-37409973

RESUMO

Based on insights obtained during the past decade, the classical concept of an activated hypothalamus-pituitary-adrenocortical axis in response to critical illness is in need of revision. After a brief central hypothalamus-pituitary-adrenocortical axis activation, the vital maintenance of increased systemic cortisol availability and action in response to critical illness is predominantly driven by peripheral adaptations rather than by an ongoing centrally activated several-fold increased production and secretion of cortisol. Besides the known reduction of cortisol-binding proteins that increases free cortisol, these peripheral responses comprise suppressed cortisol metabolism in liver and kidney, prolonging cortisol half-life, and local alterations in expression of 11ßHSD1, glucocorticoid receptor-α (GRα), and FK506 binding protein 5 (FKBP51) that appear to titrate increased GRα action in vital organs and tissues while reducing GRα action in neutrophils, possibly preventing immune-suppressive off-target effects of increased systemic cortisol availability. Peripherally increased cortisol exerts negative feed-back inhibition at the pituitary level impairing processing of pro-opiomelanocortin into ACTH, thereby reducing ACTH-driven cortisol secretion, whereas ongoing central activation results in increased circulating pro-opiomelanocortin. These alterations seem adaptive and beneficial for the host in the short term. However, as a consequence, patients with prolonged critical illness who require intensive care for weeks or longer may develop a form of central adrenal insufficiency. The new findings supersede earlier concepts such as "relative," as opposed to "absolute," adrenal insufficiency and generalized systemic glucocorticoid resistance in the critically ill. The findings also question the scientific basis for broad implementation of stress dose hydrocortisone treatment of patients suffering from acute septic shock solely based on assumption of cortisol insufficiency.


Assuntos
Insuficiência Adrenal , Doenças da Hipófise , Humanos , Hidrocortisona/metabolismo , Estado Terminal/terapia , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Sistema Hipotálamo-Hipofisário , Insuficiência Adrenal/tratamento farmacológico , Insuficiência Adrenal/metabolismo , Hipotálamo , Doenças da Hipófise/metabolismo , Hormônio Adrenocorticotrópico/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo
4.
J Endocrinol Invest ; 46(12): 2609-2616, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37233978

RESUMO

PURPOSE: Clinical control of corticotroph tumors is difficult to achieve since they usually persist or relapse after surgery. Pasireotide is approved to treat patients with Cushing's disease for whom surgical therapy is not an option. However, Pasireotide seems to be effective only in a sub-set of patients, highlighting the importance to find a response marker to this approach. Recent studies demonstrated that the delta isoform of protein kinase C (PRKCD) controls viability and cell cycle progression of an in vitro model of ACTH-secreting pituitary tumor, the AtT-20/D16v-F2 cells. This study aims at exploring the possible PRKCD role in mediating Pasireotide effects. METHODS: It was assessed cell viability, POMC expression and ACTH secretion in AtT20/D16v-F2 cells over- or under-expressing PRKCD. RESULTS: We found that Pasireotide significantly reduces AtT20/D16v-F2 cell viability, POMC expression and ACTH secretion. In addition, Pasireotide reduces miR-26a expression. PRKCD silencing decreases AtT20/D16v-F2 cell sensitivity to Pasireotide treatment; on the contrary, PRKCD overexpression increases the inhibitory effects of Pasireotide on cell viability and ACTH secretion. CONCLUSION: Our results provide new insights into potential PRKCD contribution in Pasireotide mechanism of action and suggest that PRKCD might be a possible marker of therapeutic response in ACTH-secreting pituitary tumors.


Assuntos
Hipersecreção Hipofisária de ACTH , Neoplasias Hipofisárias , Humanos , Neoplasias Hipofisárias/patologia , Corticotrofos/metabolismo , Corticotrofos/patologia , Proteína Quinase C-delta/metabolismo , Proteína Quinase C-delta/farmacologia , Proteína Quinase C-delta/uso terapêutico , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Hormônio Adrenocorticotrópico/metabolismo , Recidiva Local de Neoplasia/patologia , Linhagem Celular , Hipersecreção Hipofisária de ACTH/metabolismo , Linhagem Celular Tumoral
5.
Diabetes Obes Metab ; 25(6): 1534-1546, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36852745

RESUMO

AIM: To clarify the effects of glucose-dependent insulinotropic polypeptide (GIP) receptor agonists (GIPRAs) on feeding and body weight. MATERIALS AND METHODS: Acute and subchronic effects of subcutaneous GIPFA-085, a long-acting GIPRA, on blood glucose, food intake, body weight, respiratory exchange ratio and plasma leptin levels were measured in diet-induced obese (DIO) mice and/or functional leptin-deficient ob/ob mice. The effects of GIPFA-085 on the hypothalamic arcuate nucleus (ARC) neurons from lean and DIO mice were studied by measuring cytosolic Ca2+ concentration ([Ca2+ ]i ). RESULTS: Single bolus GIPFA-085 (30, 300 nmol/kg) dose-dependently reduced blood glucose in glucose tolerance tests, elevated plasma leptin levels at 0.5-6 hours and inhibited food intake at 2-24 hours after injection in DIO mice. Daily GIPFA-085 (300 nmol/kg) inhibited food intake and increased fat utilization on day 1, and reduced body weight gain on days 3-12 of treatment in DIO, but not ob/ob, mice. GIPFA-085 increased [Ca2+ ]i in the ARC leptin-responsive and proopiomelanocortin (POMC) neurons. GIPFA-085 and leptin cooperated to increase [Ca2+ ]i in ARC neurons and inhibit food intake. CONCLUSIONS: GIPFA-085 acutely inhibits feeding and increases lipid utilization, and sustainedly lowers body weight in DIO mice via mechanisms involving rises in leptin and activation of ARC leptin-responsive and POMC neurons. This study highlights the therapeutic potential of GIPRAs for treating obesity and diabetes.


Assuntos
Núcleo Arqueado do Hipotálamo , Leptina , Camundongos , Animais , Leptina/metabolismo , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Pró-Opiomelanocortina/uso terapêutico , Glicemia , Obesidade/tratamento farmacológico , Obesidade/etiologia , Dieta , Peso Corporal , Receptores Acoplados a Proteínas G , Neurônios/metabolismo , Camundongos Endogâmicos C57BL
6.
Pain ; 164(5): 1051-1066, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36448978

RESUMO

ABSTRACT: The nucleus of the solitary tract (NTS) contains pro-opiomelanocortin (POMC) neurons that are 1 of the 2 major sources of ß-endorphin in the brain. The functional role of these NTS POMC neurons in nociceptive and cardiorespiratory function is debated. We have shown that NTS POMC optogenetic activation produces bradycardia and transient apnoea in a working heart-brainstem preparation and chemogenetic activation with an engineered ion channel (PSAM) produced opioidergic analgesia in vivo. To better define the role of the NTS POMC neurons in behaving animals, we adopted in vivo optogenetics (ChrimsonR) and excitatory/inhibitory chemogenetic DREADD (hM3Dq/hM4Di) strategies in POMC-Cre mice. We show that optogenetic activation of NTS POMC neurons produces time-locked, graded, transient bradycardia and bradypnoea in anaesthetised mice that is naloxone sensitive (1 mg/kg, i.p.), suggesting a role of ß-endorphin. Both optogenetic and chemogenetic activation of NTS POMC neurons produces sustained thermal analgesia in behaving mice that can be blocked by naloxone. It also produced analgesia in an inflammatory pain model (carrageenan) but not in a neuropathic pain model (tibial nerve transection). Inhibiting NTS POMC neurons does not produce any effect on basal nociception but inhibits stress-induced analgesia (unlike inhibition of arcuate POMC neurons). Activation of NTS POMC neuronal populations in conscious mice did not cause respiratory depression, anxiety, or locomotor deficit (in open field) or affective preference. These findings indicate that NTS POMC neurons play a key role in the generation of endorphinergic endogenous analgesia and can also regulate cardiorespiratory function.


Assuntos
Analgesia , Pró-Opiomelanocortina , Camundongos , Animais , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Núcleo Solitário , Bradicardia , beta-Endorfina , Neurônios , Naloxona/farmacologia , Dor
7.
Molecules ; 27(15)2022 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-35956831

RESUMO

PPARγ agonists are implicated in the regulation of diabetes and metabolic syndrome and have therapeutic potential in brain disorders. PPARγ modulates appetite through its central effects, especially on the hypothalamic arcuate nucleus (ARC). Previous studies demonstrated that the small molecule GL516 is a PPARγ agonist able to reduce oxidative stress and apoptosis with a potential neuroprotective role. Herein, we investigated the effects of GL516, in vitro and ex vivo, on the levels of hypothalamic dopamine (DA) and serotonin (5-HT). The gene expressions of neuropeptide Y, CART, AgRP, and POMC, which play master roles in the neuroendocrine regulation of feeding behavior and energy balance, were also evaluated. HypoE22 cells were treated with H2O2 (300 µM) for 2 h e 30' and with different concentrations of GL516 (1 nM-100 µM). The cell viability was evaluated after 24 and 48 h of culturing using the MTT test. DA and 5-HT levels in the HypoE22 cell supernatants were analyzed through HPLC; an ex vivo study on isolated hypothalamic specimens challenged with scalar concentrations of GL516 (1-100 µM) and with pioglitazone (10 µM) was carried out. The gene expressions of CART, NPY, AgRP, and POMC were also determined by a quantitative real-time PCR. The results obtained showed that GL516 was able to reduce DA and 5-HT turnover; moreover, it was effective in stimulating NPY and AgRP gene expressions with a concomitant reduction in CART and POMC gene expressions. These results highlight the capability of GL516 to modulate neuropeptide pathways deeply involved in appetite control suggesting an orexigenic effect. These findings emphasize the potential use of GL516 as a promising candidate for therapeutical applications in neurodegenerative diseases associated with the reduction in food intake and stimulation of catabolic pathways.


Assuntos
PPAR gama , Pró-Opiomelanocortina , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/farmacologia , Peróxido de Hidrogênio/farmacologia , Hipotálamo/metabolismo , Neuropeptídeo Y/genética , Neuropeptídeo Y/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Serotonina/metabolismo , Serotonina/farmacologia
8.
Life Sci Alliance ; 5(11)2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36007929

RESUMO

Hyperphagia and obesity profoundly affect the health of children with Prader-Willi syndrome (PWS). The Magel2 gene among the genes in the Prader-Willi syndrome deletion region is expressed in proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC). Knockout of the Magel2 gene disrupts POMC neuronal circuits and functions. Here, we report that loss of the Magel2 gene exclusively in ARCPOMC neurons innervating the medial amygdala (MeA) causes a reduction in body weight in both male and female mice fed with a high-fat diet. This anti-obesity effect is associated with an increased locomotor activity. There are no significant differences in glucose and insulin tolerance in mice without the Magel2 gene in ARCPOMC neurons innervating the MeA. Plasma estrogen levels are higher in female mutant mice than in controls. Blockade of the G protein-coupled estrogen receptor (GPER), but not estrogen receptor-α (ER-α), reduces locomotor activity in female mutant mice. Hence, our study provides evidence that knockdown of the Magel2 gene in ARCPOMC neurons innervating the MeA reduces susceptibility to diet-induced obesity with increased locomotor activity through activation of central GPER.


Assuntos
Antígenos de Neoplasias/genética , Síndrome de Prader-Willi , Pró-Opiomelanocortina , Proteínas/genética , Tonsila do Cerebelo/metabolismo , Animais , Dieta Hiperlipídica/efeitos adversos , Feminino , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Obesidade/genética , Síndrome de Prader-Willi/genética , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia
9.
Acta Biochim Pol ; 69(3): 647-655, 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35877942

RESUMO

Appetite regulation in the hypothalamus is dependent on hormonal signals from the periphery, such as insulin and leptin, and can be modulated by both sugar-rich diet and stress. Our aim was to explore the effects of 9-week feeding with 20% fructose solution combined with 4-week chronic unpredictable stress, on appetite-regulating neuropeptides and modulatory role of leptin and insulin signalling in the hypothalamus of male Wistar rats. Energy intake, body mass and adiposity, as well as circulatory leptin and insulin concentrations were assessed. Hypothalamic insulin signalling was analysed at the level of glucose transporters, as well as at the protein level and phosphorylation of insulin receptor, insulin receptor supstrate-1, Akt and ERK. Phosphorylation of AMP-activated protein kinase (AMPK), level of protein tyrosine phosphatase 1B (PTP1B) and expression of leptin receptor (ObRb) and suppressor of cytokine signalling 3 (SOCS3) were also analysed, together with the expression of orexigenic agouti-related protein (AgRP) and anorexigenic proopiomelanocortin (POMC) neuropeptides. The results revealed that stress decreased body mass and adiposity, blood leptin level and expression of ObRb, SOCS3 and POMC, while combination with fructose diet led to marked increase of AgRP, associated with AMPK phosphorylation despite increased plasma insulin. Reduced Akt, enhanced ERK activity and elevated PTP1B were also observed in the hypothalamus of these animals. In conclusion, our results showed that joint effects of fructose diet and stress are more deleterious than the separate ones, since inappropriate appetite control in the hypothalamus may provide a setting for the disturbed energy homeostasis in the long run.


Assuntos
Neuropeptídeos , Pró-Opiomelanocortina , Proteínas Quinases Ativadas por AMP/metabolismo , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/farmacologia , Animais , Citocinas/metabolismo , Dieta , Frutose/efeitos adversos , Frutose/metabolismo , Glucose/metabolismo , Hipotálamo/metabolismo , Insulina , Leptina , Masculino , Neuropeptídeos/metabolismo , Neuropeptídeos/farmacologia , Fosforilação , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 1/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 1/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Receptor de Insulina/metabolismo , Receptores para Leptina/metabolismo
10.
Cell Death Dis ; 13(7): 659, 2022 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-35902579

RESUMO

Palmitic acid (PA) is significantly increased in the hypothalamus of mice, when fed chronically with a high-fat diet (HFD). PA impairs insulin signaling in hypothalamic neurons, by a mechanism dependent on autophagy, a process of lysosomal-mediated degradation of cytoplasmic material. In addition, previous work shows a crosstalk between autophagy and the primary cilium (hereafter cilium), an antenna-like structure on the cell surface that acts as a signaling platform for the cell. Ciliopathies, human diseases characterized by cilia dysfunction, manifest, type 2 diabetes, among other features, suggesting a role of the cilium in insulin signaling. Cilium depletion in hypothalamic pro-opiomelanocortin (POMC) neurons triggers obesity and insulin resistance in mice, the same phenotype as mice deficient in autophagy in POMC neurons. Here we investigated the effect of chronic consumption of HFD on cilia; and our results indicate that chronic feeding with HFD reduces the percentage of cilia in hypothalamic POMC neurons. This effect may be due to an increased amount of PA, as treatment with this saturated fatty acid in vitro reduces the percentage of ciliated cells and cilia length in hypothalamic neurons. Importantly, the same effect of cilia depletion was obtained following chemical and genetic inhibition of autophagy, indicating autophagy is required for ciliogenesis. We further demonstrate a role for the cilium in insulin sensitivity, as cilium loss in hypothalamic neuronal cells disrupts insulin signaling and insulin-dependent glucose uptake, an effect that correlates with the ciliary localization of the insulin receptor (IR). Consistently, increased percentage of ciliated hypothalamic neuronal cells promotes insulin signaling, even when cells are exposed to PA. Altogether, our results indicate that, in hypothalamic neurons, impairment of autophagy, either by PA exposure, chemical or genetic manipulation, cause cilia loss that impairs insulin sensitivity.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Animais , Autofagia , Cílios/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hipotálamo/metabolismo , Insulina/metabolismo , Resistência à Insulina/genética , Camundongos , Neurônios/metabolismo , Ácido Palmítico/metabolismo , Ácido Palmítico/farmacologia , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia
11.
BMB Rep ; 55(6): 293-298, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35651327

RESUMO

Antipsychotics have been widely accepted as a treatment of choice for psychiatric illnesses such as schizophrenia. While atypical antipsychotics such as aripiprazole are not associated with obesity and diabetes, olanzapine is still widely used based on the anticipation that it is more effective in treating severe schizophrenia than aripiprazole, despite its metabolic side effects. To address metabolic problems, metformin is widely prescribed. Hypothalamic proopiomelanocortin (POMC) neurons have been identified as the main regulator of metabolism and energy expenditure. Although the relation between POMC neurons and metabolic disorders is well established, little is known about the effects of olanzapine and metformin on hypothalamic POMC neurons. In the present study, we investigated the effect of olanzapine and metformin on the hypothalamic POMC neurons in female mice. Olanzapine administration for 5 days significantly decreased Pomc mRNA expression, POMC neuron numbers, POMC projections, and induced leptin resistance before the onset of obesity. It was also observed that coadministration of metformin with olanzapine not only increased POMC neuron numbers and projections but also improved the leptin response of POMC neurons in the olanzapine-treated female mice. These findings suggest that olanzapine-induced hypothalamic POMC neuron abnormality and leptin resistance, which can be ameliorated by metformin administration, are the possible causes of subsequent hyperphagia. [BMB Reports 2022; 55(6): 293-298].


Assuntos
Antipsicóticos , Metformina , Animais , Antipsicóticos/metabolismo , Antipsicóticos/farmacologia , Aripiprazol/metabolismo , Aripiprazol/farmacologia , Feminino , Hipotálamo/metabolismo , Leptina/metabolismo , Metformina/metabolismo , Metformina/farmacologia , Camundongos , Neurônios/metabolismo , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Olanzapina/metabolismo , Olanzapina/farmacologia , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia
12.
Sci Signal ; 15(733): eabj8204, 2022 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-35536884

RESUMO

Variants in the gene encoding ankyrin repeat and SOCS box-containing 4 (ASB4) are linked to human obesity. Here, we characterized the pathways underlying the metabolic functions of ASB4. Hypothalamic Asb4 expression was suppressed by fasting in wild-type mice but not in mice deficient in AgRP, which encodes Agouti-related protein (AgRP), an appetite-stimulating hormone, suggesting that ASB4 is a negative target of AgRP. Many ASB4 neurons in the brain were adjacent to AgRP terminals, and feeding induced by AgRP neuronal activation was disrupted in Asb4-deficient mice. Acute knockdown of Asb4 in the brain caused marked hyperphagia due to increased meal size, and Asb4 deficiency led to increased meal size and food intake at the onset of refeeding, when very large meals were consumed. Asb4-deficient mice were resistant to the meal-terminating effects of exogenously administered calcitonin and showed decreased neuronal expression of Calcr, which encodes the calcitonin receptor. Pro-opiomelanocortin (POMC) neurons in the arcuate nucleus in mice are involved in glucose homeostasis, and Asb4 deficiency specifically in POMC neurons resulted in glucose intolerance that was independent of obesity. Furthermore, individuals with type 2 diabetes showed reduced ASB4 abundance in the infundibular nuclei, the human equivalent of the arcuate nucleus. Together, our results indicate that ASB4 acts in the brain to improve glucose homeostasis and to induce satiety after substantial meals, particularly those after food deprivation.


Assuntos
Diabetes Mellitus Tipo 2 , Neuropeptídeos , Proteína Relacionada com Agouti/genética , Proteína Relacionada com Agouti/metabolismo , Proteína Relacionada com Agouti/farmacologia , Animais , Calcitonina/metabolismo , Calcitonina/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Glucose/metabolismo , Homeostase , Hipotálamo/metabolismo , Camundongos , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Obesidade/genética , Obesidade/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia
13.
Biomolecules ; 12(2)2022 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-35204737

RESUMO

Spexin (SPX) is a recently identified neuropeptide that is believed to play an important role in the regulation of energy homeostasis. Here, we describe a mediating function of SPX in hypothalamic leptin action. Intracerebroventricular (icv) SPX administration induced a decrease in food intake and body weight gain. SPX was found to be expressed in cells expressing leptin receptor ObRb in the mouse hypothalamus. In line with this finding, icv leptin injection increased SPX mRNA in the ObRb-positive cells of the hypothalamus, which was blocked by treatment with a STAT3 inhibitor. Leptin also increased STAT3 binding to the SPX promoter, as measured by chromatin immunoprecipitation assays. In vivo blockade of hypothalamic SPX biosynthesis with an antisense oligodeoxynucleotide (AS ODN) resulted in a diminished leptin effect on food intake and body weight. AS ODN reversed leptin's effect on the proopiomelanocortin (POMC) mRNA expression and, moreover, decreased leptin-induced STAT3 binding to the POMC promoter sequence. These results suggest that SPX is involved in leptin's action on POMC gene expression in the hypothalamus and impacts the anorexigenic effects of leptin.


Assuntos
Leptina , Neuropeptídeos , Animais , Comportamento Alimentar , Hipotálamo/metabolismo , Leptina/metabolismo , Leptina/farmacologia , Camundongos , Neuropeptídeos/metabolismo , Pró-Opiomelanocortina/genética , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia
14.
Environ Health Perspect ; 129(9): 97006, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34495743

RESUMO

BACKGROUND: Epidemiological studies have shown that exposure to ambient fine particulate matter with aerodynamic diameter less than or equal to 2.5 µm (PM2.5) correlates with a decrease in sperm count, but the biological mechanism remains elusive. OBJECTIVES: This study tested whether hypothalamic inflammation, an emerging pathophysiological mediator, mediates the development of lower epididymal sperm count due to PM2.5 exposure. METHODS: Inhibitor κB kinase 2 (IKK2) was conditionally knocked out either in all neurons or subtypes of hypothalamic neurons of mice. Effects of concentrated ambient PM2.5 (CAP) exposure on hypothalamic inflammation, the hypothalamic-pituitary-gonadal (HPG) axis, and epididymal sperm count of these mouse models were then assessed. Furthermore, to test whether hypothalamic inflammation is sufficient to decrease sperm production, we overexpressed constitutively active IKK2 (IKK2ca) either in all neurons or subtypes of hypothalamic neurons and assessed hypothalamic inflammation, the HPG axis, and sperm production of these overexpression mouse models. RESULTS: CAP-exposed wild-type control mice vs. filtered air (FA)-exposed wild-type control mice had a higher expression of hypothalamic inflammatory markers, lower functional indexes of the HPG axis, and a lower epididymal sperm count. In contrast, all these measurements for CAP- vs. FA-exposed mice deficient of IKK2 in all neurons were comparable. We also found that overexpression of IKK2ca in either all neurons or pro-opiomelanocortin (POMC) neurons only, but not in Agouti-related protein (AgRP) neurons only, resulted in lower functional indexes of the HPG axis and a lower epididymal sperm count. Moreover, we showed that CAP- vs. FA-exposed mice deficient of IKK2 in POMC neurons had a comparable expression of hypothalamic inflammatory markers, comparable functional indexes of the HPG axis, and a comparable epididymal sperm count. DISCUSSION: This mouse model study shows a causal role of IKK2 of POMC neurons in the development of lower epididymal sperm count due to PM2.5 exposure, providing a mechanistic insight into this emerging pathogenesis. https://doi.org/10.1289/EHP8868.


Assuntos
Material Particulado , Pró-Opiomelanocortina , Animais , Hipotálamo , Masculino , Camundongos , Neurônios , Material Particulado/metabolismo , Material Particulado/toxicidade , Pró-Opiomelanocortina/metabolismo , Pró-Opiomelanocortina/farmacologia , Espermatogênese
15.
Mol Cell Endocrinol ; 514: 110876, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32473184

RESUMO

Seasonal rhythms in energy balance are well documented across temperate and equatorial zones animals. The long-term regulated changes in seasonal physiology consists of a rheostatic system that is essential to successful time annual cycles in reproduction, hibernation, torpor, and migration. Most animals use the annual change in photoperiod as a reliable and robust environmental cue to entrain endogenous (i.e. circannual) rhythms. Research over the past few decades has predominantly examined the role of first order neuroendocrine peptides for the rheostatic changes in energy balance. These anorexigenic and orexigenic neuropeptides in the arcuate nucleus include neuropeptide y (Npy), agouti-related peptide (Agrp), cocaine and amphetamine related transcript (Cart) and pro-opiomelanocortin (Pomc). Recent studies also indicate that VGF nerve growth factor inducible (Vgf) in the arcuate nucleus is involved in the seasonal regulation of energy balance. In situ hybridization, qPCR and RNA-sequencing studies have identified that Pomc expression across fish, avian and mammalian species, is a neuroendocrine marker that reflects seasonal energetic states. Here we highlight that long-term changes in arcuate Pomc and Vgf expression is conserved across species and may provide rheostatic regulation of seasonal energy balance.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Fatores de Crescimento Neural/farmacologia , Neuropeptídeos/metabolismo , Pró-Opiomelanocortina/farmacologia , Proteína Relacionada com Agouti/farmacologia , Proteína Relacionada com Agouti/fisiologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Metabolismo Energético/fisiologia , Humanos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Proteínas do Tecido Nervoso/farmacologia , Proteínas do Tecido Nervoso/fisiologia , Neuropeptídeo Y/farmacologia , Neuropeptídeo Y/fisiologia , Neuropeptídeos/efeitos dos fármacos , Sistemas Neurossecretores/efeitos dos fármacos , Sistemas Neurossecretores/metabolismo
16.
Gen Comp Endocrinol ; 281: 91-104, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31121165

RESUMO

Two structurally related peptides, arginine vasotocin (AVT) and mesotocin (MT), are reported to regulate many physiological processes, such as anti-diuresis and oviposition in birds, and their actions are likely mediated by four AVT/MT receptors (AVPR1A, AVPR1B, MTR and AVPR2b), which are orthologous/paralogous to human AVPR1A, AVPR1B, OXTR and AVPR2 respectively. However, our knowledge regarding the functions of these avian AVT/MT receptors has been limited. Here, we examined the functionality and expression of these receptors in chickens and investigated the roles of AVT in the anterior pituitary. Our results showed that 1) AVPR1A, AVPR1B and AVPR2b could be preferentially activated by AVT, monitored by cell-based luciferase reporter assays and/or Western blot, indicating that they are AVT-specific receptors (AVPR1A; AVPR1B) or AVT-preferring receptor (AVPR2b) functionally coupled to intracellular calcium, MAPK/ERK and cAMP/PKA signaling pathways. In contrast, MTR could be activated by AVT and MT with similar potencies, indicating that MTR is a receptor common for both peptides; 2) Using qPCR, differential expression of the four receptors was found in chicken tissues including the oviduct and anterior pituitary. In particular, only AVPR1A is abundantly expressed in the uterus, suggesting its involvement in mediating AVT-induced oviposition. 3) In cultured chick pituitary cells, AVT could stimulate ACTH and PRL expression and secretion, an action likely mediated by AVPR1B and/or AVPR1A abundantly expressed in anterior pituitary. Collectively, our data helps to elucidate the roles of AVT/MT in birds, such as the 'oxytocic action' of AVT, which induces uterine muscle contraction during oviposition.


Assuntos
Oviposição/fisiologia , Hipófise/metabolismo , Prolactina/metabolismo , Receptores do Hormônio Hipofisário/metabolismo , Receptores de Vasopressinas/metabolismo , Transdução de Sinais , Vasotocina/metabolismo , Sequência de Aminoácidos , Animais , Células Cultivadas , Galinhas/metabolismo , Patos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Modelos Biológicos , Pró-Opiomelanocortina/farmacologia , Prolactina/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Distribuição Tecidual , Vasotocina/química
17.
Aging Cell ; 17(1)2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29168299

RESUMO

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


Assuntos
Estradiol/farmacologia , Comportamento Alimentar/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Pró-Opiomelanocortina/farmacologia , Animais , Núcleo Arqueado do Hipotálamo/efeitos dos fármacos , Núcleo Arqueado do Hipotálamo/metabolismo , Comportamento Animal/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Hipotálamo/metabolismo , Leptina/metabolismo , Camundongos Transgênicos , Neurônios/metabolismo , Obesidade/metabolismo , Pró-Opiomelanocortina/metabolismo
18.
J Mol Endocrinol ; 56(4): T39-48, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26759392

RESUMO

The peptide hormones contained within the sequence of proopiomelanocortin (POMC) have diverse roles ranging from pigmentation to regulation of adrenal function to control of our appetite. It is generally acknowledged to be the archetypal hormone precursor, and as its biology has been unravelled, so too have many of the basic principles of hormone biosynthesis and processing. This short review focuses on one group of its peptide products, namely, those derived from the N-terminal of POMC and their role in the regulation of adrenal growth. From a historical and a personal perspective, it describes how their role in regulating proliferation of the adrenal cortex was identified and also highlights the key questions that remain to be answered.


Assuntos
Glândulas Suprarrenais/crescimento & desenvolvimento , Glândulas Suprarrenais/metabolismo , Fragmentos de Peptídeos/metabolismo , Pró-Opiomelanocortina/metabolismo , Glândulas Suprarrenais/efeitos dos fármacos , Hormônio Adrenocorticotrópico/química , Hormônio Adrenocorticotrópico/metabolismo , Hormônio Adrenocorticotrópico/farmacologia , Animais , Endocrinologia/história , História do Século XX , Humanos , Camundongos Knockout , Receptores Nucleares Órfãos/metabolismo , Fragmentos de Peptídeos/farmacologia , Pró-Opiomelanocortina/química , Pró-Opiomelanocortina/história , Pró-Opiomelanocortina/farmacologia , Ligação Proteica , Proteólise , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais
19.
PLoS One ; 9(10): e108657, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25279464

RESUMO

The adrenal gland is a dynamic organ that undergoes constant cell turnover. This allows for rapid organ remodeling in response to the physiological demands of the HPA axis, which is controlled by proopiomelanocortin (POMC)-derived peptides, such as adrenocorticotropic hormone (ACTH) and N-Terminal peptides (N-POMC). In the rat adrenal cortex, POMC-derived peptides trigger a mitogenic effect, and this process increases cyclins D and E, while inhibiting p27Kip1. The goal of the present study was to further explore the mitogenic effect of ACTH and synthetic N-POMC1-28 peptides by investigating the differences in the expression of key genes involved in the cell cycle of the rat adrenal cortex, following inhibition of the HPA axis. Moreover, we evaluated the differences between the inner and outer fractions of the adrenal cortex (ZF-fraction and ZG-fraction) in terms of their response patterns to different stimuli. In the current study, the inhibition of the HPA axis repressed the expression of Ccnb2, Camk2a, and Nek2 genes throughout the adrenal cortex, while treatments with POMC-derived peptides stimulated Nek2, gene and protein expression, and Notch2 gene expression. Furthermore, Notch1 protein expression was restricted to the subcapsular region of the cortex, an area of the adrenal cortex that is well-known for proliferation. We also showed that different regions of the adrenal cortex respond to HPA-axis inhibition and to induction with POMC-derived peptides at different times. These results suggest that cells in the ZG and ZF fractions could be at different phases of the cell cycle. Our results contribute to the understanding of the mechanisms involved in cell cycle regulation in adrenocortical cells triggered by N-POMC peptides and ACTH, and highlight the involvement of genes such as Nek2 and Notch.


Assuntos
Córtex Suprarrenal/efeitos dos fármacos , Hormônio Adrenocorticotrópico/farmacologia , Proliferação de Células/efeitos dos fármacos , Dexametasona/farmacologia , Fragmentos de Peptídeos/farmacologia , Pró-Opiomelanocortina/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptor Notch1/metabolismo , Córtex Suprarrenal/citologia , Córtex Suprarrenal/metabolismo , Animais , Proliferação de Células/fisiologia , Ratos
20.
Mol Cell Endocrinol ; 371(1-2): 166-73, 2013 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-23219869

RESUMO

The Adrenocorticotropic hormone (ACTH) and Pro-opimelanocortin (POMC) 1-28N-terminal peptide (N-POMC(1-28)) have been shown to act as an adrenal mitogen in vivo. A possible role for cyclin E in the zona glomerulosa (ZG) proliferation, following ACTH and/or N-POMC(1-28) administration, has been previously demonstrated. In this study, we investigated the effect of ACTH and N-POMC(1-28) on the expression of adrenal cortex proteins related to cell cycle control such as cyclins D and P27(kip1). The administration of N-POMC upregulated cyclin D1 and D2 expression in the outer zone of the adrenal cortex; cyclin D3 expression was upregulated in the cortex inner zone even after administration of ACTH. Both ACTH and N-POMC peptides induced a decrease in the P27(kip1) expression in the ZG. These novel findings suggest that the POMC-derivate peptides, ACTH and N-POMC, promote proliferation in the adrenal cortex by upregulating the D2 and D3 cyclins and downregulating the P27(kip1) expression.


Assuntos
Córtex Suprarrenal/metabolismo , Hormônio Adrenocorticotrópico/farmacologia , Ciclina D/biossíntese , Inibidor de Quinase Dependente de Ciclina p27/biossíntese , Fragmentos de Peptídeos/farmacologia , Pró-Opiomelanocortina/farmacologia , Córtex Suprarrenal/efeitos dos fármacos , Hormônio Adrenocorticotrópico/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Ciclina D1/biossíntese , Ciclina D2/biossíntese , Ciclina D3/biossíntese , Regulação para Baixo , Masculino , Fragmentos de Peptídeos/metabolismo , Pró-Opiomelanocortina/metabolismo , Ratos , Ratos Sprague-Dawley , Regulação para Cima , Zona Glomerulosa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...