Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Chemosphere ; 265: 129051, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33250229

RESUMO

Bisphenol-S (BPS) and Bisphenol-F (BPF) are current Bisphenol-A (BPA) substitutes. Here we used pancreatic ß-cells from wild type (WT) and estrogen receptor ß (ERß) knockout (BERKO) mice to investigate the effects of BPS and BPF on insulin secretion, and the expression and activity of ion channels involved in ß-cell function. BPS or BPF rapidly increased insulin release and diminished ATP-sensitive K+ (KATP) channel activity. Similarly, 48 h treatment with BPS or BPF enhanced insulin release and decreased the expression of several ion channel subunits in ß-cells from WT mice, yet no effects were observed in cells from BERKO mice. PaPE-1, a ligand designed to preferentially trigger extranuclear-initiated ER pathways, mimicked the effects of bisphenols, suggesting the involvement of extranuclear-initiated ERß pathways. Molecular dynamics simulations indicated differences in ERß ligand-binding domain dimer stabilization and solvation free energy among different bisphenols and PaPE-1. Our data suggest a mode of action involving ERß whose activation alters three key cellular events in ß-cell, namely ion channel expression and activity, and insulin release. These results may help to improve the hazard identification of bisphenols.


Assuntos
Receptor beta de Estrogênio , Receptores de Estrogênio , Animais , Compostos Benzidrílicos/toxicidade , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Insulina , Canais Iônicos , Camundongos , Fenóis , Receptores de Estrogênio/genética
2.
Food Chem Toxicol ; 145: 111681, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32805339

RESUMO

Bisphenol-A (BPA) is a widespread endocrine disrupting chemical that constitutes a risk factor for type 2 diabetes mellitus (T2DM). Data from animal and human studies have demonstrated that early exposure to BPA results in adverse metabolic outcomes in adult life. In the present work, we exposed pregnant heterozygous estrogen receptor ß (ERß) knock out (BERKO) mice to 10 µg/kg/day BPA, during days 9-16 of pregnancy, and measured ß-cell mass and proliferation in wildtype (WT) and BERKO male offspring at postnatal day 30. We observed increased pancreatic ß-cell proliferation and mass in WT, yet no effect was produced in BERKO mice. Dispersed islet cells in primary culture treated with 1 nM BPA showed an enhanced pancreatic ß-cell replication rate, which was blunted in pancreatic ß-cells from BERKO mice and mimicked by the selective ERß agonist WAY200070. This increased ß-cell proliferation was found in male adult as well as in neonate pancreatic ß-cells, suggesting that BPA directly impacts ß-cell division at earliest stages of life. These findings strongly indicate that BPA during pregnancy upregulates pancreatic ß-cell division and mass in an ERß-dependent manner. Thus, other natural or artificial chemicals may use this ERß-mediated pathway to promote similar effects.


Assuntos
Compostos Benzidrílicos/toxicidade , Receptor beta de Estrogênio/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Exposição Materna/efeitos adversos , Fenóis/toxicidade , Efeitos Tardios da Exposição Pré-Natal/etiologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Receptor beta de Estrogênio/genética , Feminino , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Knockout , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética
3.
Diabetologia ; 62(9): 1667-1680, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31250031

RESUMO

AIMS/HYPOTHESIS: Bisphenol-A (BPA) is a widespread endocrine-disrupting chemical that has been associated with type 2 diabetes development. Low doses of BPA modify pancreatic beta cell function and induce insulin resistance; some of these effects are mediated via activation of oestrogen receptors α (ERα) and ß (ERß). Here we investigated whether low doses of BPA regulate the expression and function of ion channel subunits involved in beta cell function. METHODS: Microarray gene profiling of isolated islets from vehicle- and BPA-treated (100 µg/kg per day for 4 days) mice was performed using Affymetrix GeneChip Mouse Genome 430.2 Array. Expression level analysis was performed using the normalisation method based on the processing algorithm 'robust multi-array average'. Whole islets or dispersed islets from C57BL/6J or oestrogen receptor ß (ERß) knockout (Erß-/-) mice were treated with vehicle or BPA (1 nmol/l) for 48 h. Whole-cell patch-clamp recordings were used to measure Na+ and K+ currents. mRNA expression was evaluated by quantitative real-time PCR. RESULTS: Microarray analysis showed that BPA modulated the expression of 1440 probe sets (1192 upregulated and 248 downregulated genes). Of these, more than 50 genes, including Scn9a, Kcnb2, Kcnma1 and Kcnip1, encoded important Na+ and K+ channel subunits. These findings were confirmed by quantitative RT-PCR in islets from C57BL/6J BPA-treated mice or whole islets treated ex vivo. Electrophysiological measurements showed a decrease in both Na+ and K+ currents in BPA-treated islets. The pharmacological profile indicated that BPA reduced currents mediated by voltage-activated K+ channels (Kv2.1/2.2 channels) and large-conductance Ca2+-activated K+ channels (KCa1.1 channels), which agrees with BPA's effects on gene expression. Beta cells from ERß-/- mice did not present BPA-induced changes, suggesting that ERß mediates BPA's effects in pancreatic islets. Finally, BPA increased burst duration, reduced the amplitude of the action potential and enlarged the action potential half-width, leading to alteration in beta cell electrical activity. CONCLUSIONS/INTERPRETATION: Our data suggest that BPA modulates the expression and function of Na+ and K+ channels via ERß in mouse pancreatic islets. Furthermore, BPA alters beta cell electrical activity. Altogether, these BPA-induced changes in beta cells might play a role in the diabetogenic action of BPA described in animal models.


Assuntos
Compostos Benzidrílicos/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Receptor beta de Estrogênio/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Fenóis/farmacologia , Animais , Receptor alfa de Estrogênio/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Potássio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Sódio/metabolismo
4.
J Steroid Biochem Mol Biol ; 176: 16-22, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28159674

RESUMO

Endocrine Disrupting Chemicals (EDCs), including bisphenol-A (BPA) do not act as traditional toxic chemicals inducing massive cell damage or death in an unspecific manner. EDCs can work upon binding to hormone receptors, acting as agonists, antagonists or modulators. Bisphenol-A displays estrogenic activity and, for many years it has been classified as a weak estrogen, based on the classic transcriptional action of estrogen receptors serving as transcription factors. However, during the last two decades our knowledge about estrogen signaling has advanced considerably. It is now accepted that estrogen receptors ERα and ERß activate signaling pathways outside the nucleus which may or may not involve transcription. In addition, a new membrane estrogen receptor, GPER, has been proposed. Pharmacological and molecular evidence, along with results obtained in genetically modified mice, demonstrated that BPA, and its substitute BPS, are potent estrogens acting at nanomolar concentrations via extranuclear ERα, ERß, and GPER. The different signaling pathways activated by BPA and BPS explain the well-known estrogenic effects of low doses of EDCs as well as non-monotonic dose-response relationships. These signaling pathways may help to explain the actions of EDCs with estrogenic activity in the etiology of different pathologies, including type-2 diabetes and obesity.


Assuntos
Disruptores Endócrinos/farmacologia , Estrogênios/farmacologia , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Toxicologia/tendências , Animais , Núcleo Celular , Regulação da Expressão Gênica , Humanos
5.
Sci Rep ; 7(1): 11770, 2017 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-28924161

RESUMO

In regulatory toxicology, the dose-response relationship is a key element towards fulfilling safety assessments and satisfying regulatory authorities. Conventionally, the larger the dose, the greater the response, following the dogma "the dose makes the poison". Many endocrine disrupting chemicals, including bisphenol-A (BPA), induce non-monotonic dose response (NMDR) relationships, which are unconventional and have tremendous implications in risk assessment. Although several molecular mechanisms have been proposed to explain NMDR relationships, they are largely undemonstrated. Using mouse pancreatic ß-cells from wild-type and oestrogen receptor ERß-/- mice, we found that exposure to increasing doses of BPA affected Ca2+ entry in an NMDR manner. Low doses decreased plasma membrane Ca2+ currents after downregulation of Cav2.3 ion channel expression, in a process involving ERß. High doses decreased Ca2+ currents through an ERß-mediated mechanism and simultaneously increased Ca2+ currents via oestrogen receptor ERα. The outcome of both molecular mechanisms explains the NMDR relationship between BPA and Ca2+ entry in ß-cells.


Assuntos
Compostos Benzidrílicos/toxicidade , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/metabolismo , Células Secretoras de Insulina/metabolismo , Fenóis/toxicidade , Animais , Canais de Cálcio Tipo R/biossíntese , Canais de Cálcio Tipo R/genética , Sinalização do Cálcio/genética , Proteínas de Transporte de Cátions/biossíntese , Proteínas de Transporte de Cátions/genética , Relação Dose-Resposta a Droga , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Knockout
6.
Metabolism ; 65(3): 54-63, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26892516

RESUMO

OBJECTIVE: While bile acids are important for the digestion process, they also act as signaling molecules in many tissues, including the endocrine pancreas, which expresses specific bile acid receptors that regulate several cell functions. In this study, we investigated the effects of the conjugated bile acid TUDCA on glucose-stimulated insulin secretion (GSIS) from pancreatic ß-cells. METHODS: Pancreatic islets were isolated from 90-day-old male mice. Insulin secretion was measured by radioimmunoassay, protein phosphorylation by western blot, Ca(2+) signals by fluorescence microscopy and ATP-dependent K(+) (KATP) channels by electrophysiology. RESULTS: TUDCA dose-dependently increased GSIS in fresh islets at stimulatory glucose concentrations but remained without effect at low glucose levels. This effect was not associated with changes in glucose metabolism, Ca(2+) signals or KATP channel activity; however, it was lost in the presence of a cAMP competitor or a PKA inhibitor. Additionally, PKA and CREB phosphorylation were observed after 1-hour incubation with TUDCA. The potentiation of GSIS was blunted by the Gα stimulatory, G protein subunit-specific inhibitor NF449 and mimicked by the specific TGR5 agonist INT-777, pointing to the involvement of the bile acid G protein-coupled receptor TGR5. CONCLUSION: Our data indicate that TUDCA potentiates GSIS through the cAMP/PKA pathway.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , AMP Cíclico/fisiologia , Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Relação Dose-Resposta a Droga , Técnicas In Vitro , Células Secretoras de Insulina/metabolismo , Canais KATP/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos
7.
PLoS One ; 10(7): e0132884, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26222156

RESUMO

This study evaluated the chronotropic and inotropic responses to glucagon in spontaneously beating isolated right atria of rat heart. For comparison, we also investigated the effects resulting from stimulating ß-adrenoceptors with isoproterenol in this tissue. Isoproterenol increased both atrial frequency and contractility but glucagon only enhanced atrial rate. The transcript levels of glucagon receptors were about three times higher in sinoatrial node than in the atrial myocardium. Chronotropic responses to glucagon and isoproterenol were blunted by the funny current (If) inhibitor ZD 7288. Inhibitors of protein kinase A, H-89 and KT-5720 reduced the chronotropic response to glucagon but not to isoproterenol. Inhibition of ryanodine receptors and calcium/calmodulin dependent protein kinase II (important regulators of sarcoplasmic reticulum Ca2+ release), with ruthenium red and KN-62 respectively, failed to alter chronotropic responses of either glucagon or isoproterenol. Non selective inhibition of phosphodiesterase (PDE) with 3-isobutylmethylxantine or selective inhibition of PDE3 or PDE4 with cilostamide or rolipram respectively did not affect chronotropic effects of glucagon or isoproterenol. Our results indicate that glucagon increases beating rate but not contractility in rat right atria which could be a consequence of lower levels of glucagon receptors in atrial myocardium than in sinoatrial node. Chronotropic responses to glucagon or isoproterenol are mediated by If current but not by sarcoplasmic reticulum Ca2+ release, neither are regulated by PDE activity.


Assuntos
Função do Átrio Direito/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Glucagon/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Isoproterenol/farmacologia , Contração Miocárdica/efeitos dos fármacos , Animais , Função do Átrio Direito/fisiologia , Sinalização do Cálcio/fisiologia , Carbazóis/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Átrios do Coração , Isoquinolinas/farmacologia , Masculino , Miocárdio , Inibidores da Fosfodiesterase 3/farmacologia , Inibidores da Fosfodiesterase 4/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Ratos , Ratos Sprague-Dawley , Sulfonamidas/farmacologia
8.
Diabetes ; 62(6): 2015-25, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23349481

RESUMO

The estrogen receptor ß (ERß) is emerging as an important player in the physiology of the endocrine pancreas. We evaluated the role and antidiabetic actions of the ERß selective agonist WAY200070 as an insulinotropic molecule. We demonstrate that WAY200070 enhances glucose-stimulated insulin secretion both in mouse and human islets. In vivo experiments showed that a single administration of WAY200070 leads to an increase in plasma insulin levels with a concomitant improved response to a glucose load. Two-week treatment administration increased glucose-induced insulin release and pancreatic ß-cell mass and improved glucose and insulin sensitivity. In addition, streptozotocin-nicotinamide-induced diabetic mice treated with WAY200070 exhibited a significant improvement in plasma insulin levels and glucose tolerance as well as a regeneration of pancreatic ß-cell mass. Studies performed in db/db mice demonstrated that this compound restored first-phase insulin secretion and enhanced pancreatic ß-cell mass. We conclude that ERß agonists should be considered as new targets for the treatment of diabetes.


Assuntos
Receptor beta de Estrogênio/agonistas , Hipoglicemiantes/farmacologia , Oxazóis/farmacologia , Fenóis/farmacologia , Animais , Células Cultivadas , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/tratamento farmacológico , Ensaio de Imunoadsorção Enzimática , Humanos , Técnicas In Vitro , Insulina/sangue , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Niacinamida/farmacologia , Estreptozocina/farmacologia
9.
PLoS One ; 7(3): e33814, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22470480

RESUMO

Bisphenol-A (BPA) is one of the most widespread endocrine disrupting chemicals (EDC) used as the base compound in the manufacture of polycarbonate plastics. Although evidence points to consider exposure to BPA as a risk factor for insulin resistance, its actions on whole body metabolism and on insulin-sensitive tissues are still unclear. The aim of the present work was to study the effects of low doses of BPA in insulin-sensitive peripheral tissues and whole body metabolism in adult mice. Adult mice were treated with subcutaneous injection of 100 µg/kg BPA or vehicle for 8 days. Whole body energy homeostasis was assessed with in vivo indirect calorimetry. Insulin signaling assays were conducted by western blot analysis. Mice treated with BPA were insulin resistant and had increased glucose-stimulated insulin release. BPA-treated mice had decreased food intake, lower body temperature and locomotor activity compared to control. In skeletal muscle, insulin-stimulated tyrosine phosphorylation of the insulin receptor ß subunit was impaired in BPA-treated mice. This impairment was associated with a reduced insulin-stimulated Akt phosphorylation in the Thr(308) residue. Both skeletal muscle and liver displayed an upregulation of IRS-1 protein by BPA. The mitogen-activated protein kinase (MAPK) signaling pathway was also impaired in the skeletal muscle from BPA-treated mice. In the liver, BPA effects were of lesser intensity with decreased insulin-stimulated tyrosine phosphorylation of the insulin receptor ß subunit.In conclusion, short-term treatment with low doses of BPA slows down whole body energy metabolism and disrupts insulin signaling in peripheral tissues. Thus, our findings support the notion that BPA can be considered a risk factor for the development of type 2 diabetes.


Assuntos
Metabolismo Energético/efeitos dos fármacos , Estrogênios não Esteroides/toxicidade , Fenóis/toxicidade , Animais , Compostos Benzidrílicos , Glucose/metabolismo , Injeções Subcutâneas , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos
10.
PLoS One ; 7(2): e31109, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22347437

RESUMO

Bisphenol-A (BPA) is a widespread endocrine-disrupting chemical (EDC) used as the base compound in the manufacture of polycarbonate plastics. It alters pancreatic ß-cell function and can be considered a risk factor for type 2 diabetes in rodents. Here we used ERß-/- mice to study whether ERß is involved in the rapid regulation of K(ATP) channel activity, calcium signals and insulin release elicited by environmentally relevant doses of BPA (1 nM). We also investigated these effects of BPA in ß-cells and whole islets of Langerhans from humans. 1 nM BPA rapidly decreased K(ATP) channel activity, increased glucose-induced [Ca(2+)](i) signals and insulin release in ß-cells from WT mice but not in cells from ERß-/- mice. The rapid reduction in the K(ATP) channel activity and the insulinotropic effect was seen in human cells and islets. BPA actions were stronger in human islets compared to mouse islets when the same BPA concentration was used. Our findings suggest that BPA behaves as a strong estrogen via nuclear ERß and indicate that results obtained with BPA in mouse ß-cells may be extrapolated to humans. This supports that BPA should be considered as a risk factor for metabolic disorders in humans.


Assuntos
Receptor beta de Estrogênio/fisiologia , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Fenóis/toxicidade , Poluentes Ocupacionais do Ar/toxicidade , Animais , Compostos Benzidrílicos , Sinalização do Cálcio , Diabetes Mellitus Tipo 2/induzido quimicamente , Disruptores Endócrinos/toxicidade , Estrogênios não Esteroides , Humanos , Camundongos , Camundongos Knockout , Fenóis/administração & dosagem , Canais de Potássio
11.
Mol Cell Endocrinol ; 355(2): 201-7, 2012 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-22227557

RESUMO

Bisphenol-A (BPA) is an estrogenic monomer commonly used in the manufacture of numerous consumer products such as food and beverage containers. Widespread human exposure to significant doses of this compound has been reported. Traditionally, BPA has been considered a weak estrogen, based on its lower binding affinity to the nuclear estrogen receptors (ERs) compared to 17-ß estradiol (E2) as well as its low transcriptional activity after ERs activation. However, in vivo animal studies have demonstrated that it can interfere with endocrine signaling pathways at low doses during fetal, neonatal or perinatal periods as well as in adulthood. In addition, mounting evidence suggests a variety of pathways through which BPA can elicit cellular responses at very low concentrations with the same or even higher efficiency than E2. Thus, the purpose of the present review is to analyze with substantiated scientific evidence the strong estrogenic activity of BPA when it acts through alternative mechanisms of action at least in certain cell types.


Assuntos
Estrogênios/farmacologia , Fenóis/farmacologia , Transdução de Sinais , Animais , Compostos Benzidrílicos , Exposição Ambiental , Estradiol/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Receptores de Estrogênio/metabolismo
12.
Steroids ; 76(9): 856-60, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21470558

RESUMO

ATP-sensitive potassium channels (K(ATP)) regulate electrical activity and insulin secretion in pancreatic ß-cells. When glucose concentration increases, the [ATP]/[ADP] ratio rises closing K(ATP) channels, and the membrane potential depolarizes, triggering insulin secretion. This pivotal role of K(ATP) channels is used not only by glucose but also by neurotransmitters, hormones and other physiological agents to modulate electrical and secretory ß-cell response. In recent years, it has been demonstrated that estrogens and estrogen receptors are involved in glucose homeostasis, and that they can modulate the electrical activity and insulin secretion of pancreatic ß-cells. The hormone 17ß-estradiol (E2), at physiological levels, is implicated in maintaining normal insulin sensitivity for ß-cell function. Long term exposure to E2 increases insulin content, insulin gene expression and insulin release via the estrogen receptor α (ERα), while rapid responses to E2 can regulate K(ATP) channels increasing cGMP levels through the estrogen receptor ß (ERß) and type A guanylate cyclase receptor (GC-A). This review summarizes the main actions of 17ß-estradiol on K(ATP) channels and the subsequent insulin release in pancreatic ß-cells.


Assuntos
Estradiol/metabolismo , Estrogênios/metabolismo , Células Secretoras de Insulina/metabolismo , Canais KATP/metabolismo , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Humanos , Insulina/metabolismo , Secreção de Insulina , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Receptores de Droga/metabolismo , Receptores de Estrogênio/metabolismo , Receptores Acoplados a Guanilato Ciclase/metabolismo , Receptores de Sulfonilureias
13.
Front Biosci (Landmark Ed) ; 16(1): 251-60, 2011 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-21196169

RESUMO

Estrogen receptors (ER) are emerging as important molecules involved in the adaptation of beta-cells to insulin resistance. The onset of type 2 diabetes is marked by insulin secretory dysfunction and decreased beta-cell mass. During pregnancy, puberty and obesity there is increased metabolic demand and insulin resistance is developed. This metabolic state increases the demand on beta-cells to augment insulin biosynthesis and release. In this respect, ERalpha is directly implicated in the E2-regulation of insulin content and secretion, while ERbeta is in the E2-potentiation of glucose-induced insulin release. Both receptors develop their actions within the physiological range of E2. In addition, the G protein-coupled estrogen receptor (GPER1/GPR30) seems to be implicated in the E2-regulation of stimulus-secretion coupling in the three cell types of the islet. The increased demand of insulin production for long time may lead to beta-cell stress and apoptosis. ERalpha, ERbeta and GPER1/GPR30 are involved in preventing beta-cell apoptosis, impeding the loss of critical beta-cell mass. Therefore, estrogen receptors may play an essential role in the adaptation of the pancreas to insulin resistant periods.


Assuntos
Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Células Secretoras de Insulina/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Animais , Glicemia/metabolismo , Disruptores Endócrinos/farmacologia , Homeostase , Humanos , Insulina/biossíntese , Insulina/metabolismo , Secreção de Insulina , Camundongos , Receptores de Estrogênio
14.
Hormones (Athens) ; 9(2): 118-26, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20687395

RESUMO

The aim of this review was to analyze the potential effects of environmental chemicals on homeostatic control related to glycemia and energy balance. Many of the environmental chemicals can mimic or interfere with the action of hormones and are generally referred to as "endocrine disruptors". Among these compounds, polychlorinated biphenyls, dioxins, phthalates and bisphenol-A have been correlated with alterations in blood glucose homeostasis in humans. In rodents it has been demonstrated that small doses of bisphenol-A have profound effects on glucose metabolism. Therefore, this altered blood glucose homeostasis may enhance the development of type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/induzido quimicamente , Disruptores Endócrinos/efeitos adversos , Poluentes Ambientais/efeitos adversos , Estrogênios não Esteroides/efeitos adversos , Fenóis/efeitos adversos , Animais , Compostos Benzidrílicos , Glicemia/efeitos dos fármacos , Diabetes Mellitus Tipo 2/sangue , Metabolismo Energético/efeitos dos fármacos , Estradiol/metabolismo , Medicina Baseada em Evidências , Homeostase , Humanos , Medição de Risco , Fatores de Risco
15.
Metabolism ; 59(5): 635-44, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-19913855

RESUMO

A regimen of low-protein diet induces a reduction of pancreatic islet function that is associated with development of metabolic disorders including diabetes and obesity afterward. In the present study, the influence of leucine supplementation on metabolic parameters, insulin secretion to glucose and to amino acids, as well as the levels of proteins that participate in the phosphatidylinositol 3-phosphate kinase (PI3K) pathway was investigated in malnourished rats. Four groups were fed with different diets for 12 weeks: a normal protein diet (17%) without (NP) or with leucine supplementation (NPL) or a low (6%)-protein diet without (LP) or with leucine supplementation (LPL). Leucine was given in the drinking water during the last 4 weeks. As indicated by the intraperitoneal glucose tolerance test, LPL rats exhibited increased glucose tolerance as compared with NPL group. Both NPL and LPL rats had higher circulating insulin levels than controls. The LPL rats also showed increased insulin secretion by pancreatic islets in response to glucose or arginine compared with those observed in islets from LP animals. Glucose oxidation was significantly reduced in NPL, LP, and LPL isolated islets as compared with NP; but no alteration was observed for leucine and glutamate oxidation among the 4 groups. Western blotting analysis demonstrated increased PI3K and mammalian target protein of rapamycin protein contents in LPL compared with LP islets. A significant increase in insulin-induced insulin receptor substrate 1-associated PI3K activation was also observed in LPL compared with LP islets. These findings indicate that leucine supplementation can augment islet function in malnourished rats and that activation of the PI3K/mammalian target protein of rapamycin pathway may play a role in this process.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Leucina/administração & dosagem , Desnutrição/metabolismo , Animais , Proteínas Sanguíneas/metabolismo , Peso Corporal , Diabetes Mellitus Tipo 2/tratamento farmacológico , Suplementos Nutricionais , Teste de Tolerância a Glucose , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Desnutrição/tratamento farmacológico , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA/química , RNA/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas Quinases S6 Ribossômicas/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Serina-Treonina Quinases TOR
16.
Mol Endocrinol ; 23(12): 1973-82, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19855088

RESUMO

The ATP-sensitive potassium (K(ATP)) channel is a key molecule involved in glucose-stimulated insulin secretion. The activity of this channel regulates beta-cell membrane potential, glucose- induced [Ca(2+)](i) signals, and insulin release. In this study, the rapid effect of physiological concentrations of 17beta-estradiol (E2) on K(ATP) channel activity was studied in intact beta-cells by use of the patch-clamp technique. When cells from wild-type (WT) mice were used, 1 nm E2 rapidly reduced K(ATP) channel activity by 60%. The action of E2 on K(ATP) channel was not modified in beta-cells from ERalpha-/- mice, yet it was significantly reduced in cells from ERbeta-/- mice. The effect of E2 was mimicked by the ERbeta agonist 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN). Activation of ERbeta by DPN enhanced glucose-induced Ca(2+) signals and insulin release. Previous evidence indicated that the acute inhibitory effects of E2 on K(ATP) channel activity involve cyclic GMP and cyclic GMP-dependent protein kinase. In this study, we used beta-cells from mice with genetic ablation of the membrane guanylate cyclase A receptor for atrial natriuretic peptide (also called the atrial natriuretic peptide receptor) (GC-A KO mice) to demonstrate the involvement of this membrane receptor in the rapid E2 actions triggered in beta-cells. E2 rapidly inhibited K(ATP) channel activity and enhanced insulin release in islets from WT mice but not in islets from GC-A KO mice. In addition, DPN reduced K(ATP) channel activity in beta-cells from WT mice, but not in beta-cells from GC-A KO mice. This work unveils a new role for ERbeta as an insulinotropic molecule that may have important physiological and pharmacological implications.


Assuntos
Estradiol/farmacologia , Receptor beta de Estrogênio/fisiologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Canais KATP/efeitos dos fármacos , Canais KATP/metabolismo , Receptores do Fator Natriurético Atrial/fisiologia , Animais , Cálcio/metabolismo , Células Cultivadas , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/genética , Feminino , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Nitrilas/farmacologia , Técnicas de Patch-Clamp , Fenóis , Pirazóis/farmacologia , Receptores do Fator Natriurético Atrial/genética
17.
J Physiol ; 587(Pt 21): 5031-7, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19687125

RESUMO

Pregnancy is characterized by peripheral insulin resistance, which is developed in parallel with a plasma increase of maternal hormones; these include prolactin, placental lactogens, progesterone and oestradiol among others. Maternal insulin resistance is counteracted by the adaptation of the islets of Langerhans to the higher insulin demand. If this adjustment is not produced, gestational diabetes may be developed. The adaptation process of islets is characterized by an increase of insulin biosynthesis, an enhanced glucose-stimulated insulin secretion (GSIS) and an increase of beta-cell mass. It is not completely understood why, in some individuals, beta-cell mass and function fail to adapt to the metabolic demands of pregnancy, yet a disruption of the beta-cell response to maternal hormones may play a key part. The role of the maternal hormone 17beta-oestradiol (E2) in this adaptation process has been largely unknown. However, in recent years, it has been demonstrated that E2 acts directly on beta-cells to increase insulin biosynthesis and to enhance GSIS through different molecular mechanisms. E2 does not increase beta-cell proliferation but it is involved in beta-cell survival. Classical oestrogen receptors ERalpha and ERbeta, as well as the G protein-coupled oestrogen receptor (GPER) seem to be involved in these adaptation changes. In addition, as the main production of E2 in post-menopausal women comes from the adipose tissue, E2 may act as a messenger between adipocytes and islets in obesity.


Assuntos
Estrogênios/metabolismo , Resistência à Insulina/fisiologia , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Gravidez/metabolismo , Transdução de Sinais/fisiologia , Animais , Feminino , Humanos
18.
Mol Cell Endocrinol ; 304(1-2): 63-8, 2009 May 25.
Artigo em Inglês | MEDLINE | ID: mdl-19433249

RESUMO

The estrogen receptor ERalpha is emerging as a key molecule involved in glucose and lipid metabolism. The main functions of pancreatic beta-cells are the biosynthesis and release of insulin, the only hormone that can directly decrease blood glucose levels. Estrogen receptors ERalpha and ERbeta exist in beta-cells. The role of ERbeta is still unknown, yet ERalpha plays an important role in the regulation of insulin biosynthesis, insulin secretion and beta-cell survival. Activation of ERalpha by 17beta-estradiol (E2) and the environmental estrogen bisphenol-A (BPA) promotes an increase of insulin biosynthesis through a non-classical estrogen-activated pathway that involves phosphorylation of ERK1/2. The activation of ERalpha by physiological concentrations of E2 may play an important role in the adaptation of the endocrine pancreas to pregnancy. However, if ERalpha is over stimulated by an excess of E2 or the action of an environmental estrogen such as BPA, it will produce an excessive insulin signaling. This may provoke insulin resistance in the liver and muscle, as well as beta-cell exhaustion and therefore, it may contribute to the development of type II diabetes.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Homeostase , Células Secretoras de Insulina/fisiologia , Animais , Compostos Benzidrílicos , Estradiol/metabolismo , Receptor beta de Estrogênio/metabolismo , Estrogênios não Esteroides/farmacologia , Feminino , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efeitos dos fármacos , Fenóis/farmacologia , Gravidez , Transdução de Sinais/fisiologia
19.
Cell Signal ; 21(4): 523-8, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19136060

RESUMO

Embryonic stem cells (ESC) are pluripotent and could be maintained in vitro in a self-renewing state indefinitely, at the same time preserving their potential to differentiate towards more specific lineages. Despite the progress in the field, the complex network of signalling cascades involved in the maintenance of the self-renewing and pluripotent state remains not fully understood. In the present study, we have investigated the role of lysophosphatidic acid (LPA), a potent mitogen present in serum, in Ca(2+) signalling and early gene activation in mouse ESC (mESC). In these cells, we detected the expression of the G-protein coupled LPA receptor subtypes LPA(1), LPA(2) and LPA(3). Using fluorescence Ca(2+) imaging techniques, we showed that LPA induced an increase in intracellular Ca(2+) concentration. This increase was also observed in the absence of extracellular Ca(2+), suggesting the involvement of internal stores. Pre-treatment with BAPTA-AM, thapsigargin or U-73122 efficiently blocked this Ca(2+) release, indicating that LPA was evoking Ca(2+) mobilization from the endoplasmic reticulum via the phospholipase C (PLC) pathway. Interestingly, this signalling cascade initiated by LPA was involved in inducing the expression of the Ca(2+)-dependent early response gene c-myc, a key gene implicated in ESC self-renewal and pluripotency. Additionally, LPA increased the proliferation rate of mESC. Our findings therefore outline the physiological role of LPA in mESC.


Assuntos
Sinalização do Cálcio/efeitos dos fármacos , Células-Tronco Embrionárias/efeitos dos fármacos , Genes myc/efeitos dos fármacos , Lisofosfolipídeos/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/biossíntese , Receptores de Ácidos Lisofosfatídicos/fisiologia , Fosfolipases Tipo C/fisiologia , Animais , Compartimento Celular , Divisão Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Replicação do DNA/efeitos dos fármacos , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Células-Tronco Embrionárias/citologia , Retículo Endoplasmático/metabolismo , Estrenos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Células-Tronco Pluripotentes/citologia , Pirrolidinonas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptores de Ácidos Lisofosfatídicos/biossíntese , Receptores de Ácidos Lisofosfatídicos/efeitos dos fármacos , Receptores de Ácidos Lisofosfatídicos/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/fisiologia , Tapsigargina/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores
20.
Infect Disord Drug Targets ; 8(1): 61-4, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18473909

RESUMO

Rapid estrogen actions are triggered after estrogens are bound to a variety of proteins in organelles other than the nucleus. Those include classic estrogen receptors ERalpha and ERbeta, novel membrane proteins that behave as estrogen receptors such as GPR30, ion channels, and other ligand receptors. In pancreatic alpha and beta-cells, estrogens binding to a non-classical membrane estrogen receptors at physiological concentrations regulate ion channels and [Ca(2+)](i) signals, provoking important physiological responses. In beta-cells, 17beta-estradiol regulates K(ATP) channel activity and glucose-induced [Ca(2+)](i) oscillations, eliciting changes in insulin release and the activation of Ca(2+)-dependent transcription factors. In alpha-cells, 17beta-estradiol abolishes low glucose-induced [Ca(2+)](i) oscillations.


Assuntos
Estrogênios/fisiologia , Células Secretoras de Glucagon/fisiologia , Células Secretoras de Insulina/fisiologia , Animais , Canais de Cálcio/fisiologia , GMP Cíclico/metabolismo , Estradiol/fisiologia , Humanos , Canais KATP/fisiologia , Transdução de Sinais , Fatores de Transcrição/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA