Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Life Sci ; 307: 120854, 2022 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-35917939

RESUMO

AIMS: Synthetic glucocorticoids, including dexamethasone (DEX), are clinically prescribed due to their immunoregulatory properties. In excess they can perturb glucose homeostasis, with individuals predisposed to glucose intolerance more sensitive to these negative effects. While DEX is known to negatively impact ß-cell function, it is unclear how. Hence, our aim was to investigate the effect of DEX on ß-cell function, both alone and in combination with a diabetogenic milieu in the form of elevated glucose and palmitate. MAIN METHODS: Human pancreatic EndoC-ßH1 cells were cultured in the presence of high glucose and palmitate (glucolipotoxicity) and/or a pharmacological concentration of DEX, before functional and molecular analyses. KEY FINDINGS: Either treatment alone resulted in reduced insulin content and secretion, while the combination of DEX and glucolipotoxicity promoted a strong synergistic effect. These effects were associated with reduced insulin biosynthesis, likely due to downregulation of PDX1, MAFA, and the proinsulin converting enzymes, as well as reduced ATP response upon glucose stimulation. Genome-wide DNA methylation analysis found changes on PDE4D, MBNL1 and TMEM178B, all implicated in ß-cell function, after all three treatments. DEX alone caused very strong demethylation of the glucocorticoid-regulated gene ZBTB16, also known to influence the ß-cell, while the combined treatment caused altered methylation of many known ß-cell regulators and diabetes candidate genes. SIGNIFICANCE: DEX treatment and glucolipotoxic conditions separately alter the ß-cell epigenome and function. The combination of both treatments exacerbates these changes, showing that caution is needed when prescribing potent glucocorticoids in patients with dysregulated metabolism.


Assuntos
Glucocorticoides , Células Secretoras de Insulina , Trifosfato de Adenosina/metabolismo , Dexametasona/metabolismo , Dexametasona/toxicidade , Epigenoma , Glucocorticoides/metabolismo , Glucocorticoides/farmacologia , Glucose/metabolismo , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Palmitatos/farmacologia , Proinsulina/metabolismo , Proinsulina/farmacologia
2.
Arch. endocrinol. metab. (Online) ; 66(3): 312-323, June 2022. tab, graf
Artigo em Inglês | LILACS-Express | LILACS | ID: biblio-1393858

RESUMO

ABSTRACT Objectives: To evaluate the effect of sitagliptin treatment in early type 2 diabetes mellitus (T2DM) and the impact of different macronutrient compositions on hormones and substrates during meal tolerance tests (MTT). Materials and methods: Half of the drug-naive patients with T2DM were randomly assigned for treatment with 100 mg of sitagliptin, q.d., or placebo for 4 weeks and then submitted to 3 consecutive MTT intercalated every 48 h. The MTTs differed in terms of macronutrient composition, with 70% of total energy from carbohydrates, proteins, or lipids. After 4 weeks of washout, a crossover treatment design was repeated. Both patients and researchers were blinded, and a repeated-measures ANOVA was employed for statistical analysis. Results: Sitagliptin treatment reduced but did not normalize fasting and post-meal glucose values in the three MTTs, with lowered area-under-glucose-curve values varying from 7% to 15%. The sitagliptin treatment also improved the insulinogenic index (+86%) and the insulin/glucose (+25%), glucagon-like peptide-1/glucose (+46%) incremental area under the curves. Patients with early T2DM maintained the lowest glucose excursion after a protein- or lipid-rich meal without any major change in insulin, C-peptide, glucagon, or NEFA levels. Conclusion: We conclude that sitagliptin treatment is tolerable and contributes to better control of glucose homeostasis in early T2DM, irrespective of macronutrient composition. The blood glucose excursion during meal ingestion is minimal in protein- or fat-rich meals, which can be a positive ally for the management of T2DM. Clinical trial no: NCT00881543

3.
FASEB J ; 35(8): e21828, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34325494

RESUMO

Since prenatal glucocorticoids (GC) excess increases the risk of metabolic dysfunctions in the offspring and its effect on ß-cell recovery capacity remains unknown we investigated these aspects in offspring from mice treated with dexamethasone (DEX) in the late pregnancy. Half of the pups were treated with streptozotocin (STZ) on the sixth postnatal day (PN). Functional and molecular analyses were performed in male offspring on PN25 and PN225. Prenatal DEX treatment resulted in low birth weight. At PN25, both the STZ-treated offspring developed hyperglycemia and had lower ß-cell mass, in parallel with higher α-cell mass and glucose intolerance, with no impact of prenatal DEX on such parameters. At PN225, the ß-cell mass was partially recovered in the STZ-treated mice, but they remained glucose-intolerant, irrespective of being insulin sensitive. Prenatal exposition to DEX predisposed adult offspring to sustained hyperglycemia and perturbed islet function (lower insulin and higher glucagon response to glucose) in parallel with exacerbated glucose intolerance. ß-cell-specific knockdown of the Hnf4α in mice from the DS group resulted in exacerbated glucose intolerance. We conclude that high GC exposure during the prenatal period exacerbates the metabolic dysfunctions in adult life of mice exposed to STZ early in life, resulting in a lesser ability to recover the islets' function over time. This study alerts to the importance of proper management of exogenous GCs during pregnancy and a healthy postnatal lifestyle since the combination of adverse factors during the prenatal and postnatal period accentuates the predisposition to metabolic disorders in adult life.


Assuntos
Dexametasona/toxicidade , Glucocorticoides/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/fisiologia , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Dexametasona/administração & dosagem , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/administração & dosagem , Teste de Tolerância a Glucose , Insulina/farmacologia , Camundongos , Neoplasias Experimentais , Gravidez , Efeitos Tardios da Exposição Pré-Natal , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
4.
Life Sci ; 277: 119509, 2021 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-33865877

RESUMO

AIMS: Pregnancy is associated with the development of a transitory insulin resistance that parallels with the upregulation of pancreatic ß-cell function and mass. These metabolic adaptations guarantee the higher insulin demand, but there is no evidence of whether insulin clearance contributes to this process. Thus, we investigated some of the hepatic parameters related to insulin clearance during rat pregnancy. We also investigated some molecular parameters in the hypothalamus. MAIN METHODS: We evaluated the body mass and food intake, insulin sensitivity, ß- and α-cell masses, insulin clearance based on an exogenous insulin load, hepatic insulin-degrading enzyme (IDE) activity, and hepatic and hypothalamic protein content of IDE and carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM-1) in three periods of gestation in Wistar rats. KEY FINDINGS: In the first week of pregnancy, both insulin sensitivity and clearance increased, a pattern that inverted in the third week of gestation (reduced insulin sensitivity and clearance). Diminished insulin clearance was associated with lower hepatic IDE activity and higher pancreatic ß- and α-cell masses. No alteration in the hepatic IDE and CEACAM protein content was observed throughout pregnancy, but hypothalamic IDE protein content was significantly reduced in the late gestation period. SIGNIFICANCE: In conclusion, elevated insulin demand in the late period of gestation occurs not only as a result of increased ß-cell mass and function but also by a potential reduction in hepatic insulin clearance. Knowing this physiological process may be valuable when considering gestational diabetes mellitus results from a failure in insulin supply during pregnancy.


Assuntos
Células Secretoras de Glucagon/metabolismo , Resistência à Insulina/fisiologia , Células Secretoras de Insulina/metabolismo , Insulisina/metabolismo , Animais , Glicemia/metabolismo , Tamanho Celular , Diabetes Gestacional/fisiopatologia , Feminino , Glucose/metabolismo , Teste de Tolerância a Glucose , Hiperinsulinismo/metabolismo , Insulina/metabolismo , Fígado/metabolismo , Fígado/patologia , Masculino , Gravidez , Ratos , Ratos Wistar
5.
Cancer Lett ; 502: 44-57, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33429006

RESUMO

Obesity is a major risk factor for breast cancer, especially in post-menopausal women. In the breast tissue of obese women, cyclooxygenase-2 (COX-2)-dependent prostaglandin E2 (PGE2) production has been correlated with inflammation and local estrogen biosynthesis via aromatase. Using a mouse model of 7,12-dimethylbenz[a]anthracene/medroxyprogesterone-acetate (DMBA/MPA)-induced carcinogenesis, we demonstrated that an obesogenic diet promotes mammary tissue inflammation and local estrogen production, and accelerates mammary tumor formation in a COX-2-dependent manner. High-sugar/fat (HSF) diet augmented the levels of the pro-inflammatory mediators MCP-1, IL-6, COX-2, and PGE2 in mammary tissue, and this was accompanied by crown-like structures of breast (CLS-B) formation and aromatase/estrogen upregulation. Treatment with a COX-2 selective inhibitor, etoricoxib, decreased PGE2, IL-6, MCP-1, and CLS-B formation as well as reduced aromatase protein and estrogen levels in the mammary tissue of mice fed a HSF diet. Etoricoxib-treated mice showed increased latency and decreased incidence of mammary tumors, which resulted in prolonged animal survival when compared to HSF diet alone. Inhibition of tumor angiogenesis also seemed to account for the prolonged survival of COX-2 inhibitor-treated animals. In conclusion, obesogenic diet-induced COX-2 is sufficient to trigger inflammation, local estrogen biosynthesis, and mammary tumorigenesis.


Assuntos
Neoplasias da Mama/metabolismo , Ciclo-Oxigenase 2/metabolismo , Dieta Hiperlipídica/efeitos adversos , Dinoprostona/biossíntese , Açúcares/efeitos adversos , Regulação para Cima , 9,10-Dimetil-1,2-benzantraceno/efeitos adversos , Animais , Aromatase/metabolismo , Neoplasias da Mama/induzido quimicamente , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Quimiocina CCL2/metabolismo , Modelos Animais de Doenças , Etoricoxib/administração & dosagem , Etoricoxib/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Interleucina-6/metabolismo , Células MCF-7 , Acetato de Medroxiprogesterona/efeitos adversos , Camundongos
6.
J Nutr Biochem ; 90: 108572, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33388348

RESUMO

We investigated whether combined long-term fructose and prednisolone intake would be more detrimental to the glucose homeostasis than if ingested separately. We also evaluated whether fish oil administration or interruption of treatments has any positive impact. For this, male adult Wistar rats ingested fructose (20%) (F) or prednisolone (12.5 µg/mL) (P) or both (FP) through drinking water for 12 weeks. A separate group of fructose and prednisolone-treated rats received fish oil treatment (1 g/kg) in the last 6 weeks. In another group, the treatment with fructose and prednisolone was interrupted after 12 weeks, and the animals were followed for more 12 weeks. Control groups ran in parallel (C). The F group had higher plasma TG (+42%) and visceral adiposity (+63%), whereas the P group had lower insulin sensitivity (-33%) and higher insulinemia (+200%). Only the the FP group developed these alterations combined with higher circulating uric acid (+126%), hepatic triacylglycerol content (+16.2-fold), lipid peroxidation (+173%) and lower catalase activity (-32%) that were associated with lower protein kinase B content and AMP-activated protein kinase (AMPK) phosphorylation in the liver, lower AMPK phosphorylation in the adipose tissue and higher beta-cell mass. Fish oil ingestion attenuated the elevation in circulating triacylglycerol and uric acid values, while the interruption of sugar and glucocorticoid intake reverted almost all modified parameters. In conclusion, long-term intake of fructose and prednisolone by male rats are more detrimental to glucose and lipid homeostasis than if ingested separately and the benefits of treatment interruption are broader than fish oil treatment.


Assuntos
Óleos de Peixe/farmacologia , Frutose/farmacologia , Glucocorticoides/farmacologia , Glucose/metabolismo , Metabolismo dos Lipídeos , Prednisolona/farmacologia , Tecido Adiposo/metabolismo , Adiposidade/efeitos dos fármacos , Animais , Óleos de Peixe/administração & dosagem , Frutose/administração & dosagem , Glucocorticoides/administração & dosagem , Homeostase , Humanos , Insulina/metabolismo , Resistência à Insulina , Peroxidação de Lipídeos , Fígado/metabolismo , Masculino , Prednisolona/administração & dosagem , Ratos , Ratos Wistar , Bebidas Adoçadas com Açúcar , Triglicerídeos/sangue , Ácido Úrico/metabolismo
7.
J Nutr Biochem ; 86: 108494, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32920089

RESUMO

Overfeeding and rapid weight gain during early life are risk factors for the development of obesity in adulthood. This metabolic malprogramming may be mediated by endocrine disturbances during critical periods of development. Cholecystokinin (CCK) acts on the central nervous system by elevating thermogenesis and the activity of anorectic neurons, modulating overall energy balance. Therefore, we tested the hypothesis that postnatal overfeeding impaired CCK effects. Pups were raised in either a litter of three (neonatal overnutrition/small litter group) or 12 (controls/normal litter group) pups per dam to study the effects of postnatal overfeeding on the central and peripheral CCK systems in adulthood. Rats raised in small litters became overweight during lactation and remained overweight as adults, with increased adiposity and plasma levels of lipids, glucose, insulin, and leptin. Neonatally over-nourished rats showed attenuation of gastric emptying and anorexigenic response to CCK, suggesting that offspring from the SL group may present CCK resistance as adult male rats. Consistent with this idea, overweight rats displayed impaired central response in c-Fos immunoreactivity on the nucleus tractus solitarius, area postrema, paraventricular nucleus, central amygdala, arcuate nucleus, and dorsomedial hypothalamus in response to peripheral CCK at adulthood. The small litter group of adult male rats also exhibited reduced norepinephrine- and CCK-stimulated thermogenesis. Unresponsiveness to the effects of CCK may contribute to overweight and metabolic dysfunctions observed in postnatally over-nourished adult rats. Thus, the involvement of an impaired CCK system, among other neurohormonal failures, may contribute to the development of obesity.


Assuntos
Adiposidade , Sistema Nervoso Central/fisiopatologia , Colecistocinina/metabolismo , Sistema Endócrino/fisiopatologia , Hipernutrição/fisiopatologia , Tecido Adiposo/metabolismo , Animais , Animais Recém-Nascidos , Mapeamento Encefálico , Metabolismo Energético , Feminino , Esvaziamento Gástrico , Glucose/metabolismo , Homeostase , Hipotálamo , Leptina/sangue , Lipídeos/química , Masculino , Obesidade/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ratos , Ratos Wistar , Termogênese , Aumento de Peso
8.
J Cell Physiol ; 233(1): 486-496, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28370189

RESUMO

In the present study, we investigated the relationship between early life protein malnutrition-induced redox imbalance, and reduced glucose-stimulated insulin secretion. After weaning, male Wistar rats were submitted to a normal-protein-diet (17%-protein, NP) or to a low-protein-diet (6%-protein, LP) for 60 days. Pancreatic islets were isolated and hydrogen peroxide (H2 O2 ), oxidized (GSSG) and reduced (GSH) glutathione content, CuZn-superoxide dismutase (SOD1), glutathione peroxidase (GPx1) and catalase (CAT) gene expression, as well as enzymatic antioxidant activities were quantified. Islets that were pre-incubated with H2 O2 and/or N-acetylcysteine, were subsequently incubated with glucose for insulin secretion measurement. Protein malnutrition increased CAT mRNA content by 100%. LP group SOD1 and CAT activities were 50% increased and reduced, respectively. H2 O2 production was more than 50% increased whereas GSH/GSSG ratio was near 60% lower in LP group. Insulin secretion was, in most conditions, approximately 50% lower in LP rat islets. When islets were pre-incubated with H2 O2 (100 µM), and incubated with glucose (33 mM), LP rats showed significant decrease of insulin secretion. This effect was attenuated when LP islets were exposed to N-acetylcysteine.


Assuntos
Glicemia/metabolismo , Dieta com Restrição de Proteínas , Insulina/sangue , Ilhotas Pancreáticas/metabolismo , Estresse Oxidativo , Desnutrição Proteico-Calórica/metabolismo , Fenômenos Fisiológicos da Nutrição Animal , Animais , Antioxidantes/farmacologia , Catalase/genética , Catalase/metabolismo , Modelos Animais de Doenças , Regulação Enzimológica da Expressão Gênica , Glutationa/metabolismo , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Peróxido de Hidrogênio/metabolismo , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Estado Nutricional , Oxirredução , Estresse Oxidativo/efeitos dos fármacos , Desnutrição Proteico-Calórica/sangue , Desnutrição Proteico-Calórica/genética , Desnutrição Proteico-Calórica/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Wistar , Superóxido Dismutase-1/genética , Superóxido Dismutase-1/metabolismo , Fatores de Tempo
9.
Int J Exp Pathol ; 98(6): 329-340, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29226508

RESUMO

Sepsis is associated with high mortality. Both critically ill humans and animal models of sepsis exhibit changes in their glucose homeostasis, that is, hypoglycaemia, with the progression of infection. However, the relationship between basal glycaemia, glucose tolerance and insulin sensitivity is not well understood. Thus, we aimed to evaluate this glucose homeostasis triad at the late stage of sepsis (24 h after surgery) in male Swiss mice subjected to lethal and sublethal sepsis by the caecal ligation and puncture (CLP) model. The percentage of survival 24 h after CLP procedure in the Lethal and Sublethal groups was around 66% and 100% respectively. Both Lethal and Sublethal groups became hypoglycaemic in fasting and fed states 24 h after surgery. The pronounced fed hypoglycaemia in the Lethal group was not due to worsening anorexic behaviour or hepatic inability to deliver glucose in relation to the Sublethal group. Reduction in insulin sensitivity in CLP mice occurred in a lethality-dependent manner and was not associated with glucose intolerance. Analysis of oral and intraperitoneal glucose tolerance tests, as well as the gastrointestinal motility data, indicated that CLP mice had reduced intestinal glucose absorption. Altogether, we suggest cessation of appetite and intestinal glucose malabsorption are key contributors to the hypoglycaemic state observed during experimental severe sepsis.


Assuntos
Glicemia/biossíntese , Ceco/metabolismo , Homeostase/fisiologia , Sepse/mortalidade , Animais , Ceco/cirurgia , Modelos Animais de Doenças , Hipoglicemiantes , Resistência à Insulina , Ligadura/métodos , Fígado/metabolismo , Masculino , Camundongos , Punções/métodos
10.
Biomed Res Int ; 2017: 4378328, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28929113

RESUMO

Background. Continuous fructose consumption may cause elevation of circulating triacylglycerol. However, how much of this alteration is reverted after the removal of fructose intake is not known. We explored this question and compared the efficacy of this approach with fish oil supplementation. Methods. Male Wistar rats were divided into the following groups: control (C), fructose (F) (water intake with 10% or 30% fructose for 9 weeks), fish oil (FO), and fructose/fish oil (FFO). Fish oil was supplemented only for the last 33 days of fructose ingestion. Half of the F group remained for additional 8 weeks without fructose ingestion (FR). Results. Fructose ingestion reduced food intake to compensate for the increased energy obtained through water ingestion, independent of fructose concentration. Fish oil supplementation exerted no impact on these parameters, but the removal of fructose from water recovered both ingestion behaviors. Plasma triacylglycerol augmented significantly during the second and third weeks (both fructose groups). Fish oil supplementation did not attenuate the elevation in triacylglycerol caused by fructose intake, but the interruption of sugar consumption normalized this parameter. Conclusion. Elevation in triacylglyceridemia may be recovered by removing fructose from diet, suggesting that it is never too late to repair improper dietary habits.


Assuntos
Suplementos Nutricionais , Óleos de Peixe/administração & dosagem , Frutose/administração & dosagem , Glucose/metabolismo , Metabolismo dos Lipídeos , Animais , Dieta , Ingestão de Alimentos , Comportamento Alimentar , Óleos de Peixe/metabolismo , Frutose/metabolismo , Homeostase , Masculino , Ratos , Ratos Wistar
11.
Appl Physiol Nutr Metab ; 41(4): 382-90, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26939043

RESUMO

Dexamethasone is an anti-inflammatory glucocorticoid that may alter glucose and lipid homeostasis when administered in high doses or for long periods of time. Omega-3 fatty acids, present in fish oil (FO), can be used as potential modulators of intermediary glucose and lipid metabolism. Herein, we evaluate the effects of FO supplementation (1 g·kg(-1) body weight (BW)) on glucose and lipid metabolism in rats treated with dexamethasone (0.5 mg·kg(-1) BW) for 15 days. Adult male Wistar rats were distributed among 4 groups: control (saline, 1 mL·kg(-1) BW and mineral oil, 1 g·kg(-1) BW), DEX (dexamethasone and mineral oil), FO (fish oil and saline), and DFO (fish oil and dexamethasone). Dexamethasone and saline were administered intraperitoneally, and fish oil and mineral oil were administered by gavage. We evaluated functional and molecular parameters of lipid and glycemic profiles at 8 days and at the end of treatment. FO supplementation increased hepatic docosahexaenoic acid (DEX: 5.6% ± 0.7%; DFO: 10.5% ± 0.8%) and eicosapentaenoic acid (DEX: 0.3% ± 0.0%; DFO: 1.3% ± 0.1%) contents and attenuated the increase of plasma triacylglycerol, total cholesterol, and non-high-density lipoprotein cholesterol concentrations in DFO rats compared with DEX rats. These effects seem not to depend on hepatic expression of insulin receptor substrate 1, protein kinase B, peroxisome proliferator-activated receptor γ coactivator 1-α, and peroxisome proliferator-activated receptor γ. There was no effect of supplementation on body weight loss, fasting glycemia, and glucose tolerance in rats treated with dexamethasone. In conclusion, we show that FO supplementation for 15 days attenuates the dyslipidemia induced by dexamethasone treatment.


Assuntos
HDL-Colesterol/sangue , LDL-Colesterol/sangue , Dexametasona/efeitos adversos , Suplementos Nutricionais , Óleos de Peixe/farmacologia , Triglicerídeos/sangue , Animais , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/metabolismo , Dislipidemias/induzido quimicamente , Dislipidemias/tratamento farmacológico , Ácido Eicosapentaenoico/metabolismo , Ácidos Graxos Ômega-3/farmacologia , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , PPAR gama/genética , PPAR gama/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar
12.
J Steroid Biochem Mol Biol ; 145: 1-12, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25268311

RESUMO

OBJECTIVES: Peripheral insulin resistance (IR) is one of the main side effects caused by glucocorticoid (GC)-based therapies, and the molecular mechanisms of GC-induced IR are not yet fully elucidated. Thus, we aimed to investigate the effects of dexamethasone treatment on the main components of insulin and inflammatory signaling in the adipose tissue of rats. MATERIALS/METHODS: Male Wistar rats received daily injections of dexamethasone (1mg/kg body weight (b.w.), intraperitoneally (i.p.)) for 5 days (DEX), whereas control rats received saline (CTL). The metabolic status was investigated, and the epididymal fat fragments were collected for lipolysis and western blot analyses. RESULTS: The DEX rats became hyperglycemic, hyperinsulinemic, insulin resistant and glucose intolerant, compared with the CTL rats (P<0.05). The basal glycerol release in the fat fragments was 1.5-fold higher in the DEX rats (P<0.05). The phosphorylation of protein kinase B (PKB) at ser(473) decreased by 44%, whereas, the phosphorylation of insulin receptor substrate (IRS)-1 at ser(307) increased by 93% in the adipose tissue of the DEX rats after an oral bolus of glucose (P<0.05). The basal phosphorylation of c-jun-N-terminal kinase (JNK) and inhibitor of nuclear factor kappa-B (IKKß) proteins was reduced by 46% and 58%, respectively, in the adipose tissue of the DEX rats (P<0.05). This was paralleled with a significant reduction (47%) in the glucocorticoid receptor (GR) protein content in the adipose tissue of the DEX rats (P<0.05). CONCLUSION: The insulin-resistant status of rats induced by dexamethasone administration have PKB and IRS-1 activity attenuated in epididymal fat without increases in the phosphorylation of the proinflammatory signals JNK and IKKß.


Assuntos
Tecido Adiposo/metabolismo , Glucocorticoides/metabolismo , Quinase I-kappa B/metabolismo , MAP Quinase Quinase 4/metabolismo , Animais , Peso Corporal , Citocinas/metabolismo , Dexametasona/farmacologia , Epididimo/metabolismo , Glicogênio/metabolismo , Inflamação , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Resistência à Insulina , Masculino , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ácido Pirúvico/metabolismo , Ratos , Ratos Wistar , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
13.
Int J Exp Pathol ; 96(1): 21-30, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25529509

RESUMO

Recent studies have shown a positive association of cancer and obesity, but the morphological and molecular mechanisms involved in this relationship are still unknown. This study analysed the impact of long-term obesity on rat prostate, focusing on stromal changes. Male adult Wistar rats were treated with high-fat diet to induce obesity, while the control group received a balanced diet. After 30 weeks of feeding, the ventral prostate was analysed by immunohistochemistry for cell proliferation, smooth muscle α-actin, vimentin, chondroitin sulphate and metalloproteinases (MMP-2 and 9). The content of androgen receptor (AR), oestrogen receptors (ERs) and vascular endothelial growth factor (VEGF) was measured by Western blotting, and activity of catalase and Glutathione-S-Transferase (GST) were quantified by enzymatic assay. Long-term obesity decreased testosterone plasma levels by 70% and resulted in stromal prostate hyperplasia, as evidenced by increased collagen fibres. Such stromal hyperplasia was associated with increased number of blood vessels and raised VEGF content, and increased expression of chondroitin sulphate, vimentin, α-actin and MMP-9. In spite of the high cell density in prostate, the proliferative activity was lower in the prostates of obese rats, indicating that hyperplasia was established during the early phases in this obesity model. AR levels increased significantly, whereas the ERα decreased in this group. Moreover, the levels of catalase and GST were changed considerably. These findings indicate that long-term obesity, besides disturbing the antioxidant control, causes intense stromal remodelling and release of factors that create an environment that can promote proliferative disorders in the gland, culminating with diffuse hyperplasia.


Assuntos
Matriz Extracelular/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Obesidade/complicações , Próstata/enzimologia , Hiperplasia Prostática/etiologia , Células Estromais/enzimologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Biomarcadores/sangue , Glicemia/metabolismo , Catalase/metabolismo , Proliferação de Células , Microambiente Celular , Modelos Animais de Doenças , Glutationa Transferase/metabolismo , Insulina/sangue , Masculino , Malondialdeído/metabolismo , Oxirredução , Próstata/patologia , Hiperplasia Prostática/sangue , Hiperplasia Prostática/enzimologia , Hiperplasia Prostática/patologia , Ratos Wistar , Receptores Androgênicos/metabolismo , Fatores de Risco , Células Estromais/patologia , Testosterona/sangue , Fatores de Tempo , Regulação para Cima
14.
J Androl ; 33(5): 854-65, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22441765

RESUMO

In this study, we evaluated the effects of obesity and insulin resistance induced by a high-fat diet on prostate morphophysiology, focusing on cell proliferation, expression of androgen (AR) and estrogen receptors (ER) and proteins of the insulin signaling pathway. Adult male Wistar rats were fed a high-fat diet (20% fat) for 15 weeks, whereas control animals received a balanced diet (4% fat). Both groups were then divided and treated for 2 weeks with 1 mg/kg body weight/day of the aromatase inhibitor letrozole or vehicle only. The ventral prostate was analyzed with immunohistochemical, histopathological, stereological, and Western blotting methods. Obese rats showed insulin resistance, hyperinsulinemia, and reduced plasma testosterone levels. The incidence of prostatic intraepithelial neoplasia (PIN) was 2.7 times higher in obese rats and affected 0.4% of the gland compared with 0.1% PIN areas found in control rats. Obesity doubled cell proliferation in both prostate epithelium and stroma. AR content decreased in the prostate of obese rats and estrogen receptor beta (ERß) increased in this group. Protein levels of insulin receptor substrate 1 and protein kinase B diminished in the obese group, whereas phosphatidylinositol 3-kinase (PI3K) increased significantly. Most structural changes observed in the prostate of obese rats normalized after letrozole treatment, except for increased stromal cell proliferation and ERß expression, which might be associated with insulin resistance. This experimental model of obesity and insulin resistance induced by a high-fat diet increases cell proliferation in rat prostate. Such alterations are associated with decreased levels of AR and increased ERß and PI3K proteins. This change can facilitate the establishment of proliferative lesions in rat prostate.


Assuntos
Proliferação de Células , Dieta Hiperlipídica , Receptor beta de Estrogênio/metabolismo , Resistência à Insulina , Obesidade/etiologia , Fosfatidilinositol 3-Quinase/metabolismo , Próstata/metabolismo , Animais , Inibidores da Aromatase/farmacologia , Western Blotting , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Hiperinsulinismo/etiologia , Hiperinsulinismo/metabolismo , Imuno-Histoquímica , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Letrozol , Masculino , Nitrilas/farmacologia , Obesidade/metabolismo , Obesidade/patologia , Obesidade/fisiopatologia , Próstata/efeitos dos fármacos , Próstata/patologia , Neoplasia Prostática Intraepitelial/etiologia , Neoplasia Prostática Intraepitelial/metabolismo , Neoplasias da Próstata/etiologia , Neoplasias da Próstata/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Receptores Androgênicos/metabolismo , Transdução de Sinais , Células Estromais/metabolismo , Testosterona/sangue , Fatores de Tempo , Triazóis/farmacologia
15.
Biol. Res ; 44(3): 251-257, 2011. ilus
Artigo em Inglês | LILACS | ID: lil-608621

RESUMO

Chronic administration of glucocorticoids induces insulin resistance that is compensated by an increase in p-cell function and mass. Since insulin signaling is involved in the control of p-cell function and mass, we investigated the content of insulin pathway proteins in pancreatic islets. Rats were made insulin resistant by daily administration of dexamethasone (1mg/kg, b.w., i.p.) for 5 consecutive days (DEX), whilst control rats received saline (CTL). Circulating insulin and insulin released from isolated islets were measured by radioimmunoassay whereas the content of proteins was analyzed by Western blotting. DEX rats were hyperinsulinemic and exhibited augmented insulin secretion in response to glucose (P < 0.01). The IRa-subunit, IRS-1, Shc, AKT, p-p70S6K, ERK1/2, p-ERK1/2, and glucocorticoid receptor protein levels were similar between DEX and CTL islets. However, the IRp-subunit, p-IRp-subunit, IRS-2, PI3-K, p-AKT and p70S6K protein contents were increased in DEX islets (P < 0.05). We conclude that IRS-2 may have a major role, among the immediate substrates of the insulin receptor, to link activated receptors to downstream signaling components related to islet function and growth in this insulin-resistant rat model.


Assuntos
Animais , Masculino , Ratos , Dexametasona/efeitos adversos , Glucocorticoides/efeitos adversos , Resistência à Insulina , Proteínas Substratos do Receptor de Insulina/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Insulina , Ilhotas Pancreáticas/metabolismo , Ratos Wistar , Transdução de Sinais , Proteínas Adaptadoras da Sinalização Shc/metabolismo
16.
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
17.
Am J Physiol Endocrinol Metab ; 296(4): E681-9, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19158320

RESUMO

Activation of insulin signaling and cell cycle intermediates is required for adult beta-cell proliferation. Here, we report a model to study beta-cell proliferation in living rats by administering three different doses of dexamethasone (0.1, 0.5, and 1.0 mg/kg ip, DEX 0.1, DEX 0.5, and DEX 1.0, respectively) for 5 days. Insulin sensitivity, insulin secretion, and histomorphometric data were investigated. Western blotting was used to analyze the levels of proteins related to the control of beta-cell growth. DEX 1.0 rats, which present moderate hyperglycemia and marked hyperinsulinemia, exhibited a 5.1-fold increase in beta-cell proliferation and an increase (17%) in beta-cell size, with significant increase in beta-cell mass, compared with control rats. The hyperinsulinemic but euglycemic DEX 0.5 rats also showed a significant 3.6-fold increase in beta-cell proliferation. However, DEX 0.1 rats, which exhibited the lowest degree of insulin resistance, compensate for insulin demand by improving only islet function. Activation of the insulin receptor substrate 2/phosphatidylinositol 3-kinase/serine-threonine kinase/ribosomal protein S6 kinase pathway, as well as protein retinoblastoma in islets from DEX 1.0 and DEX 0.5, but not in DEX 0.1, rats was also observed. Therefore, increasing doses of dexamethasone induce three different degrees of insulin requirement in living rats, serving as a model to investigate compensatory beta-cell alterations. Augmented beta-cell mass involves beta-cell hyperplasia and, to a lower extent, beta-cell hypertrophy. We suggest that alterations in circulating insulin and, to a lesser extent, glucose levels could be the major stimuli for beta-cell proliferation in the dexamethasone-induced insulin resistance.


Assuntos
Proliferação de Células/efeitos dos fármacos , Dexametasona/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Animais , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Dexametasona/administração & dosagem , Relação Dose-Resposta a Droga , Resistência à Insulina , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/fisiologia , Ilhotas Pancreáticas/anatomia & histologia , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiologia , Masculino , Síndrome Metabólica/induzido quimicamente , Síndrome Metabólica/patologia , Tamanho do Órgão , Ratos , Ratos Wistar
18.
Int J Exp Pathol ; 89(4): 264-75, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18429991

RESUMO

It is well known that glucocorticoids induce peripheral insulin resistance in rodents and humans. Here, we investigated the structural and ultrastructural modifications, as well as the proteins involved in beta-cell function and proliferation, in islets from insulin-resistant rats. Adult male Wistar rats were made insulin resistant by daily administration of dexamethasone (DEX; 1mg/kg, i.p.) for five consecutive days, whilst control (CTL) rats received saline alone. Structure analyses showed a marked hypertrophy of DEX islets with an increase of 1.7-fold in islet mass and of 1.6-fold in islet density compared with CTL islets (P < 0.05). Ultrastructural evaluation of islets revealed an increased amount of secreting organelles, such as endoplasmic reticulum and Golgi apparatus in DEX islets. Mitotic figures were observed in DEX islets at structural and ultrastructural levels. Beta-cell proliferation, evaluated at the immunohistochemical level using anti-PCNA (proliferating cell nuclear antigen), showed an increase in pancreatic beta-cell proliferation of 6.4-fold in DEX islets compared with CTL islets (P < 0.0001). Increases in insulin receptor substrate-2 (IRS-2), phosphorylated-serine-threonine kinase AKT (p-AKT), cyclin D(2) and a decrease in retinoblastoma protein (pRb) levels were observed in DEX islets compared with CTL islets (P < 0.05). Therefore, during the development of insulin resistance, the endocrine pancreas adapts itself increasing beta-cell mass and proliferation, resulting in an amelioration of the functions. The potential mechanisms that underlie these events involve the activation of the IRS-2/AKT pathway and activation of the cell cycle, mediated by cyclin D(2). These adaptations permit the maintenance of glycaemia at near-physiological ranges.


Assuntos
Ciclinas/análise , Resistência à Insulina , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ilhotas Pancreáticas/patologia , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/fisiologia , Animais , Proteínas de Ciclo Celular/análise , Proliferação de Células , Ciclina D2 , Ciclinas/metabolismo , Dexametasona , Glucagon/análise , Técnicas Imunoenzimáticas , Insulina/análise , Proteínas Substratos do Receptor de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Microscopia Eletrônica de Transmissão , Modelos Animais , Ratos , Ratos Wistar
19.
Biosci. j. (Online) ; 23(3)July-Sept. 2007.
Artigo em Português | LILACS | ID: lil-482726

RESUMO

Terapias a base de glicocorticóides estão freqüentemente associadas a alteração da sensibilidade à insulina. No presente trabalho avaliamos alguns parâmetros metabólicos como glicose, insulina, proteínas e colesterolplasmáticos em ratos tratados com dexametasona (DEX) (1mg/kg, peso corpóreo, ip.) por diferentes períodos de tempo (24h, 72h e 120h). Os ratos tratados com dexametasona apresentaram resistência periférica à insulina após 24h de administração da droga como indicam os valores de insulina plasmática de jejum (1,3 vs. 6,8 ng/ml para ratos controle [CTL] e DEX, respectivamente) e do índice HOMA. Resistência periférica à insulina adicional ocorre até o final do tratamento nos ratos DEX. A glicemia permanece moderadamente elevada até o período de 72h. Entretanto, observa-se marcante hiperglicemia após 120h (79 vs. 160 mg/dl para ratos CTL e DEX, respectivamente). Aumento significativo dos níveis de proteínas totais e albuminas plasmáticas ocorre a partir de 72h de tratamento e de colesterol total a partir de 120h. Glicogênio e gordura hepáticos aumentam de maneira tempo-dependente nos ratos DEX. Correlação negativa foi observada entre os valores de insulinemia de jejum e peso nos grupos tratados com dexametasona (r > 0,95). Portanto, administração de dexametasona, 1mg/kg, induz resistência periférica à insulina de maneira tempo-dependente a partir de 24h e aumento dos níveis circulantes de glicose e proteínas plasmáticos após 72h de tratamento.


Glucocorticoid therapies are often associated with insulin sensitivity alteration. In the present study we evaluated some metabolical parameters such as plasma glucose, insulin, protein and cholesterol levels in ratstreated with dexamethasone (DEX) (1mg/kg, body weight, ip.) in different periods (24h, 72h and 120h). Dexamethasonetreated rats show peripherical resistance after 24h of drug administration as indicated by the fasting plasma insulin values (1.3 vs.6.8 ng/ml for controls [CTL] and DEX rats, respectively) and by HOMA index. Additional peripheral insulin resistance occurred until the end of treatment in DEX rats. The glycaemia remained slightly elevated until 72h period. However, marked hyperglycaemia was observed after 120h (79 vs.160 mg/dl for CTL and DEX rats, respectively). Significantly increase of plasma albumin and total proteins levels occurred from 72h of treatment and total cholesterol from 120h. Hepatic glycogen and hepatic fat increased in a time-dependent manner in DEX rats. Negative correlation was observed between fasting insulin and body weight values in dexamethasone-treated groups (r > 0.95). Therefore, dexamethasone administration, 1mg/kg, induces insulin peripheral resistance in a time-dependent manner from 24h and increase of circulating plasma glucose and proteins levels after 72h of treatment.


Assuntos
Animais , Ratos , Carboidratos , Dexametasona , Gorduras , Proteínas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA