Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cell Biochem Funct ; 42(2): e3976, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38489223

ABSTRACT

Fluoride (F) has been employed worldwide to control dental caries. More recently, it has been suggested that the consumption of low doses of F in the drinking water may reduce blood glucose levels, introducing a new perspective for the use of F for the management of blood glucose. However, the exact mechanism by which F affects blood glucose levels remains largely unexplored. Given that the small gut plays a pivotal role in glucose homeostasis, the aim of this study was to investigate the proteomic changes induced by low doses of F in the ileum of female nonobese-diabetic (NOD) mice. Forty-two female NOD mice were divided into two groups based on the F concentration in their drinking water for 14 weeks: 0 (control) or 10 mgF/L. At the end of the experimental period, the ileum was collected for proteomic and Western blot analyses. Proteomic analysis indicated an increase in isoforms of actin, gastrotropin, several H2B histones, and enzymes involved in antioxidant processes, as well as a decrease in enzymes essential for energy metabolism. In summary, our data indicates an adaptive response of organism to preserve protein synthesis in the ileum, despite significant alterations in energy metabolism typically induced by F, therefore highlighting the safety of controlled fluoridation in water supplies.


Subject(s)
Dental Caries , Drinking Water , Mice , Animals , Female , Fluorides/pharmacology , Fluorides/analysis , Mice, Inbred NOD , Blood Glucose/analysis , Proteomics , Drinking Water/analysis , Ileum/chemistry , Ileum/metabolism
2.
J Agric Food Chem ; 66(6): 1419-1427, 2018 Feb 14.
Article in English | MEDLINE | ID: mdl-29345910

ABSTRACT

Although Jatropha aethiopica, popularly known in Cuba as "mata diabetes", is used in salads and as a dietary supplement, its chemical composition and antidiabetic properties yet remains unclear. In this work, we evaluate the qualitative and quantitative composition of ethanolic extract (EE) and phenolic fraction (PF) of Jatropha aethiopica leaves and their hypoglycemic and hypolipidemic activity. Chemical fractionation of the ethanolic extract yielded nine compounds, which included protocatechuic acid (1), chlorogenic acid (2), caffeic acid (3), quercetin 3-O-α-l-rhamnopyranosyl-(1 → 2)-[α-l-rhamnopyranolsyl-(1 → 6)]-ß-d-galactopyranoside (4), a new kaempferol 3-O-α-l-rhamnopyranosyl-(1 → 4)-[α-l-rhamnopyranolsyl-(1 → 6)]-ß-d-galactopyranoside (5), kaempferol 3-O-α-l-rhamnopyranosyl-(1 → 2)-[α-l-rhamnopyranolsyl-(1 → 6)]-ß-d-glucopyranoside (6), rutin (7), kaempferol 3-O-α-l-rhamnopyranosyl-(1 → 6)-ß-d-glucopyranoside (8), and quercetin (9). The compounds (1, 4-7) were quantified by high-performance liquid chromatography photodiode array detection (HPLC-PDA) in both the ethanolic extract (62.65 ± 0.15 mg/g) and phenolic fraction (61.72 ± 0.23 mg/g). The results obtained show that both ethanolic extract and phenolic fraction contributed toward the improvement of glucose tolerance, which in turn led to a decline in the glucose levels. Remarkably, the ethanolic extract presented a relatively higher promising effect compared to metformin.


Subject(s)
Diabetes Mellitus/drug therapy , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/isolation & purification , Jatropha/chemistry , Phenols/chemistry , Phenols/isolation & purification , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Animals , Blood Glucose/metabolism , Diabetes Mellitus/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Male , Mice , Phenols/administration & dosage , Plant Extracts/administration & dosage , Plant Leaves/chemistry
3.
Int J Exp Pathol ; 95(5): 351-63, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25186305

ABSTRACT

Glucocorticoids (GCs) induce insulin resistance (IR), a condition known to alter oral homeostasis. This study investigated the effects of long-term dexamethasone administration on morphofunctional aspects of salivary glands. Male Wistar rats received daily injections of dexamethasone [0.1 mg/kg body weight (b.w.), intraperitoneally] for 10 days (DEX), whereas control rats received saline. Subsequently, glycaemia, insulinaemia, insulin secretion and salivary flow were analysed. The parotid and submandibular glands were collected for histomorphometric evaluation and Western blot experiments. The DEX rats were found to be normoglycaemic, hyperinsulinaemic, insulin resistant and glucose intolerant (P < 0.05). DEX rat islets secreted more insulin in response to glucose (P < 0.05). DEX rats had significant reductions in the masses of the parotid (29%) and submandibular (16%) glands (P < 0.05) that was associated with reduced salivary flux rate. The hypotrophy in both glands observed in the DEX group was associated with marked reduction in the volume of the acinar cells in these glands of 50% and 26% respectively (P < 0.05). The total number of acinar cells was increased in the submandibular glands of the DEX rats (P < 0.05) but not in the parotid glands. The levels of proteins related to insulin and survival signalling in both glands did not differ between the groups. In conclusion, the long-term administration of dexamethasone caused IR, which was associated with significant reductions in both mass and flux rate of the salivary glands. The parotid and submandibular glands exhibited reduced acinar cell volume; however, the submandibular glands displayed acinar hyperplasia, indicating a gland-specific response to GCs. Our data emphasize that GC-based therapies and insulin-resistant states have a negative impact on salivary gland homeostasis.


Subject(s)
Acinar Cells/cytology , Dexamethasone/pharmacology , Parotid Gland/drug effects , Salivary Glands/drug effects , Submandibular Gland/pathology , Acinar Cells/drug effects , Animals , Blood Glucose/metabolism , Cell Shape , Glucocorticoids/metabolism , Insulin/metabolism , Male , Parotid Gland/metabolism , Rats , Rats, Wistar , Salivary Glands/metabolism , Submandibular Gland/metabolism , Time
4.
Int J Exp Pathol ; 93(3): 188-95, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22583132

ABSTRACT

Previous studies by our research group using a model of insulin resistance induced by dexamethasone (DEX) showed that in the rat ventral prostate there was epithelial and smooth muscle cell atrophy and there were also alterations in fibroblasts. Proteins of the insulin signalling pathway are known to be very important for cell proliferation and development. Thus, we investigated the insulin signalling pathway and epithelial proliferation in the rat ventral prostate in this model and correlated the findings with expression of glucocorticoid (GR) and androgen (AR) receptors. Insulin resistance was induced in adult male Wistar rats by injection of DEX (1 mg/kg, ip for 5 consecutive days), whereas control (CTL) rats received saline. DEX treatment resulted in a significant decrease in body weight, but not in prostate weight. Reductions in insulin receptor 1 (IRS-1) (CTL 1.11 ± 0.06; DEX 0.85 ± 0.03), IRS-2 (CTL 0.95 ± 0.05; DEX 0.49 ± 0.04), AKT (CTL 0.98 ± 0.03; DEX 0.78 ± 0.02), mammalian target of rapamycin (mTOR; CTL 0.65 ± 0.08; DEX 0.22 ± 0.05), GR (CTL 1.30 ± 0.09; DEX 0.57 ± 0.10) and AR (CTL 1.83 ± 0.16; DEX 0.55 ± 0.08) protein levels were observed in the prostate of DEX-treated rats. The expression of the IRα-subunit, phosphoinositide 3-kinase, p-AKT, p70(S6K) , extracellular signal-regulated kinase (ERK) and p-ERK was not altered. The frequency of AR-positive cells in the epithelium of the prostate decreased in the glucocorticoid-treated group, and the intensity of the reaction for this receptor in the cell nuclei was lower in this group. Furthermore, the treatment with DEX reduced the frequency of proliferating cell nuclear antigen-positive (PCNA) cells 30-fold. This study suggests that the reduction in the insulin signalling pathway proteins IRS-1/IRS-2/AKT/mTOR in the prostate of DEX-treated rats may be associated with the morphological alterations observed previously.


Subject(s)
Cell Proliferation/drug effects , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Glucocorticoids/pharmacology , Insulin/metabolism , Prostate/drug effects , Signal Transduction/drug effects , Animals , Epithelial Cells/metabolism , Insulin Receptor Substrate Proteins/metabolism , Male , Prostate/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , TOR Serine-Threonine Kinases/metabolism
5.
Gen Physiol Biophys ; 31(1): 65-76, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22447832

ABSTRACT

Chronic administration of glucocorticoids (GC) leads to characteristic features of type 2 diabetes in mammals. The main action of dexamethasone in target cells occurs through modulation of gene expression, although the exact mechanisms are still unknown. We therefore investigated the gene expression profile of pancreatic islets from rats treated with dexamethasone using a cDNA array screening analysis. The expression of selected genes and proteins involved in mitochondrial apoptosis was further analyzed by PCR and immunoblotting. Insulin, triglyceride and free fatty acid plasma levels, as well as glucose-induced insulin secretion, were significantly higher in dexamethasone-treated rats compared with controls. Out of 1176 genes, 60 were up-regulated and 28 were down-regulated by dexamethasone treatment. Some of the modulated genes are involved in apoptosis, stress response, and proliferation pathways. RT-PCR confirmed the cDNA array results for 6 selected genes. Bax α protein expression was increased, while Bcl-2 was decreased. In vivo dexamethasone treatment decreased the mitochondrial production of NAD(P)H, and increased ROS production. Concluding, our data indicate that dexamethasone modulates the expression of genes and proteins involved in several pathways of pancreatic-islet cells, and mitochondria dysfunction might be involved in the deleterious effects after long-term GC treatment.


Subject(s)
Dexamethasone/pharmacology , Gene Expression Regulation/physiology , Islets of Langerhans/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Animals , Cells, Cultured , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Islets of Langerhans/drug effects , Mitochondria/drug effects , Rats , Rats, Wistar , Signal Transduction/drug effects
6.
J Appl Physiol (1985) ; 112(5): 711-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22174407

ABSTRACT

Endurance training has been shown to increase pancreatic ß-cell function and mass. However, whether exercise modulates ß-cell growth and survival pathways signaling is not completely understood. This study investigated the effects of exercise on growth and apoptotic markers levels in rat pancreatic islets. Male Wistar rats were randomly assigned to 8-wk endurance training or to a sedentary control group. After that, pancreatic islets were isolated; gene expression and the total content and phosphorylation of several proteins related to growth and apoptotic pathways as well as the main antioxidant enzymes were determined by real-time polymerase chain reaction and Western blot analysis, respectively. Reactive oxygen species (ROS) production was measured by fluorescence. Endurance training increased the time to reach fatigue by 50%. Endurance training resulted in increased protein phosphorylation content of AKT (75%), AKT substrate (AS160; 100%), mTOR (60%), p70s6k (90%), and ERK1/2 (50%), compared with islets from control group. Catalase protein content was 50% higher, whereas ROS production was 49 and 77% lower in islets from trained rats under basal and stimulating glucose conditions, respectively. Bcl-2 mRNA and protein levels increased by 46 and 100%, respectively. Bax and cleaved caspase-3 protein contents were reduced by 25 and 50% in islets from trained rats, respectively. In conclusion, these results demonstrate that endurance training favors the ß-cell growth and survival by activating AKT and ERK1/2 pathways, enhancing antioxidant capacity, and reducing ROS production and apoptotic proteins content.


Subject(s)
Insulin-Secreting Cells/physiology , Islets of Langerhans/physiology , Physical Endurance/physiology , Signal Transduction/physiology , Animals , Antioxidants/metabolism , Apoptosis/physiology , Body Weight , Fatigue/genetics , Fatigue/metabolism , Fatigue/physiopathology , Gene Expression , Glucose/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Male , Oxidation-Reduction , Phosphorylation , Physical Conditioning, Animal , Physical Endurance/genetics , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
7.
Biol Res ; 44(3): 251-7, 2011.
Article in English | MEDLINE | ID: mdl-22688912

ABSTRACT

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.


Subject(s)
Dexamethasone/adverse effects , Glucocorticoids/adverse effects , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance , Insulin/metabolism , Islets of Langerhans/drug effects , Animals , Insulin Secretion , Islets of Langerhans/metabolism , Male , Rats , Rats, Wistar , Shc Signaling Adaptor Proteins/metabolism , Signal Transduction
8.
Biol. Res ; 44(3): 251-257, 2011. ilus
Article in English | LILACS | ID: lil-608621

ABSTRACT

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.


Subject(s)
Animals , Male , Rats , Dexamethasone/adverse effects , Glucocorticoids/adverse effects , Insulin Resistance , Insulin Receptor Substrate Proteins/metabolism , Insulin/metabolism , Islets of Langerhans/drug effects , Insulin , Islets of Langerhans/metabolism , Rats, Wistar , Signal Transduction , Shc Signaling Adaptor Proteins/metabolism
9.
Endocrinology ; 151(1): 85-95, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19880808

ABSTRACT

Although glucocorticoids are widely used as antiinflammatory agents in clinical therapies, they may cause serious side effects that include insulin resistance and hyperinsulinemia. To study the potential functional adaptations of the islet of Langerhans to in vivo glucocorticoid treatment, adult Wistar rats received dexamethasone (DEX) for 5 consecutive days, whereas controls (CTL) received only saline. The analysis of insulin release in freshly isolated islets showed an enhanced secretion in response to glucose in DEX-treated rats. The study of Ca(2+) signals by fluorescence microscopy also demonstrated a higher response to glucose in islets from DEX-treated animals. However, no differences in Ca(2+) signals were found between both groups with tolbutamide or KCl, indicating that the alterations were probably related to metabolism. Thus, mitochondrial function was explored by monitoring oxidation of nicotinamide dinucleotide phosphate autofluorescence and mitochondrial membrane potential. Both parameters revealed a higher response to glucose in islets from DEX-treated rats. The mRNA and protein content of glucose transporter-2, glucokinase, and pyruvate kinase was similar in both groups, indicating that changes in these proteins were probably not involved in the increased mitochondrial function. Additionally, we explored the status of Ca(2+)-dependent signaling kinases. Unlike calmodulin kinase II, we found an augmented phosphorylation level of protein kinase C alpha as well as an increased response of the phospholipase C/inositol 1,4,5-triphosphate pathway in DEX-treated rats. Finally, an increased number of docked secretory granules were observed in the beta-cells of DEX animals using transmission electron microscopy. Thus, these results demonstrate that islets from glucocorticoid-treated rats develop several adaptations that lead to an enhanced stimulus-secretion coupling and secretory capacity.


Subject(s)
Glucocorticoids/pharmacology , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Adaptation, Biological/drug effects , Animals , Calcium/metabolism , Cell Separation , Cells, Cultured , Dexamethasone/pharmacology , Drug Resistance , Drug Synergism , Insulin Resistance , Insulin Secretion , Islets of Langerhans/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Rats , Rats, Wistar , Signal Transduction/drug effects , Up-Regulation/drug effects
10.
Free Radic Biol Med ; 47(10): 1386-93, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19698781

ABSTRACT

Pancreatic beta cells are very sensitive to reactive oxygen species (ROS) and this might play an important role in beta cell death in diabetes. Dexamethasone is a synthetic diabetogenic glucocorticoid, which impairs pancreatic beta cell function. Therefore we investigated the toxicity of dexamethasone in RINm5F insulin-producing cells and its dependence on the expression level of the antioxidant enzyme catalase, which inactivates hydrogen peroxide. This was correlated with oxidative stress and cell death. An increased generation of ROS was observed in dexamethasone-treated cells together with an increase in caspase-3 activity and apoptosis rate. Interestingly, exposure to dexamethasone increased the cytosolic superoxide dismutase Cu/ZnSOD protein expression and activity, whereas the mitochondrial MnSOD isoform was not affected by the glucocorticoid. Catalase overexpression in insulin-producing cells prevented all the cytotoxic effects of dexamethasone. In conclusion, dexamethasone-induced cell death in insulin-producing cells is ROS mediated. Increased levels of expression and activity of the Cu/ZnSOD might favor the generation of hydrogen peroxide in dexamethasone-treated cells. Increased ROS scavenging capacity in insulin-producing cells, through overexpression of catalase, prevents a deleterious increase in hydrogen peroxide generation and thus prevents dexamethasone-induced apoptosis.


Subject(s)
Antioxidants/metabolism , Catalase/metabolism , Dexamethasone/toxicity , Insulin-Secreting Cells/drug effects , Animals , Caspase 3/metabolism , Catalase/biosynthesis , Catalase/genetics , Cell Death , Cells, Cultured , Gene Expression , Humans , Hydrogen Peroxide/metabolism , Insulin-Secreting Cells/metabolism , Oxidative Stress , Rats , Reactive Oxygen Species/metabolism
11.
Am J Physiol Endocrinol Metab ; 296(4): E681-9, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19158320

ABSTRACT

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.


Subject(s)
Cell Proliferation/drug effects , Dexamethasone/pharmacology , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Animals , Cell Cycle/drug effects , Cell Death/drug effects , Dexamethasone/administration & dosage , Dose-Response Relationship, Drug , Insulin Resistance , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Islets of Langerhans/anatomy & histology , Islets of Langerhans/pathology , Islets of Langerhans/physiology , Male , Metabolic Syndrome/chemically induced , Metabolic Syndrome/pathology , Organ Size , Rats , Rats, Wistar
12.
Regul Pept ; 148(1-3): 39-45, 2008 Jun 05.
Article in English | MEDLINE | ID: mdl-18378016

ABSTRACT

Islet Neogenesis Associated Protein (INGAP) increases pancreatic beta-cell mass and potentiates glucose-induced insulin secretion. Here, we investigated the effects of the pentadecapeptide INGAP-PP in adult cultured rat islets upon the expression of proteins constitutive of the K(+)(ATP) channel, Ca(2+) handling, and insulin secretion. The islets were cultured in RPMI medium with or without INGAP-PP for four days. Thereafter, gene (RT-PCR) and protein expression (Western blotting) of Foxa2, SUR1 and Kir6.2, cytoplasmic Ca(2+) ([Ca(2+)](i)), static and dynamic insulin secretion, and (86)Rb efflux were measured. INGAP-PP increased the expression levels of Kir6.2, SUR1 and Foxa2 genes, and SUR1 and Foxa2 proteins. INGAP-PP cultured islets released significantly more insulin in response to 40 mM KCl and 100 muM tolbutamide. INGAP-PP shifted to the left the dose-response curve of insulin secretion to increasing concentrations of glucose (EC(50) of 10.0+/-0.4 vs. 13.7+/-1.5 mM glucose of the controls). It also increased the first phase of insulin secretion elicited by either 22.2 mM glucose or 100 microM tolbutamide and accelerated the velocity of glucose-induced reduction of (86)Rb efflux in perifused islets. These effects were accompanied by a significant increase in [Ca(2+)](i) and the maintenance of a considerable degree of [Ca(2+)](i) oscillations. These results confirm that the enhancing effect of INGAP-PP upon insulin release, elicited by different secretagogues, is due to an improvement of the secretory function in cultured islets. Such improvement is due, at least partly, to an increased K(+)(ATP) channel protein expression and/or changing in the kinetic properties of these channels and augmented [Ca(2+)](i) response. Accordingly, INGAP-PP could potentially be used to maintain the functional integrity of cultured islets and eventually, for the prevention and treatment of diabetes.


Subject(s)
Calcium/metabolism , Cytokines/pharmacology , Islets of Langerhans/drug effects , KATP Channels/metabolism , Peptide Fragments/pharmacology , Animals , Blotting, Western , Gene Expression/drug effects , Glucose/pharmacology , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , KATP Channels/genetics , Pancreatitis-Associated Proteins , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Potassium Chloride/pharmacology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Culture Techniques , Tolbutamide/pharmacology
13.
Metabolism ; 57(5): 617-24, 2008 May.
Article in English | MEDLINE | ID: mdl-18442623

ABSTRACT

The effects of dexamethasone (Dex) on the metabolic parameters, peripheral insulin, and glucose sensitivity in vivo as well as on islet function ex vivo of rats submitted to low-protein diet were analyzed. Dexamethasone (1.0 mg/kg body weight) was administered intraperitoneally daily to adult Wistar rats fed on a normal-protein diet or low-protein diet (LPD) for 5 days, whereas control rats fed on a normal-protein diet or low-protein diet (LP) received saline alone. At the end of the experimental period, LP rats showed a significant reduction in serum insulin, total serum protein, and serum albumin levels compared with rats fed on a normal-protein diet (P<.05). All these parameters tended to be normalized in LPD rats (P<.05); furthermore, these rats exhibited increased serum glucose and nonesterified fatty acid levels compared with LP rats (P<.05). Rats submitted to the low-protein diet demonstrated normal peripheral glucose sensitivity and improved peripheral insulin sensitivity, which was reversed by Dex treatment. A reduced area of islets from LP rats was partially recovered in LPD rats (P<.05). At 16.7 mmol/L glucose, insulin secretion from LPD islets was also partially recovered and was significantly higher than that from LP islets (P<.05). In conclusion, induction of insulin resistance by Dex treatment reverses most of the metabolic alterations in rats submitted to a low-protein diet. In addition, several islet functions were also improved by Dex, confirming the plasticity of pancreatic islets in adverse conditions.


Subject(s)
Dexamethasone/pharmacology , Islets of Langerhans/metabolism , Protein Deficiency/metabolism , Animals , Blood Glucose/analysis , Fatty Acids, Nonesterified/blood , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Islets of Langerhans/pathology , Male , Protein Deficiency/pathology , Rats , Rats, Wistar
14.
Pancreas ; 36(3): 284-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18362843

ABSTRACT

OBJECTIVES: We have analyzed the peripheral insulin and glucose sensitivity in vivo, and islet function ex vivo in rats with different degrees of insulin resistance induced by dexamethasone (DEX). METHODS: Dexamethasone, in the concentrations of 0.1 (DEX 0.1), 0.5 (DEX 0.5), and 1.0 mg/kg body weight (DEX 1.0) was administered daily, intraperitoneally, to adult Wistar rats for 5 days, whereas controls received saline. RESULTS: Dexamethasone treatment induced peripheral insulin resistance in a dose-dependent manner. At the end of the treatment, only DEX 1.0 rats showed significant increase of postabsorptive blood glucose and serum triglycerides, and nonesterified fatty acids levels. Incubation of pancreatic islets in increasing glucose concentrations (2.8-22 mM) led to an augmented insulin secretion in all DEX-treated rats. Leucine, carbachol, and high KCl concentrations induced the insulin release in DEX 0.5 and DEX 1.0, whereas arginine augmented secretion in all DEX-treated groups. CONCLUSIONS: We demonstrate that in DEX 0.5 and, especially in DEX 0.1 groups, but not in DEX 1.0, the adaptations that occurred in the endocrine pancreas are able to counteract metabolic disorders (glucose intolerance and dyslipidemia). These animal models seem to be interesting approaches for the study of degrees of subjacent effects that may mediate type 2 diabetes (DEX 1.0) and islet function alterations, without collateral effects (DEX 0.1 and DEX 0.5).


Subject(s)
Dexamethasone/toxicity , Glucocorticoids/toxicity , Insulin Resistance/physiology , Islets of Langerhans/drug effects , Islets of Langerhans/physiopathology , Animals , Blood Glucose/metabolism , Dexamethasone/administration & dosage , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Fatty Acids, Nonesterified/blood , Glucocorticoids/administration & dosage , Glucose/pharmacology , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Male , Rats , Rats, Wistar , Triglycerides/blood
15.
J Endocrinol ; 195(1): 17-27, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17911393

ABSTRACT

During pregnancy, the maternal endocrine pancreas undergoes, as a consequence of placental lactogens and prolactin (PRL) action, functional changes that are characterized by increased glucose-induced insulin secretion. After delivery, the maternal endocrine pancreas rapidly returns to non-pregnant state, which is mainly attributed to the increased serum levels of glucocorticoids (GCs). Although GCs are known to decrease insulin secretion and counteract PRL action, the mechanisms for these effects are poorly understood. We have previously demonstrated that signal transducer and activator of transcription 3 (STAT3) is increased in islets treated with PRL. In the present study, we show that STAT3 expression and serine phosphorylation are increased in pancreatic islets at the end of pregnancy (P19). STAT3 serine phosphorylation rapidly returned to basal levels 3 days after delivery (L3). The expression of the sarcoendoplasmic reticulum Ca(2+)-ATPase 2 (SERCA2), a crucial protein involved in the regulation of calcium handling in beta-cells, was also increased in P19, returning to basal levels at L3. PRL increased SERCA2 and STAT3 expressions and STAT3 serine phosphorylation in RINm5F cells. The upregulation of SERCA2 by PRL was abolished after STAT3 knockdown. Moreover, PRL-induced STAT3 serine phosphorylation and SERCA2 expression were inhibited by dexamethasone (DEX). Insulin secretion from islets of P19 rats pre-incubated with thapsigargin and L3 rats showed a dramatic suppression of first phase of insulin release. The present results indicate that PRL regulates SERCA2 expression by a STAT3-dependent mechanism. PRL effect is counteracted by DEX and might contribute to the adaptation of maternal endocrine pancreas during the peripartum period.


Subject(s)
Glucocorticoids/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Prolactin/metabolism , STAT3 Transcription Factor/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Adaptation, Physiological , Animals , Blotting, Western , Cell Line , Cells, Cultured , Dexamethasone/pharmacology , Female , Gene Expression/drug effects , Insulin/analysis , Insulin Secretion , Islets of Langerhans/chemistry , Lactation/physiology , Oligonucleotides, Antisense/genetics , Phosphorylation , Pregnancy , Prolactin/genetics , RNA, Messenger/analysis , Rats , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/analysis , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Signal Transduction/drug effects , Transfection/methods
16.
Arq. bras. endocrinol. metab ; 42(1): 29-35, fev. 1998. ilus, graf
Article in Portuguese | LILACS | ID: lil-214539

ABSTRACT

Glicose provoca a secreçao de insulina através do aumento da relaçao ATP/ADP no citoplasma das células beta. Isto leva ao bloqueio de canais de K+ sensíveis ao ATP (KATP), reduçao da saída deste cátion da célula, despolarizaçao celular, ativaçao da permeabilidade ao Ca2+ sensível à voltagem, entrada e acúmulo deste cátion nas células e consequente secreçao de insulina. O canal KATP parece ser composto por duas unidades distintas; uma delas, denominada Kir6,2, constitui o canal propriamente dito, por onde fluem as correntes de K+. A outra é o receptor de sulfoniluréias (SUR1), que é provida de sítios de ligaçao para o referido fármaco, para ATP, MgADP e diazoxida, atuando como unidade regulatória. Neste artigo, fazemos uma breve revisao da fisiologia dos canais KATP, considerando também sua importância na fisiopatologia do processo secretório.


Subject(s)
Humans , Adenosine Triphosphate/pharmacology , Potassium Channels , Glucose/pharmacology , Hyperinsulinism/genetics , Hyperinsulinism/physiopathology , Insulin/metabolism , Potassium Channels/deficiency , Potassium Channels/physiology , Hypoglycemic Agents/pharmacology , Sulfonylurea Compounds/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...