Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
J Clin Endocrinol Metab ; 106(4): e1574-e1585, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33421070

ABSTRACT

CONTEXT: Congenital adrenal hyperplasia (CAH) patients have potential normal longevity. However, a greater risk for cardiovascular disease has been reported. Insulin resistance and hyperinsulinemia have been described in CAH patients, whereas the prevalence of overt type 2 diabetes is not higher in CAH than in normal population. OBJECTIVE: To examine the contributions of insulin secretion and of hepatic insulin clearance to compensatory hyperinsulinemia in young insulin-resistant adults with classic CAH due to 21-hydroxylase deficiency (21-OHD). DESIGN: Cross-sectional. SETTING: University outpatient clinics. METHODS: Fifty-one participants: 21 controls, and 30 CAH (15 virilizing and 15 salt-wasting phenotypes), female/male (33/18), age (mean [SD]): 24.0 (3.6) years, body mass index: 24.6 (4.9)kg/m2 with normal glucose tolerance, were submitted to a hyperglycemic clamp study. MAIN OUTCOME MEASURES: Insulin sensitivity, beta cell function, and hepatic insulin clearance using appropriate modeling. RESULTS: We found an increased insulin resistance in 21-OHD. The systemic hyperinsulinemia (posthepatic insulin delivery) was elevated in CAH patients. No increases were observed in insulin secretory rate (beta cell function) in the first phase or during the hyperglycemic clamp. The increase in insulin concentrations was totally due to a ~33% reduction in insulin clearance. CONCLUSION: 21-OHD nonobese subjects have reduced insulin sensitivity and beta cell response unable to compensate for the insulin resistance, probably due to overexposure to glucocorticoids. Compensatory hyperinsulinemia is most related with reduced hepatic insulin clearance. The exclusive adaptation of the liver acts as a gating mechanism to regulate the access of insulin to insulin-sensitive tissues to maintain glucose homeostasis.


Subject(s)
Adrenal Hyperplasia, Congenital/metabolism , Hyperinsulinism/metabolism , Insulin Resistance , Insulin/metabolism , Adrenal Hyperplasia, Congenital/complications , Adult , Cross-Sectional Studies , Female , Humans , Hyperinsulinism/complications , Insulin-Secreting Cells/metabolism , Male , Young Adult
2.
Front Endocrinol (Lausanne) ; 11: 599165, 2020.
Article in English | MEDLINE | ID: mdl-33324349

ABSTRACT

ARHGAP21 is a RhoGAP protein implicated in the modulation of insulin secretion and energy metabolism. ARHGAP21 transient-inhibition increase glucose-stimulated insulin secretion (GSIS) in neonatal islets; however, ARHGAP21 heterozygote mice have a reduced insulin secretion. These discrepancies are not totally understood, and it might be related to functional maturation of beta cells and peripheral sensitivity. Here, we investigated the real ARHGAP21 role in the insulin secretion process using an adult mouse model of acute ARHGAP21 inhibition, induced by antisense. After ARHGAP21 knockdown induction by antisense injection in 60-day old male mice, we investigated glucose and insulin tolerance test, glucose-induced insulin secretion, glucose-induced intracellular calcium dynamics, and gene expression. Our results showed that ARHGAP21 acts negatively in the GSIS of adult islet. This effect seems to be due to the modulation of important points of insulin secretion process, such as the energy metabolism (PGC1α), Ca2+ signalization (SYTVII), granule-extrusion (SNAP25), and cell-cell interaction (CX36). Therefore, based on these finds, ARHGAP21 may be an important target in Diabetes Mellitus (DM) treatment.


Subject(s)
GTPase-Activating Proteins/physiology , Gene Expression Regulation/drug effects , Glucose/pharmacology , Hyperinsulinism/prevention & control , Insulin Secretion , Insulin-Secreting Cells/drug effects , Animals , Homeostasis , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sweetening Agents/pharmacology
3.
Molecules ; 25(14)2020 Jul 20.
Article in English | MEDLINE | ID: mdl-32698439

ABSTRACT

Dietary intake of eicosapentaenoic/docosahexaenoic acid (EPA/DHA) reduces insulin resistance and hepatic manifestations through the regulation of metabolism in the liver. Obese mice present insulin resistance and lipid accumulation in intracellular lipid droplets (LDs). LD-associated proteins perilipin (Plin) have an essential role in both adipogenesis and lipolysis; Plin5 regulates lipolysis and thus contributes to fat oxidation. The purpose of this study was to compare the effects of deodorized refined salmon oil (DSO) and its polyunsaturated fatty acids concentrate (CPUFA) containing EPA and DHA, obtained by complexing with urea, on obesity-induced metabolic alteration. CPUFA maximum content was determined using the Box-Behnken experimental design based on Surface Response Methodology. The optimized CPUFA was administered to high-fat diet (HFD)-fed mice (200 mg/kg/day of EPA + DHA) for 8 weeks. No significant differences (p > 0.05) in cholesterol, glycemia, LDs or transaminase content were found. Fasting insulin and hepatic Plin5 protein level increased in the group supplemented with the EPA + DHA optimized product (38.35 g/100 g total fatty acids) compared to obese mice without fish oil supplementation. The results suggest that processing salmon oil by urea concentration can generate an EPA+DHA dose useful to prevent the increase of fasting insulin and the decrease of Plin5 in the liver of insulin-resistant mice.


Subject(s)
Diet, High-Fat , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Feeding Behavior , Hyperinsulinism/metabolism , Liver/metabolism , Perilipin-5/metabolism , Urea/chemistry , Analysis of Variance , Animals , Body Weight/drug effects , Fish Oils/pharmacology , Lipid Droplets/chemistry , Liver/drug effects , Male , Mice, Inbred C57BL , Oxidation-Reduction
4.
Gac. méd. Méx ; Gac. méd. Méx;155(5): 500-503, Sep.-Oct. 2019. graf
Article in English | LILACS | ID: biblio-1286550

ABSTRACT

The metabolic syndrome describes a group of signs that increase the likelihood for developing type 2 diabetes mellitus, cardiovascular diseases and some types of cancer. The action of insulin depends on its binding to membrane receptors on its target cells. We wonder if blood insulin could travel bound to proteins and if, in the presence of hyperinsulinemia, a soluble insulin receptor might be generated. We used young adult Wistar rats (which have no predisposition to obesity or diabetes), whose drinking water was added 20 % of sugar and that were fed a standard diet ad libitum for two and six months. They were compared with control rats under the same conditions, but that had running water for consumption. At two months, the rats developed central obesity, moderate hypertension, high triglyceride levels, hyperinsulinemia, glucose intolerance and insulin resistance, i.e., metabolic syndrome. Electrophoresis of the rats’ plasma proteins was performed, followed by Western Blot (WB) for insulin and for the outer portion of the insulin receptor. The bands corresponding to insulin and to the receptor external part were at the same molecular weight level, 25-fold higher than that of free insulin. We demonstrated that insulin, both in control animals and in those with hyperinsulinemia, travels bound to the receptor outer portion (ectodomain), which we called soluble insulin receptor, and that is released al higher amounts in response to plasma insulin increase; in rats with metabolic syndrome and hyperinsulinemia, plasma levels are much higher than in controls. Soluble insulin receptor increase in blood might be an early sign of metabolic syndrome.


Subject(s)
Humans , Animals , Rats , Insulin Resistance/physiology , Receptor, Insulin/metabolism , Metabolic Syndrome/etiology , Hyperinsulinism/metabolism , Insulin/metabolism , Hypertriglyceridemia/etiology , Rats, Wistar , Glucose Intolerance/etiology , Metabolic Syndrome/metabolism , Diabetes Mellitus, Type 2/etiology , Disease Models, Animal , Obesity, Abdominal/etiology , Hypertension/etiology , Insulin/blood
5.
Nutrients ; 11(7)2019 Jul 05.
Article in English | MEDLINE | ID: mdl-31284400

ABSTRACT

This study evaluates the progressive participation of enzymes involved in lipolysis and lipogenesis, leading to adipocyte hypertrophy in a metabolic syndrome (MS) rat model caused by chronic consumption of 30% sucrose in drinking water. A total of 70 male Wistar rats were divided into two groups: C and MS. Each of these groups were then subdivided into five groups which were sacrificed as paired groups every month from the beginning of the treatment until 5 months. The intra-abdominal fat was dissected, and the adipocytes were extracted. Lipoprotein lipase (LPL), hormone-sensitive lipase (HSL), protein kinases A (PKA), and perilipin A expressions were determined. The LPL and HSL activities were evaluated by spectrophotometry. Histological staining was performed in adipose tissue. Significant increases were observed in blood pressure, HOMA-IR, leptin, triglycerides, insulin, intra-abdominal fat, and number of fat cells per field (p = 0.001) and in advanced glycosylation products, adipocyte area, LPL, HSL activities and/or expression (p ≤ 0.01) in the MS groups progressively from the third month onward. Lipogenesis and lipolysis were increased by LPL activity and HSL activity and/or expression. This was associated with hyperinsulinemia and release of non-esterified fatty acids causing a positive feedback loop that contributes to the development of adipocyte hypertrophy.


Subject(s)
Abdominal Fat/metabolism , Lipogenesis , Lipolysis , Metabolic Syndrome/metabolism , Abdominal Fat/pathology , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Dietary Sucrose , Disease Models, Animal , Disease Progression , Fatty Acids, Nonesterified/metabolism , Feedback, Physiological , Hyperinsulinism/etiology , Hyperinsulinism/metabolism , Hypertrophy , Lipoprotein Lipase/metabolism , Male , Metabolic Syndrome/etiology , Metabolic Syndrome/pathology , Perilipin-1/metabolism , Rats, Wistar , Signal Transduction , Sterol Esterase/metabolism , Time Factors
6.
J Steroid Biochem Mol Biol ; 190: 54-63, 2019 06.
Article in English | MEDLINE | ID: mdl-30923014

ABSTRACT

Oral contraception is the most commonly used interventional method in the world. However, several women employ the continuous use of these hormones to avoid pre- and menstruation discomforts. Some studies indicate that oral contraceptives are associated with disturbances in glycemia and the effects of the use of a continuous regime are poorly elucidated. Herein, we evaluated the effects of the continuous administration of a combined oral contraceptive (COC) composed by ethinyl estradiol (EE) and drospirenone (DRSP) on glucose homeostasis in female mice. Adult Swiss mice received 0.6 µg EE and 60 µg DRSP (COC group) or vehicle [control (CTL)] daily by gavage for 35 days. COC treatment had no effect on body weight or adiposity, but increased uterus weight and induced hepatomegaly. Importantly, COC females displayed normal glycemia and glucose tolerance, but hyperinsulinemia and lower plasma C-peptide/insulin ratio, indicating reduced insulin clearance. Furthermore, COC mice displayed reduced protein content of the ß subunit of the insulin receptor (IRß) in the liver. Additionally, pancreatic islets isolated from COC mice secreted more insulin in response to increasing glucose concentrations. This effect was associated with the activity of steroid hormones, since INS-1E cells incubated with EE plus DRSP also secreted more insulin. Therefore, we provide the first evidence that the continuous administration of EE and DRSP lead to hyperinsulinemia, due to enhancement of insulin secretion and the reduction of insulin degradation, which possibly lead to the down-regulation of hepatic IRß. These findings suggest that the continuous administration of COC could cause insulin resistance with the prolongation of treatment.


Subject(s)
Androstenes/adverse effects , Contraceptives, Oral, Combined/adverse effects , Ethinyl Estradiol/adverse effects , Hyperinsulinism/chemically induced , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Animals , Female , Glucose/metabolism , Hyperinsulinism/metabolism , Insulin Resistance , Insulin-Secreting Cells/metabolism , Mice
7.
Diabetes Metab Syndr ; 13(1): 258-263, 2019.
Article in English | MEDLINE | ID: mdl-30641708

ABSTRACT

AIM: To evaluate the association between vitamin D deficiency and insulin resistance (IR) or hyperinsulinemia after oral glucose tolerance test (OGTT) in euthyroid non-diabetic individuals. MATERIALS AND METHODS: We carried out an analytical cross-sectional study in euthyroid non-diabetic adults of both sexes, who attended the outpatient service of a private clinic in Lima-Peru during the 2012-2016 period. Participants were categorized in two groups according to their serum vitamin D levels: normal vitamin D levels (serum vitamin D values ≥ 20 ng/dL) and vitamin D deficiency (serum vitamin D values < 20 ng/dL). IR was defined as a Homeostasis Model Assessment (HOMA-IR) value ≥ 3.8 and hyperinsulinemia after OGTT was defined as a serum insulin value ≥ 80µU/mL after 120 min of 75-g glucose intake. We elaborated crude and adjusted Poisson regression models to assess the association between serum vitamin D levels and IR or hyperinsulinemia after OGTT. The reported association measure was the prevalence ratio (PR) with their respective 95% confidence intervals (95%CI). RESULTS: We analyzed 204 participants, the average age was 38.5 ±â€¯10.6 (SD) years, 40 (19.6%) were males and the vitamin D median was 25.0 (IQR: 19.0-33.3) ng/dL. The prevalence of vitamin D deficiency, IR and hyperinsulinemia after OGTT was 29.4% (n = 60), 29.9% (n = 61) and 25.0% (n = 51). In the adjusted Poisson regression models, the prevalence of hyperinsulinemia after OGTT was higher among the vitamin D deficient group (aPR=1.75; 95%CI: 1.06-2.90); however, we did not find statistically significant association between vitamin D deficiency and IR (aPR=0.99; 95%CI: 0.61-1.63). CONCLUSIONS: We found an association between vitamin D deficiency and hyperinsulinemia after OGTT in euthyroid people with no T2DM.Our findings are consistent with previous reports; providing evidence that serum vitamin D deficiency could be an IR marker.


Subject(s)
Biomarkers/metabolism , Hyperinsulinism/epidemiology , Insulin Resistance , Thyroid Gland/physiopathology , Vitamin D Deficiency/epidemiology , Adult , Cross-Sectional Studies , Female , Follow-Up Studies , Humans , Hyperinsulinism/diagnosis , Hyperinsulinism/metabolism , Male , Peru/epidemiology , Prevalence , Prognosis , Vitamin D Deficiency/diagnosis , Vitamin D Deficiency/metabolism
8.
Gac Med Mex ; 155(5): 500-503, 2019.
Article in English | MEDLINE | ID: mdl-32091018

ABSTRACT

The metabolic syndrome describes a group of signs that increase the likelihood for developing type 2 diabetes mellitus, cardiovascular diseases and some types of cancer. The action of insulin depends on its binding to membrane receptors on its target cells. We wonder if blood insulin could travel bound to proteins and if, in the presence of hyperinsulinemia, a soluble insulin receptor might be generated. We used young adult Wistar rats (which have no predisposition to obesity or diabetes), whose drinking water was added 20 % of sugar and that were fed a standard diet ad libitum for two and six months. They were compared with control rats under the same conditions, but that had running water for consumption. At two months, the rats developed central obesity, moderate hypertension, high triglyceride levels, hyperinsulinemia, glucose intolerance and insulin resistance, i.e., metabolic syndrome. Electrophoresis of the rats' plasma proteins was performed, followed by Western Blot (WB) for insulin and for the outer portion of the insulin receptor. The bands corresponding to insulin and to the receptor external part were at the same molecular weight level, 25-fold higher than that of free insulin. We demonstrated that insulin, both in control animals and in those with hyperinsulinemia, travels bound to the receptor outer portion (ectodomain), which we called soluble insulin receptor, and that is released al higher amounts in response to plasma insulin increase; in rats with metabolic syndrome and hyperinsulinemia, plasma levels are much higher than in controls. Soluble insulin receptor increase in blood might be an early sign of metabolic syndrome.


Subject(s)
Hyperinsulinism/metabolism , Insulin Resistance/physiology , Insulin/metabolism , Metabolic Syndrome/etiology , Receptor, Insulin/metabolism , Animals , Diabetes Mellitus, Type 2/etiology , Disease Models, Animal , Glucose Intolerance/etiology , Humans , Hypertension/etiology , Hypertriglyceridemia/etiology , Insulin/blood , Metabolic Syndrome/metabolism , Obesity, Abdominal/etiology , Rats , Rats, Wistar
9.
J Cell Physiol ; 234(6): 9802-9809, 2019 06.
Article in English | MEDLINE | ID: mdl-30370604

ABSTRACT

Human life expectancy is increasing faster lately and, consequently, the number of patients with age-related diseases such as type 2 diabetes (T2D) is rising every year. Cases of hyperinsulinemia have been extensively reported in elderly subjects and this alteration in blood insulin concentration is postulated to be a cause of insulin resistance, which in some cases triggers T2D onset. Thus, it is important to know the underlying mechanisms of age-dependent hyperinsulinemia to find new strategies to prevent T2D in elderly subjects. Two processes control blood insulin concentration: Insulin secretion by the endocrine portion of the pancreas and insulin clearance, which occurs mainly in the liver by the action of the insulin-degrading enzyme (IDE). Here, we demonstrated that 10-month-old mice (old) display increased body and fat pad weight, compared with 3-month-old mice (control), and these alterations were accompanied by glucose and insulin intolerance. We also confirm hyperinsulinemia in the old mice, which was related to increased insulin secretion but not to reduced insulin clearance. Although no changes in insulin clearance were observed, IDE activity was lower in the liver of old compared with the control mice. However, this decreased IDE activity was compensated by increased expression of IDE protein in the liver, thus explaining the similar insulin clearance observed in both groups. In conclusion, at the beginning of aging, 10-month-old mice do not display any alterations in insulin clearance. Therefore, hyperinsulinemia is initiated primarily due to a higher insulin secretion in the age-related metabolic dysfunction in mice.


Subject(s)
Aging , Glucose/metabolism , Hyperinsulinism/etiology , Insulin/metabolism , Animals , Area Under Curve , Blood Glucose , Body Weight , Glucose/pharmacology , Homeostasis , Hyperinsulinism/metabolism , Insulin/blood , Insulysin , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL
10.
Cell Biol Int ; 43(4): 373-383, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30353973

ABSTRACT

Recent studies have been trying to find out how diet and metabolic changes such as dyslipidaemia, hyperglycaemia, and hyperinsulinaemia can stimulate cancer progression. This investigation aimed to evaluate the effect of high concentrations of fatty acids and/or glucose in tumour prostate cells, focusing on the proliferation/migration profile and oxidative stress. PC3 cells were treated with high concentration of saturated fatty acid (palmitate, 100 µM), glucose (220 mg/dL), or both for 24 or 48 h. Results demonstrated that PC3 cells showed a significant increase in proliferation after 48 h of treatment with glucose and palmitate+glucose. Cell proliferation was associated with reduced levels of AMPK phosphorylation in glucose group at 24 and 48 h of treatment, while palmitate group presented this result only after 48 h of treatment. Also, there was a significant increase in cell migration between time 0 and 48 h after all treatments, except in the control. Catalase activity was increased by palmitate in the beginning of treatment, while glucose presented a later effect. Also, nitrite production was increased by glucose only after 48 h, and the total antioxidant activity was enhanced by palmitate in the initial hours. Thus, we conclude that the high concentration of the saturated fatty acid palmitate and glucose in vitro influences PC3 cells and stimulates cellular activities related to carcinogenesis such as cell proliferation, migration, and oxidative stress in different ways. Palmitate presents a rapid and initial effect, while a glucose environment stimulates cells later on, maintaining high levels of cell proliferation.


Subject(s)
Glucose/metabolism , Palmitates/metabolism , Prostatic Neoplasms/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Fatty Acids/metabolism , Glucose/adverse effects , Glucose/physiology , Humans , Hyperinsulinism/metabolism , Insulin/metabolism , Male , PC-3 Cells/drug effects , Palmitates/pharmacology , Phosphorylation , Prostate/metabolism
11.
Neuropeptides ; 63: 18-27, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28427866

ABSTRACT

Aggregation of the amyloid beta (Aß) peptide and hyperphosphorylation of tau protein, which are markers of Alzheimer's disease (AD), have been reported also in diabetes mellitus (DM). One regulator of tau phosphorylation is O-GlcNAcylation, whereas for hyperphosphorylation it could be GSK3beta, which is activated in hyperglycemic conditions. With this in mind, both O-GlcNAcylation and phosphorylation of tau protein were evaluated in the brain of rats with streptozotocin (STZ)-induced hyperglycemia and hyperinsulinemia and treated with the Aß25-35 peptide in the hippocampal region CA1. Weight, glycated hemoglobin, glucose, and insulin were determined. Male Wistar rats were divided in groups (N=20): a) control, b) treated only with the Aß25-35 peptide, c) treated with Aß25-35 and STZ, and d) treated only with STZ. Results showed statistically significant differences in the mean weight, glucose levels, insulin concentration, and HbA1c percentage, between C- and D-treated groups and not STZ-treated A and B (P<0.05). Interestingly, our results showed diminution of O-GlcNAcylation and increase in P-tau-Ser-396 in the hippocampal area of the Aß25-35- and STZ-treated groups; moreover, enhanced expression of GSK3beta was observed in this last group. Our results suggest that hyperinsulinemia-Aß25-35-hyperglycemia is relevant for the down regulation of O-GlcNAcylation and up-regulation of the glycogen synthase kinase-3 beta (GSK3beta), favoring Aß25-35-induced neurotoxicity in the brain of rats.


Subject(s)
Amyloid beta-Peptides/pharmacology , CA1 Region, Hippocampal/drug effects , Diabetes Mellitus, Experimental/metabolism , Hyperglycemia/metabolism , Hyperinsulinism/metabolism , Peptide Fragments/pharmacology , tau Proteins/metabolism , Acylation/drug effects , Animals , Blood Glucose , CA1 Region, Hippocampal/metabolism , Insulin/blood , Male , Phosphorylation/drug effects , Rats , Rats, Wistar
12.
Eur J Nutr ; 56(2): 705-713, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26621632

ABSTRACT

PURPOSE: Obesity is usually associated with low-grade inflammation, which impairs insulin action. The amino acid, taurine (TAU), regulates glucose homeostasis and lipid metabolism and presents anti-inflammatory actions. Here, we evaluated whether inflammatory markers are altered in the serum and retroperitoneal adipose tissue of monosodium glutamate (MSG) obese rats, supplemented or not with TAU. METHODS: Male Wistar rats received subcutaneous injections of MSG (4 mg/kg body weight/day, MSG group) or hypertonic saline (CTL) during the first 5 days of life. From 21 to 120 days of age, half of each of the MSG and CTL groups received 2.5 % TAU in their drinking water (CTAU and MTAU). RESULTS: At 120 days of age, MSG rats were obese and hyperinsulinemic. TAU supplementation reduced fat deposition without affecting insulinemia in MTAU rats. MSG rats presented increased pIκ-Bα/Iκ-Bα protein expression in the retroperitoneal adipose tissue. TAU supplementation decreased the ratio of pIκ-Bα/Iκ-Bα protein, possibly contributing to the increased Iκ-Bα content in MTAU adipose tissue. Furthermore, MSG obesity or supplementation did not alter TNF-α, IL-1ß or IL-6 content in adipose tissue. In contrast, MSG rats presented lower serum TNF-α, IL-4 and IL-10 concentrations, and these alterations were prevented by TAU treatment. CONCLUSION: MSG obesity in rats was not associated with alterations in pro-inflammatory markers in retroperitoneal fat stores; however, reductions in the serum concentrations of anti-inflammatory cytokines and of TNF-α were observed. TAU treatment decreased adiposity, and this effect was associated with the normalization of circulating TNF-α and IL-4 concentrations in MTAU rats.


Subject(s)
Anti-Obesity Agents/therapeutic use , Dietary Supplements , Gene Expression Regulation , Intra-Abdominal Fat/metabolism , NF-KappaB Inhibitor alpha/metabolism , Obesity/diet therapy , Taurine/therapeutic use , Adiposity , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/blood , Biomarkers/metabolism , Hyperinsulinism/diet therapy , Hyperinsulinism/etiology , Hyperinsulinism/immunology , Hyperinsulinism/metabolism , I-kappa B Proteins/agonists , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Injections, Subcutaneous , Interleukin-4/antagonists & inhibitors , Interleukin-4/blood , Interleukin-4/metabolism , Intra-Abdominal Fat/immunology , Male , NF-KappaB Inhibitor alpha/agonists , NF-KappaB Inhibitor alpha/genetics , Obesity/etiology , Obesity/immunology , Obesity/metabolism , Phosphorylation , Protein Processing, Post-Translational , Rats, Wistar , Sodium Glutamate/administration & dosage , Sodium Glutamate/adverse effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism
13.
J Physiol Biochem ; 72(4): 625-633, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27351887

ABSTRACT

Herein, we investigated whether subdiaphragmatic vagotomy has benefits on obesity, body glucose homeostasis, and insulin secretion in cafeteria (CAF)-obese rats. Wistar rats were fed a standard or CAF diet for 12 weeks. Subsequently, CAF rats were randomly submitted to truncal vagotomy (CAF Vag) or sham operation (CAF Sham). CAF Sham rats were hyperphagic, obese, and presented metabolic disturbances, including hyperinsulinemia, glucose intolerance, insulin resistance, hyperglycemia, and hypertriglyceridemia. Twelve weeks after vagotomy, CAF Vag rats presented reductions in body weight and perigonadal fat stores. Vagotomy did not modify glucose tolerance but normalized fed glycemia, insulinemia, and insulin sensitivity. Isolated islets from CAF Sham rats secreted more insulin in response to the cholinergic agent, carbachol, and when intracellular cyclic adenine monophosphate (cAMP) is enhanced by forskolin or 3-isobutyl-1-methylxanthine. Vagotomy decreased glucose-induced insulin release due to a reduction in the cholinergic action on ß-cells. This effect also normalized islet secretion in response to cAMP. Therefore, vagotomy in rats fed on a CAF-style diet effectively decreases adiposity and restores insulin sensitivity. These effects were mainly associated with the lack of cholinergic action on the endocrine pancreas, which decreases insulinemia and may gradually reduce fat storage and improve insulin sensitivity.


Subject(s)
Hyperglycemia/surgery , Hyperinsulinism/surgery , Hypertriglyceridemia/surgery , Obesity/surgery , Vagotomy , Vagus Nerve/surgery , 1-Methyl-3-isobutylxanthine/pharmacology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Body Weight/drug effects , Carbachol/pharmacology , Colforsin/pharmacology , Cyclic AMP/metabolism , Diet, High-Fat , Disease Models, Animal , Glucose/metabolism , Glucose/pharmacology , Hyperglycemia/etiology , Hyperglycemia/metabolism , Hyperglycemia/pathology , Hyperinsulinism/etiology , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Hypertriglyceridemia/etiology , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/pathology , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Rats , Rats, Wistar , Tissue Culture Techniques , Vagus Nerve/metabolism
14.
J Endocrinol ; 230(1): 157-69, 2016 07.
Article in English | MEDLINE | ID: mdl-27154336

ABSTRACT

The metabolic syndrome is a growing epidemic; it increases the risk for diabetes, cardiovascular disease, fatty liver, and several cancers. Several reports have indicated a link between hormonal imbalances and insulin resistance or obesity. Transgenic (TG) female mice overexpressing the human chorionic gonadotropin ß-subunit (hCGß+ mice) exhibit constitutively elevated levels of hCG, increased production of testosterone, progesterone and prolactin, and obesity. The objective of this study was to investigate the influence of hCG hypersecretion on possible alterations in the glucose and lipid metabolism of adult TG females. We evaluated fasting serum insulin, glucose, and triglyceride levels in adult hCGß+ females and conducted intraperitoneal glucose and insulin tolerance tests at different ages. TG female mice showed hyperinsulinemia, hypertriglyceridemia, and dyslipidemia, as well as glucose intolerance and insulin resistance at 6 months of age. A 1-week treatment with the dopamine agonist cabergoline applied on 5-week-old hCGß+ mice, which corrected hyperprolactinemia, hyperandrogenism, and hyperprogesteronemia, effectively prevented the metabolic alterations. These data indicate a key role of the hyperprolactinemia-induced gonadal dysfunction in the metabolic disturbances of hCGß+ female mice. The findings prompt further studies on the involvement of gonadotropins and prolactin on metabolic disorders and might pave the way for the development of new therapeutic strategies.


Subject(s)
Chorionic Gonadotropin, beta Subunit, Human/metabolism , Glucose Intolerance/metabolism , Hyperinsulinism/metabolism , Hyperprolactinemia/metabolism , Hypertriglyceridemia/metabolism , Insulin Resistance/physiology , Animals , Blood Glucose/metabolism , Cabergoline , Chorionic Gonadotropin, beta Subunit, Human/genetics , Ergolines/therapeutic use , Female , Glucose Intolerance/drug therapy , Glucose Intolerance/genetics , Hyperinsulinism/drug therapy , Hyperinsulinism/genetics , Hyperprolactinemia/drug therapy , Hyperprolactinemia/genetics , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/genetics , Insulin/blood , Mice , Mice, Transgenic , Prolactin/blood , Triglycerides/blood
15.
J Steroid Biochem Mol Biol ; 155(Pt A): 1-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26386462

ABSTRACT

OBJECTIVES: Glucocorticoid treatment induces insulin resistance (IR), which is counteracted by a compensatory hyperinsulinemia, due to increased pancreatic ß-cell function. There is evidence for also reduced hepatic insulin clearance, but whether this correlates with altered activity of insulin-degrading enzyme (IDE) in the liver, is not fully understood. Here, we investigated whether hyperinsulinemia, in glucocorticoid-treated rodents, is associated with any alteration in the insulin clearance and activity of the IDE in the liver. MATERIALS/METHODS: Adult male Swiss mice and Wistar rats were treated with the synthetic glucocorticoid dexamethasone intraperitoneally [1mg/kg body weight (b.w.)] for 5 consecutive days. RESULTS: Glucocorticoid treatment induced IR and hyperinsulinemia in both species, but was more impactful in rats that also displayed glucose intolerance and hyperglycemia. Insulin clearance was reduced in glucocorticoid-treated rats and mice, as judged by the reduction of insulin decay rate and increased insulin area-under-the-curve (47% and 87%, respectively). These results were associated with reduced activity (35%) of hepatic IDE in rats and a tendency to reduction (p=0.068) in mice, without alteration in hepatic IDE mRNA content, in both species. CONCLUSION: In conclusion, the reduced insulin clearance in glucocorticoid-treated rodents was due to the reduction of hepatic IDE activity, at least in rats, which may contributes to the compensatory hyperinsulinemia. These findings corroborate the idea that short-term and/or partial inhibition of IDE activity in the liver could be beneficial for the glycemic control.


Subject(s)
Dexamethasone/adverse effects , Hyperinsulinism/chemically induced , Insulin/metabolism , Insulysin/metabolism , Animals , Dexamethasone/pharmacology , Glucocorticoids/adverse effects , Glucocorticoids/pharmacology , Glucose Tolerance Test , Hyperinsulinism/metabolism , Insulin Resistance , Insulysin/genetics , Liver/drug effects , Liver/metabolism , Male , Mice , Rats, Wistar
17.
Horm Metab Res ; 47(12): 901-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26197851

ABSTRACT

Hyperandrogenemia, hyperinsulinemia, and obesity affect 60-70% of patients with Polycystic Ovarian Syndrome (PCOS), who exhibit an altered endometrial insulin signaling. The aim of the study was to evaluate whether hyperandrogenism, hyperinsulinism, and obesity present in PCOS patients impair the endometrial adiponectin signaling pathway. The ex vivo study was conducted on 27 samples from lean (n=9), obese (n=9), and obese-PCOS (n=9) patients. The in vitro assays were performed in immortalized human endometrial stromal cells stimulated with testosterone, insulin, or testosterone plus insulin. Serum steroid-hormones, adiponectin, glucose, and insulin; body mass index, free androgen index, ISI-Composite, and HOMA were evaluated in the 3 groups. Ex vivo and in vitro gene expression and protein content of adiponectin, AdipoR1, AdipoR2, and APPL1 were determined. Adiponectin serum levels were decreased in obese-PCOS patients compared to lean (78%) and obese (54%) controls (p<0.05). AdipoR1 protein and gene expression were increased in obese group vs. obese-PCOS and lean groups (2-fold, p<0.05). In turn, AdipoR2 protein and mRNA content was similar between the 3 groups. APPL1 protein levels were reduced in endometria from both obese groups, compared to lean group (6-fold, p<0.05). Testosterone plus insulin stimulation of T-HESC and St-T1b leads to a reduction of adiponectin, AdipoR1, AdipoR2, and APPL1 protein content in both endometrial cell lines (p<0.05), whereas, in the presence of testosterone or insulin alone, protein levels were similar to basal. Therefore, endometrial adiponectin-signaling pathway is impaired in hyperandrogenemic and hyperinsulinemic obese-PCOS patients, corroborated in the in vitro model, which could affect endometrial function and potentially the implantation process.


Subject(s)
Adiponectin/physiology , Endometrium/metabolism , Hyperinsulinism/metabolism , Obesity/metabolism , Polycystic Ovary Syndrome/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/analysis , Adiponectin/analysis , Adult , Cells, Cultured , Female , Humans , Receptors, Adiponectin/analysis
18.
Ann Anat ; 200: 88-97, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25819502

ABSTRACT

In this study, we investigated a possible sexual dimorphism regarding metabolic response and structural and functional adaptations of the endocrine pancreas after exposure to a high-fat diet (HFd). On chow diet, male and female C57BL/6/JUnib mice showed similar metabolic and morphometric parameters, except that female islets displayed a relatively lower ß-cell:non-ß-cell ratio. After 30 days on HFd, both male and female mice showed increased weight gain, however only the males displayed glucose intolerance associated with high postprandial glycemia when compared to their controls. After 60 days on HFd, both genders became obese, hyperglycemic, hyperinsulinemic, insulin resistant and glucose intolerant, although the metabolic changes were more pronounced in males, while females displayed greater weight gain. In both genders, insulin resistance induced by HFd feeding was compensated by expansion of ß-cell mass without changes in islet cytoarchitecture. Interestingly, we found a strong correlation between the degree of ß-cell expansion and the levels of hyperglycemia in the fed state: male mice fed a 60d-HFd, showing higher glycemic levels also displayed a greater ß-cell mass increase in comparison with female mice. Additionally, sexual dimorphism was also observed regarding the source of ß-cell mass expansion following 60d-HFd: while in males, both hypertrophy and hyperplasia (revealed by morphometry and Ki67 immunoreaction) of ß-cells were observed, female islets displayed only a significant increase in ß-cell size. In conclusion, this study describes gender differences in metabolic response to high fat diet, paralleled by distinct compensatory morphometric changes in pancreatic islets.


Subject(s)
Diet, High-Fat/adverse effects , Diet , Islets of Langerhans/pathology , Metabolic Diseases/pathology , Animals , Blood Glucose/metabolism , Cell Proliferation , Cell Size , Female , Glucose Intolerance , Hyperglycemia/metabolism , Hyperglycemia/pathology , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/ultrastructure , Male , Metabolic Diseases/etiology , Mice , Mice, Inbred C57BL , Sex Characteristics , Weight Gain
19.
Arq Bras Endocrinol Metabol ; 58(6): 600-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25211442

ABSTRACT

Overall excess of fat, usually defined by the body mass index, is associated with metabolic (e.g. glucose intolerance, type 2 diabetes mellitus (T2DM), dyslipidemia) and non-metabolic disorders (e.g. neoplasias, polycystic ovary syndrome, non-alcoholic fat liver disease, glomerulopathy, bone fragility etc.). However, more than its total amount, the distribution of adipose tissue throughout the body is a better predictor of the risk to the development of those disorders. Fat accumulation in the abdominal area and in non-adipose tissue (ectopic fat), for example, is associated with increased risk to develop metabolic and non-metabolic derangements. On the other hand, observations suggest that individuals who present peripheral adiposity, characterized by large hip and thigh circumferences, have better glucose tolerance, reduced incidence of T2DM and of metabolic syndrome. Insulin resistance (IR) is one of the main culprits in the association between obesity, particularly visceral, and metabolic as well as non-metabolic diseases. In this review we will highlight the current pathophysiological and molecular mechanisms possibly involved in the link between increased VAT, ectopic fat, IR and comorbidities. We will also provide some insights in the identification of these abnormalities.


Subject(s)
Adipose Tissue/physiopathology , Hyperinsulinism/complications , Insulin Resistance , Obesity/complications , Adipose Tissue/pathology , Animals , Apoptosis , Body Fat Distribution , Endoplasmic Reticulum/metabolism , Humans , Hyperinsulinism/metabolism , Mitochondria/metabolism , Obesity/metabolism , Obesity/physiopathology , Oxidation-Reduction , Oxidative Stress , Risk Assessment
20.
Revista Latinoamericana de Cirugía ; 4(2): 83-90, mayo-ago. 2014. ilus
Article in Spanish | BVSNACUY | ID: bnu-180638

ABSTRACT

Introducción: El retardo en la evacuación gástrica (REG) es de las complicaciones más frecuentes luego de la duodenopancreatectomía cefálica (DPC). Se encuentra muchas veces subregistrado. Este estudio mostrará su incidencia en una serie de 90 DPC según la clasificación de REG del consenso 2007 del ISGPS y analizaremos los factores desencadenantes. Material y métodos: Se realizó un estudio retrospectivo, descriptivo y observacional. Buscamos pacientes operados entre los años 1999 y 2014 por nuestro equipo. Se realizaron 16 duodenopancreatectomías con preservación pilórica y 57 operaciones tipo Whipple. Excluimos del estudio a 17 pacientes que tuvieron diferentes complicaciones asociadas que dificultaban la interpretación de los resultados, en especial la fístula pancreática tipo C que se asocia al REG, quedando 73 pacientes. Resultados: Siete pacientes tuvieron REG (9.4%). Se encontraron seis REG tipo C (85%) y uno tipo B (15%), y se analizaron las características de dichos pacientes. De los tipo C, tres fueron mujeres y tres hombres, todos en patología neoplásica maligna. La edad fue entre 49 y 67 años. Media: 58.16. Dos tuvieron preservación pilórica (12.5%) y cuatro, Whipple clásicos(7%), p =0.606. Se analizan todos los potenciales factores que pudieron incidir en dicha complicación. Conclusiones: El REG es una complicación frecuente luego de la DPC, que ocurre tanto en la preservación pilórica como en el Whipple clásico, y deben estudiarse las características de cada enfermo, así como los factores de riesgo, para diseñar una estrategia que minimice su incidencia.


Subject(s)
Adolescent , Young Adult , Adult , Gastrectomy , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/surgery , Hyperinsulinism/metabolism , Hyperinsulinism/surgery , Uruguay , Bariatric Surgery , Obesity, Morbid/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL