Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Language
Publication year range
1.
Int J Obes (Lond) ; 31(10): 1586-95, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17471296

ABSTRACT

OBJECTIVE: We recently described that hypertriglyceridemic apolipoprotein (apo) CIII transgenic mice show increased whole body metabolic rate. In this study, we used these apo CIII-expressing mice, combined or not with the expression of the natural promoter-driven CETP gene, to test the hypothesis that both proteins modulate diet-induced obesity. MEASUREMENTS AND RESULTS: Mice expressing apo CIII, CIII/CETP, CETP and nontransgenic (NonTg) mice were maintained on a high-fat diet (14% fat by weight) during 20 weeks after weaning. At the end of this period, all groups exhibited the expected lipemic phenotype. Fasting glucose levels were neither affected by the high-fat diet nor by the distinct genotypes. However, apo CIII mice showed significantly higher glycemia ( approximately 35%) and lower insulin levels ( approximately 45%) in the fed state, compared with the NonTg mice. The apo CIII mice presented significantly increased body weight, lipid content of the carcass ( approximately 25%), visceral adipose tissue mass (about twofold) and adipocyte size ( approximately 25%) compared with the CETP and NonTg mice. The CETP expression in the apo CIII background normalized the subcutaneous adipose depot and visceral adipocyte size to the levels of NonTg mice. Plasma leptin levels were lower in CETP groups (25-50%) and higher in the apo CIII mice. Similar core body temperature in all groups and similar liver mitochondrial resting respiration rates in CIII and NonTg mice indicate no differences in basal energy expenditure rates among these mice fed a high-fat diet. CONCLUSION: The elevation of plasma apo CIII levels aggravates diet-induced obesity and the expression of physiological levels of circulating CETP reverses this adipogenic effect, indicating a novel role for CETP in modulating adiposity.


Subject(s)
Apolipoprotein C-III/physiology , Cholesterol Ester Transfer Proteins/physiology , Fasting/metabolism , Hypertriglyceridemia/metabolism , Leptin/metabolism , Animals , Body Composition/physiology , Body Weight/physiology , Fasting/blood , Hypertriglyceridemia/blood , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity
2.
J Endocrinol ; 181(1): 25-38, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15072564

ABSTRACT

Autocrine and paracrine insulin signaling may participate in the fine control of insulin secretion. In the present study, tissue distribution and protein amounts of the insulin receptor and its major substrates, insulin receptor substrate (IRS)-1 and IRS-2, were evaluated in a model of impaired glucose-induced insulin secretion, the protein-deficient rat. Immunoblot and RT-PCR studies showed that the insulin receptor and IRS-2 expression are increased, whilst IRS-1 protein and mRNA contents are decreased in pancreatic islets of protein-deficient rats. Immunohistochemical studies revealed that the insulin receptor and IRS-1 and -2 are present in the great majority of islet cells; however, the greatest staining was localized at the periphery, suggesting a co-localization with non-insulin-secreting cells. Exogenous insulin stimulation of isolated islets promoted higher insulin receptor and IRS-1 and -2 tyrosine phosphorylation in islets from protein-deficient rats, as compared with controls. Moreover, insulin-induced IRS-1- and IRS-2-associated phosphatidylinositol 3-kinase activity are increased in islets of protein-deficient rats. The reduction of IRS-1 and IRS-2 protein expression in islets isolated from protein-deficient rats by the use of antisense IRS-1 or IRS-2 phosphorthioate-modified oligonucleotides partially restored glucose-induced insulin secretion. Thus, the impairment of insulin cell signaling through members of the IRS family of proteins in isolated rat pancreatic islets improves glucose-induced insulin secretion. The present data reinforced the role of insulin paracrine and autocrine signaling in the control of its own secretion.


Subject(s)
Insulin/metabolism , Islets of Langerhans/metabolism , Phosphoproteins/metabolism , Protein Deficiency/metabolism , Animals , Glucagon/metabolism , Glucose/administration & dosage , Immunohistochemistry/methods , Insulin Receptor Substrate Proteins , Insulin Secretion , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/chemistry , Male , Paracrine Communication/physiology , Phosphoproteins/analysis , Protein Deficiency/physiopathology , Rats , Rats, Wistar , Somatostatin/analysis
3.
Horm Metab Res ; 35(5): 282-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12915997

ABSTRACT

During pregnancy, pancreatic islets undergo structural and functional changes in response to an increased demand for insulin. Different hormones, especially placental lactogens, mediate these adaptive changes. Prolactin (PRL) mainly exerts its biological effects by activation of the JAK2/STAT5 pathway. PRL also stimulates some biological effects via activation of IRS-1, IRS-2, PI 3-kinase, and MAPK in different cell lines. Since IRS-2 is important for the maintenance of pancreatic islet cell mass, we investigated whether PRL affects insulin-signaling pathways in neonatal rat islets. PRL significantly potentiated glucose-induced insulin secretion in islets cultured for 7 days. This effect was blocked by the specific PI 3-kinase inhibitor wortmannin. To determine possible effects of PRL on insulin-signaling pathways, fresh islets were incubated with or without the hormone for 5 or 15 min. Immunoprecipitation and immunoblotting with specific antibodies showed that PRL induced a dose-dependent IRS-1 and IRS-2 phosphorylation compared to control islets. PRL-induced increase in IRS-1/-2 phosphorylation was accompanied by an increase in the association with and activation of PI 3-kinase. PRL-induced IRS-2 phosphorylation and its association with PI 3-kinase did not add to the effect of insulin. PRL also induced JAK2, SHC, ERK1 and ERK2 phosphorylation in neonatal islets, demonstrating that PRL can activate MAPK. These data indicate that PRL can stimulate the IRSs/PI 3-kinase and SHC/ERK pathways in islets from neonatal rats.


Subject(s)
Animals, Newborn/physiology , Insulin/physiology , Islets of Langerhans/metabolism , Prolactin/physiology , Proto-Oncogene Proteins , Signal Transduction/physiology , Animals , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Immunoblotting , Insulin/metabolism , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/enzymology , Janus Kinase 2 , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Rats , Receptor Cross-Talk/physiology , Tyrosine/metabolism
4.
Horm Metab Res ; 34(1): 21-6, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11832997

ABSTRACT

In this study, we examined glucose homeostasis and insulin secretion in transgenic mice overexpressing the human apolipoprotein CIII gene (apo CIII tg). These mice have elevated plasma levels of triglycerides, FFA and cholesterol compared to control mice. The body weight, plasma glucose, and insulin levels, glucose disappearance rates, areas under the ipGTT curve for adult (4 - 8 mo. old) and aged (20 - 24 mo. old) apo CIII tg mice and the determination of insulin during the ipGTT were not different from those of control mice. However, an additional elevation of plasma FFA by treatment with heparin for 2 - 4 h impaired the ipGTT responses in apo CIII tg mice compared to saline-treated mice. The glucose disappearance rate in heparin-treated transgenic mice was slightly lower than in heparin-treated controls. Glucose (22.2 mmol/l) stimulated insulin secretion in isolated islets to the same extent in saline-treated control and apo CIII tg mice. In islets from heparin-treated apo CIII tg mice, the insulin secretion at 2.8 and 22.2 mmol glucose/l was lower than in heparin-treated control mice. In conclusion, hypertriglyceridemia per se or a mild elevation in FFA did not affect insulin secretion or insulin resistance in adult or aged apo CIII tg mice. Nonetheless, an additional elevation of FFA induced by heparin in hypertriglyceridemic mice impaired the ipGTT by reducing insulin secretion.


Subject(s)
Blood Glucose/metabolism , Hypertriglyceridemia/blood , Insulin/metabolism , Islets of Langerhans/metabolism , Animals , Apolipoprotein C-III , Apolipoproteins C/physiology , Area Under Curve , Body Weight , Cholesterol/blood , Fatty Acids, Nonesterified , Female , Glucose Tolerance Test , Heparin/administration & dosage , Hypertriglyceridemia/physiopathology , Insulin/blood , Insulin/physiology , Insulin Secretion , Male , Mice , Mice, Transgenic , Statistics, Nonparametric , Triglycerides/blood
5.
Diabetes Metab ; 28(6 Pt 2): 3S13-7; discussion 3S108-12, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12688628

ABSTRACT

Pancreatic islet isolated from hyperinsulinemic obese rodents showed high glucose sensitivity to stimulation of insulin secretion. Current research evaluates the effect of subdiaphragmatic vagotomy on insulin secretion stimulated by glucose and by a cholinergic agonist carbachol (Cch) in islets isolated from monosodium L-glutamate (MSG)-obese rats. During the first 5 days after birth, MSG was intradermically injected into the cervical area of male Wistar rats (n=26). Control animals were injected with hyperosmotic saline solution (n=26). Vagotomy was performed on 30-day-old rats. On the 90th day, after a 12-h fast, the animals were killed. Pancreatic islets were isolated by collagenase. Batches of islets (n=30) were incubated for 60 min in glucose 2.8-20.0 mM or 0.1-2.0 mM Cch in the presence of glucose 8.3 mM. Released insulin was measured by radioimmunoassay. Insulin secretion stimulated by glucose in islets from MSG-obese rats shifted to the left when compared to that of controls, 63.8+/-3.9 and 42.2+/-2.6 ng/ml/islet/60 min (p<0.001), respectively. Vagotomy decreased by 56% (p<0.001) the secretion induced by all glucose concentrations in islets from MSG-obese rats. In the controls the operation caused a 30% (p<0.01) reduction. Cch stimulated insulin secretion in normal and obese rat islets. Response to obese rat islets was 1/3 less than normal ones (p<0.05). Vagotomy produced a greater potentiation of Cch induced insulin secretion on islets from obese rats. Data suggested that parasympathetic modulation is important to insulin secretion control.


Subject(s)
Islets of Langerhans/metabolism , Parasympathetic Nervous System/physiology , Sodium Glutamate/pharmacology , Vagotomy , Analysis of Variance , Animals , Carbachol/pharmacology , Glucose/pharmacology , In Vitro Techniques , Islets of Langerhans/drug effects , Kinetics , Male , Neurotransmitter Agents/metabolism , Obesity/physiopathology , Parasympathetic Nervous System/drug effects , Rats , Rats, Wistar , Reference Values
6.
J Nutr Biochem ; 12(5): 285-291, 2001 May.
Article in English | MEDLINE | ID: mdl-11382546

ABSTRACT

High protein content in the diet during childhood and adolescence has been associated to the onset insulin-dependent diabetes mellitus. We investigated the effect of interleukin-1beta (IL-1beta) on insulin secretion, glucose metabolism, and nitrite formation by islets isolated from rats fed with normal protein (NP, 17%) or low protein (LP, 6%) after weaning. Pretreatment of islets with IL-1beta for 1 h or 24 h inhibited the insulin secretion induced by glucose in both groups, but it was less marked in LP than in NP group. Islets from LP rats exhibited a decreased IL-1beta-induced nitric oxide (NO) production, lower inhibition of D-[U(14)C]-glucose oxidation to (14)CO(2) and less pronounced effect of IL-1beta on alpha-ketoisocaproic acid-induced insulin secretion than NP islets. However, when the islets were stimulated by high concentrations of K(+) the inhibitory effect of IL-1beta on insulin secretion was not different between groups. In conclusion, protein restriction protects beta-cells of the deleterious effect of IL-1beta, apparently, by decreasing NO production. The lower NO generation in islets from protein deprived rats may be due to increased free fatty acids oxidation and consequent alteration in Ca(2+) homeostasis.

7.
Journal of Arachnology ; 28(1): 7-15, 2000.
Article in English | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP, SESSP-IBACERVO | ID: biblio-1064175

ABSTRACT

Three Brazilian species of the genus Paradossenus F.O. Pickard-Cambridge 1903 are included in this paper: Paradossenus minimus (Mello-Leitão 1940), whose holotype was located and is here redescribed; Paradossenus corumba new species is described from Mato Grosso do Sul, Brazil and preliminary data on its biology are presented. Morphological data and new records of P. longipes (Taczanowski 1874) are included.


Subject(s)
Animals , Spiders/anatomy & histology , Spiders/classification , Species Specificity , Brazil
SELECTION OF CITATIONS
SEARCH DETAIL
...