Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Adv Exp Med Biol ; 1370: 267-278, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35882802

RESUMO

Adaptation of islet ß-cell mass and function under limiting or excess nutrient availability is critical for maintenance of glucose homeostasis. Taurine regulates islet function of obese mice in normal and low dietary protein conditions, but whether this involves remodeling of the endocrine pancreas architecture is not well understood. Here, we carried functional and morphometric evaluation of the endocrine pancreas of normal and protein-restricted mice fed a high-fat diet (HFD) and investigated the role of taurine supplementation. Weaned mice were placed in a normal (C) or a low-protein diet (R) for 6 weeks, followed by HFD for 8 weeks (CH and RH). Half of HFD groups received 5% taurine supplementation since weaning (CHT and RHT) until the end of the experiment. Isolated islets from both CH and RH groups showed increased insulin release in association with increased pancreas weight and independently of changes in islet or ß-cell area. In normal protein CHT mice, taurine supplementation prevented obesity-induced insulin hypersecretion and promoted increased islet and ß-cell areas in association with increased protein expression of the proliferation marker, PCNA. On a low-protein background, taurine effects on islet function and morphology were blunted, but it prevented obesity-induced DNA fragmentation. In summary, taurine regulates islet function and morphology to improve the adaptive response to diet-induced obesity, but these effects are dependent on adequate dietary protein levels.


Assuntos
Ilhotas Pancreáticas , Taurina , Animais , Dieta Hiperlipídica/efeitos adversos , Proteínas Alimentares/metabolismo , Suplementos Nutricionais , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Taurina/metabolismo , Taurina/farmacologia
2.
Exp Physiol ; 106(7): 1482-1497, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33913203

RESUMO

NEW FINDINGS: What is the central question of this study? Type 1 diabetes mellitus (T1D) leads to hyperglycaemia owing to pancreatic ß-cell destruction by the immune system. Physical exercise has been shown to have potentially beneficial protective roles against cytokine-induced pancreatic ß-cell death, but its benefits are yet to be proved and should be understood better, especially in the islet environment. What is the main finding and its importance? Physical exercise protects against ß-cell loss in a well-described animal model for T1D, induced by multiple low doses of streptozotocin. This seems to be related to reduced cytokine-induced ß-cell death and increased islet cell proliferation. Contributions of islet neogenesis and/or transdifferentiation of pancreatic non-ß-cells into ß-cells cannot be excluded. ABSTRACT: Physical exercise has beneficial effects on pancreatic ß-cell function and survival in a pro-inflammatory environment. Although these effects have been linked to decreased islet inflammation and modulation of pro-apoptotic pathways, little is known about the islet microenvironment. Our aim was to evaluate the effects of physical exercise in islet histomorphology in a mouse model of type 1 diabetes mellitus induced by multiple low doses of streptozotocin. As expected, induction of type 1 diabetes mellitus led to ß-cell loss and, consequently, decreased islet area. Interestingly, although the decrease in islet area was not prevented by physical exercise, this was not the case for the decrease in ß-cell mass. This was probably related to induction of ß-cell regeneration, because we observed increased proliferation and regeneration markers, such as Ki67 and Pcna, in islets of trained mice. These were found in the central and peripheral regions of the islets. An increase in the percentage of α- and δ-cells in these conditions, combined with an increase in proliferation and Pax4 labelling in peripheral regions, suggest that ß-cell regeneration might also occur by transdifferentiation. This agrees with the presence of cells double stained for insulin and glucagon only in islets of diabetic trained mice. In addition, this group had more extra-islet insulin-positive cells and islets associated with ducts than diabetic mice. Physical exercise also decreased nuclear factor-κB activation in islet cells of diabetic trained compared with diabetic untrained mice, indicating a decrease in pro-inflammatory cytokine-induced ß-cell death. Taken together, these findings indicate that preservation of ß-cell mass induced by physical exercise involves an increase in ß-cell replication and decrease in ß-cell death, together with islet neogenesis and islet cell transdifferentiation.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Ilhotas Pancreáticas , Animais , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Glucagon/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Camundongos
3.
FASEB J ; 31(9): 4078-4087, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28572444

RESUMO

Taurine (Tau) restores ß-cell function in obesity; however, its action is lost in malnourished obese rodents. Here, we investigated the mechanisms involved in the lack of effects of Tau in this model. C57BL/6 mice were fed a control diet (CD) (14% protein) or a protein-restricted diet (RD) (6% protein) for 6 wk. Afterward, mice received a high-fat diet (HFD) for 8 wk [CD + HFD (CH) and RD + HFD (RH)] with or without 5% Tau supplementation after weaning on their drinking water [CH + Tau (CHT) and RH + Tau (RHT)]. The HFD increased insulin secretion through mitochondrial metabolism in CH and RH. Tau prevented all those alterations in CHT only. The expression of the taurine transporter (Tau-T), as well as Tau content in pancreatic islets, was increased in CH but had no effect on RH. Protein malnutrition programs ß cells and impairs Tau-induced restoration of mitochondrial metabolism and biogenesis. This may be associated with modulation of the expression of Tau-T in pancreatic islets, which may be responsible for the absence of effect of Tau in protein-malnourished obese mice.-Branco, R. C. S., Camargo, R. L., Batista, T. M., Vettorazzi, J. F., Borck, P. C., dos Santos-Silva, J. C. R., Boschero, A. C., Zoppi, C. C., Carneiro, E. M. Protein malnutrition blunts the increment of taurine transporter expression by a high-fat diet and impairs taurine reestablishment of insulin secretion.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Proteínas Alimentares/administração & dosagem , Insulina/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Deficiência de Proteína/metabolismo , Taurina/farmacologia , Animais , Linhagem Celular , Suplementos Nutricionais , Regulação da Expressão Gênica/fisiologia , Ilhotas Pancreáticas , Masculino , Glicoproteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Taurina/administração & dosagem
4.
Amino Acids ; 50(6): 765-774, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29556780

RESUMO

The sulfur-containing amino acid, taurine (Tau), regulates glucose and lipid homeostasis under normal, pre- and diabetic conditions. Here, we aimed to verify whether Tau supplementation exerts its beneficial effects against obesity, hyperglycemia and alterations in islet functions, in leptin-deficient obese (ob/ob), over a long period of treatment. From weaning until 12 months of age, female ob/ob mice received, or not, 5% Tau in drinking water (obTau group). After this period, a reduction in hypertriglyceridemia and an improvement in glucose tolerance and insulin sensitivity were observed in obTau mice. In addition, the daily metabolic flexibility was restored in obTau mice. In the gastrocnemius muscle of obTau mice, the activation of AMP-activated protein kinase (AMPK) was increased, while total AMPK protein content was reduced. Finally, isolated islets from obTau mice expressed high amounts of pyruvate carboxylase (PC) protein and lower glucose-induced insulin secretion. Taking these evidences together Tau supplementation had long-term positive actions on glucose tolerance and insulin sensitivity, associated with a reduction in glucose-stimulated insulin secretion, in ob/ob mice. The improvement in insulin actions in obTau mice was due, at least in part, to increased activation of AMPK in skeletal muscle, while the increased content of the PC enzyme in pancreatic islets may help to preserve glucose responsiveness in obTau islets, possibly contributing to islet cell survive.


Assuntos
Glicemia/metabolismo , Homeostase/efeitos dos fármacos , Hipertrigliceridemia , Taurina/farmacologia , Animais , Teste de Tolerância a Glucose , Hipertrigliceridemia/sangue , Hipertrigliceridemia/tratamento farmacológico , Hipertrigliceridemia/patologia , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Obesos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia
5.
Neural Plast ; 2017: 9652978, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28951790

RESUMO

The aim of this study was to investigate the effect of subdiaphragmatic vagotomy on insulin sensitivity, secretion, and degradation in metabolic programmed mice, induced by a low-protein diet early in life, followed by exposure to a high-fat diet in adulthood. Weaned 30-day-old C57Bl/6 mice were submitted to a low-protein diet (6% protein). After 4 weeks, the mice were distributed into three groups: LP group, which continued receiving a low-protein diet; LP + HF group, which started to receive a high-fat diet; and LP + HFvag group, which underwent vagotomy and also was kept at a high-fat diet. Glucose-stimulated insulin secretion (GSIS) in isolated islets, ipGTT, ipITT, in vivo insulin clearance, and liver expression of the insulin-degrading enzyme (IDE) was accessed. Vagotomy improved glucose tolerance and reduced insulin secretion but did not alter adiposity and insulin sensitivity in the LP + HFvag, compared with the LP + HF group. Improvement in glucose tolerance was accompanied by increased insulinemia, probably due to a diminished insulin clearance, as judged by the lower C-peptide : insulin ratio, during the ipGTT. Finally, vagotomy also reduced liver IDE expression in this group. In conclusion, when submitted to vagotomy, the metabolic programmed mice showed improved glucose tolerance, associated with an increase of plasma insulin concentration as a result of insulin clearance reduction, a phenomenon probably due to diminished liver IDE expression.


Assuntos
Resistência à Insulina/fisiologia , Insulina/metabolismo , Obesidade/cirurgia , Vagotomia/métodos , Animais , Dieta Hiperlipídica , Dieta com Restrição de Proteínas , Glucose/metabolismo , Insulisina/metabolismo , Fígado/metabolismo , Camundongos , Obesidade/metabolismo
6.
Amino Acids ; 43(4): 1791-801, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22418865

RESUMO

Taurine (Tau) is involved in beta (ß)-cell function and insulin action regulation. Here, we verified the possible preventive effect of Tau in high-fat diet (HFD)-induced obesity and glucose intolerance and in the disruption of pancreatic ß-cell morpho-physiology. Weaning Swiss mice were distributed into four groups: mice fed on HFD diet (36 % of saturated fat, HFD group); HTAU, mice fed on HFD diet and supplemented with 5 % Tau; control (CTL); and CTAU. After 19 weeks of diet and Tau treatments, glucose tolerance, insulin sensitivity and islet morpho-physiology were evaluated. HFD mice presented higher body weight and fat depots, and were hyperglycemic, hyperinsulinemic, glucose intolerant and insulin resistant. Their pancreatic islets secreted high levels of insulin in the presence of increasing glucose concentrations and 30 mM K(+). Tau supplementation improved glucose tolerance and insulin sensitivity with a higher ratio of Akt phosphorylated (pAkt) related to Akt total protein content (pAkt/Akt) following insulin administration in the liver without altering body weight and fat deposition in HTAU mice. Isolated islets from HTAU mice released insulin similarly to CTL islets. HFD intake induced islet hypertrophy, increased ß-cell/islet area and islet and ß-cell mass content in the pancreas. Tau prevented islet and ß-cell/islet area, and islet and ß-cell mass alterations induced by HFD. The total insulin content in HFD islets was higher than that of CTL islets, and was not altered in HTAU islets. In conclusion, for the first time, we showed that Tau enhances liver Akt activation and prevents ß-cell compensatory morpho-functional adaptations induced by HFD.


Assuntos
Dieta Hiperlipídica , Suplementos Nutricionais , Intolerância à Glucose/prevenção & controle , Hiperglicemia/prevenção & controle , Células Secretoras de Insulina/efeitos dos fármacos , Obesidade/prevenção & controle , Taurina/farmacologia , Animais , Glicemia/metabolismo , Peso Corporal , Feminino , Intolerância à Glucose/etiologia , Intolerância à Glucose/metabolismo , Teste de Tolerância a Glucose , Hiperglicemia/etiologia , Hiperglicemia/metabolismo , Insulina/metabolismo , Resistência à Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Obesidade/etiologia , Obesidade/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo
7.
Can J Physiol Pharmacol ; 90(7): 837-50, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22712703

RESUMO

In this study, we investigated the cellular distribution of junctional proteins and the dependence on cell-cell contacts of pancreatic beta cells during animal development. Fetus and newborn rat islets, which display a relatively poor insulin secretory response to glucose, present an immature morphology and cytoarchitecture when compared with young and adult islets that are responsive to glucose. At the perinatal stage, beta cells display a low junctional content of neural cell adhesion molecule (N-CAM), α- and ß-catenins, ZO-1, and F-actin, while a differential distribution of N-CAM and Pan-cadherin was seen in beta cells and nonbeta cells only from young and adult islets. In the absence of intercellular contacts, the glucose-stimulated insulin secretion was completely blocked in adult beta cells, but after reaggregation they partially reestablished the secretory response to glucose. By contrast, neonatal beta cells were poorly responsive to sugar, regardless of whether they were arranged as intact islets or as isolated cells. Interestingly, after 10 days of culturing, neonatal beta cells, known to display increased junctional protein content in vitro, became responsive to glucose and concomitantly dependent on cell-cell contacts. Therefore, our data suggest that the developmental acquisition of an adult-like insulin secretory pattern is paralleled by a dependence on direct cell-cell interactions.


Assuntos
Comunicação Celular/fisiologia , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Proteínas Musculares/metabolismo , Actinas/metabolismo , Animais , Feminino , Glucose/metabolismo , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/patologia , Masculino , Moléculas de Adesão de Célula Nervosa/metabolismo , Ratos , Ratos Wistar , Proteína da Zônula de Oclusão-1/metabolismo , alfa Catenina/metabolismo , beta Catenina/metabolismo
8.
Biomed Pharmacother ; 141: 111807, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34120066

RESUMO

Agomelatine (AGO) is an antidepressant drug with agonistic activity at melatonin receptor 1 (MT1) and MT2 and with neutral antagonistic activity at serotonin receptor 5-HT2C. Although experimental studies show that melatonin reduces hypertriglyceridemia and hepatic steatosis induced by excessive fructose intake, no studies have tested if AGO exerts similar actions. To address this issue we have treated male Wistar rats with fructose (15% in the drinking water) and/or AGO (40 mg/kg/day) for two weeks. AGO reduced body weight gain, feeding efficiency and hepatic lipid levels without affecting caloric intake in fructose-treated rats. AGO has also decreased very low-density lipoprotein (VLDL) production and circulating TAG levels after an oral load with olive oil. Accordingly, treatment with AGO reduced the hepatic expression of fatty acid synthase (Fasn), a limiting step for hepatic de novo lipogenesis (DNLG). The expression of apolipoprotein B (Apob) and microsomal triglyceride transfer protein (Mttp) in the ileum, two crucial proteins for intestinal lipoprotein production, were also downregulated by treatment with AGO. Altogether, the present data show that AGO mimics the metabolic benefits of melatonin when used in fructose-treated rats. This study also suggests that it is relevant to evaluate the potential of AGO to treat metabolic disorders in future clinical trials.


Assuntos
Acetamidas/farmacologia , Fígado Gorduroso/tratamento farmacológico , Frutose/farmacologia , Hipolipemiantes/farmacologia , Receptores de Melatonina/agonistas , Triglicerídeos/farmacologia , Acetamidas/uso terapêutico , Animais , Apolipoproteínas B/metabolismo , Peso Corporal/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Ingestão de Energia , Hipertrigliceridemia , Hipolipemiantes/uso terapêutico , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipoproteínas VLDL/sangue , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Melatonina/metabolismo , Azeite de Oliva/farmacologia , Ratos , Ratos Wistar , Triglicerídeos/uso terapêutico
9.
Life Sci ; 265: 118765, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33189820

RESUMO

The progeny of rats born and breastfed by mothers receiving dexamethasone (DEX) during pregnancy exhibits permanent reduction in body weight and adiposity but the precise mechanisms related to this programming are not fully understood. In order to clarify this issue, the present study investigated key aspects of lipoprotein production and lipid metabolism by the liver and the intestine that would explain the reduced adiposity seen in the adult offspring exposed to DEX in utero. Female Wistar rats were treated with DEX (0.1 mg/kg/day) between the 15th and the 21st days of pregnancy, while control mothers were treated with vehicle. Male offspring born to control mothers were nursed by either adoptive control mothers (CTL/CTL) or DEX-treated mothers (CTL/DEX). Male offspring born to DEX-treated mothers were nursed by either control mothers (DEX/CTL) or adoptive DEX-treated mothers (DEX/DEX). We found that only the male DEX/DEX offspring had reduced adiposity. Additionally, male DEX/DEX progeny had lower circulating triacylglycerol (TAG) levels only in fed-state. The four groups of offspring presented similar energy expenditure, respiratory quotient and very low-density lipoprotein (VLDL) production. On the other hand, DEX/DEX rats displayed reduced TAG levels after gavage with olive oil and reduced expression of fatty acid translocase Cd36 (Fat/Cd36) and peroxisome proliferator-activated receptor γ (Pparg) in the jejunum. Altogether, our study supports the notion that reduced fat absorption by the jejunum may contribute to the lower adiposity of the adult offspring born and breastfed by mothers treated with DEX during pregnancy.


Assuntos
Antígenos CD36/metabolismo , Dexametasona/farmacologia , Ácidos Graxos/metabolismo , Jejuno/efeitos dos fármacos , PPAR gama/metabolismo , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Animais , Calorimetria Indireta , Colesterol/metabolismo , Feminino , Trânsito Gastrointestinal/efeitos dos fármacos , Jejuno/metabolismo , Masculino , Reação em Cadeia da Polimerase , Gravidez , Ratos , Ratos Wistar , Triglicerídeos/metabolismo
13.
Life Sci ; 255: 117810, 2020 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-32473248

RESUMO

AIMS: The aim of the present study was to clarify if in utero exposure to DEX would affect the development of different types of pancreatic endocrine cells during postnatal life. MAIN METHODS: We investigated morphological and transcriptional features of both pancreatic ß- and α-cell populations within the pancreatic islets during the early postnatal life of rats born to mothers treated with DEX (0.1 mg/kg) from day 14 to 19 of pregnancy. Untreated pregnant Wistar rats of the same age (12-week-old) were used as control (CTL). Pups were euthanized on the 1st, 3rd and 21st (PND1, PND3 and PND21, respectively) days of life, regardless of sex. Serum insulin and glucagon levels were also evaluated. KEY FINDINGS: Rats born to DEX-treated mothers exhibited increased pancreatic α-cell mass, circulating glucagon levels and Gcg, Pax6, MafB and Nkx2.2 expression. Rats born to DEX-treated mothers also presented a rise in serum insulin levels on the PND3 that was paralleled by reduced ß-cell mass. Such increase in serum insulin levels, instead, was associated with increased expression of genes associated to insulin secretion such as Gck and Slc2a2. SIGNIFICANCE: Altogether, the present data reveals yet unknown changes in endocrine pancreas during early postnatal life of rats exposed to DEX in utero. Such data may contribute to the understanding of the metabolic features of rats born to DEX-treated mothers.


Assuntos
Dexametasona/toxicidade , Células Secretoras de Glucagon/efeitos dos fármacos , Glucocorticoides/toxicidade , Células Secretoras de Insulina/efeitos dos fármacos , Animais , Dexametasona/administração & dosagem , Feminino , Regulação da Expressão Gênica , Glucagon/sangue , Células Secretoras de Glucagon/citologia , Glucocorticoides/administração & dosagem , Proteína Homeobox Nkx-2.2 , Insulina/sangue , Secreção de Insulina/fisiologia , Células Secretoras de Insulina/citologia , Masculino , Gravidez , Efeitos Tardios da Exposição Pré-Natal/fisiopatologia , Ratos , Ratos Wistar
14.
Endocr Connect ; 9(4): 299-308, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32182583

RESUMO

PURPOSE: Observational studies show that longer breastfeeding periods reduce maternal risk of type 2 diabetes mellitus. However, it is currently unknown if the long-term benefits of breastfeeding for maternal glucose homeostasis are linked to changes in the endocrine pancreas. METHODS: We presently evaluated functional, morphological and molecular aspects of the endocrine pancreas of mice subjected to two sequential cycles of pregnancy and lactation (L21). Age-matched mice not allowed to breastfeed (L0) and virgin mice were used as controls. RESULTS: L21 mice exhibited increased tolerance and increased glucose-stimulated insulin secretion (GSIS) by isolated islets. Pancreatic islets of L21 mice did not present evident morphological changes to justify the increased GSIS. On the other hand, islets of L21 mice exhibited a reduction in Cavb3 and Kir6.2 expression with concordant increased intracellular Ca2+ levels after challenge with glucose. CONCLUSION: Altogether, the present findings show the breastfeeding exerts long-term benefits for maternal endocrine pancreas by increasing intracellular Ca2+ levels and GSIS.

15.
Endocr Connect ; 8(2): 120-131, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30768422

RESUMO

Pancreatic islets from pregnant rats develop a transitory increase in the pancreatic ß-cell proliferation rate and mass. Increased apoptosis during early lactation contributes to the rapid reversal of those morphological changes. Exposure to synthetic glucocorticoids during pregnancy has been previously reported to impair insulin secretion, but its impacts on pancreatic islet morphological changes during pregnancy and lactation have not been described. To address this issue, we assessed the morphological and molecular characteristics of pancreatic islets from rats that underwent undisturbed pregnancy (CTL) or were treated with dexamethasone between the 14th and 19th days of pregnancy (DEX). Pancreatic islets were analyzed on the 20th day of pregnancy (P20) and on the 3rd, 8th, 14th and 21st days of lactation (L3, L8, L14 and L21, respectively). Pancreatic islets from CTL rats exhibited transitory increases in cellular proliferation and pancreatic ß-cell mass at P20, which were reversed at L3, when a transitory increase in apoptosis was observed. This was followed by the appearance of morphological features of pancreatic islet neogenesis at L8. Islets from DEX rats did not demonstrate an increase in apoptosis at L3, which coincided with an increase in the expression of M2 macrophage markers relative to M1 macrophage and T lymphocyte markers. Islets from DEX rats also did not exhibit the morphological characteristics of pancreatic islet neogenesis at L8. Our data demonstrate that maternal pancreatic islets undergo a renewal process during lactation that is impaired by exposure to DEX during pregnancy.

16.
Life Sci ; 217: 261-270, 2019 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-30562489

RESUMO

AIMS: The present investigation evaluated whether pregnancy followed by lactation exerts long-term impacts on maternal hepatic lipid metabolism. MAIN METHODS: Female mice were subjected to two pregnancies, after which they were either allowed to breastfeed their pups for 21 days (L21) or had their litter removed (L0). Age-matched virgin mice were used as controls (CTL). Three months after the second delivery, serum was collected for lipid profiling, and fragments of liver were used to assess lipid content and to evaluate the key steps of de novo non-esterified fatty acid (NEFA) synthesis, esterification and ß-oxidation, very low density lipoprotein (VLDL) assembly and secretion and autophagy. KEY FINDINGS: L0 exhibited a significant increase in hepatic TG and reduced apolipoprotein B-100 (ApoB-100) expression. L21 mice had increased ATP citrate lyase (ACLY) activity and reduced acetyl-CoA carboxylase (ACC) phosphorylation but no increased hepatic TG. On the other hand, L21 mice had reduced hepatic sequestosome 1 (SQSTM1/p62) levels. Increased high density lipoprotein (HDL) cholesterol and hepatic apolipoprotein A-1 (ApoA-1) expression were found exclusively in L21. SIGNIFICANCE: The present study reveals that long-term hepatic lipid accumulation is induced by the history of pregnancy without lactation. On the other hand, reduced SQSTM1/p62 levels indicate that increased autophagic flux during life may prevent hepatic fat in dams subjected to lactation. Lactation after pregnancy is also obligatory for a long-term increase in maternal HDL. The present data may contribute to the understanding of the mechanisms leading to elevated cardiometabolic risk in women limited to short periods of lactation.


Assuntos
Lactação , Metabolismo dos Lipídeos , Fígado/metabolismo , Animais , Ácidos Graxos não Esterificados/análise , Ácidos Graxos não Esterificados/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Oxirredução , Gravidez , Triglicerídeos/análise , Triglicerídeos/metabolismo
17.
Sci Rep ; 7(1): 10367, 2017 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-28871187

RESUMO

We investigated the effect of dexamethasone during the last week of pregnancy on glucose and lipid metabolism in male offspring. Twelve-week old offspring were evaluated after fasting for 12-hours (physiological) and 60-hours (prolonged). Physiological fasting resulted in glucose intolerance, decreased glucose clearance after pyruvate load and increased PEPCK expression in rats born to dexamethasone-treated mothers (DEX). Prolonged fasting resulted in increased glucose tolerance and increased glucose clearance after pyruvate load in DEX. These modulations were accompanied by accumulation of hepatic triglycerides (TG). Sixty-hour fasted DEX also showed increased citrate synthase (CS) activity, ATP citrate lyase (ACLY) content, and pyruvate kinase 2 (pkm2), glucose transporter 1 (slc2a1) and lactate dehydrogenase-a (ldha) expressions. Hepatic AKT2 was increased in 60-hour fasted DEX, in parallel with reduced miRNAs targeting the AKT2 gene. Altogether, we show that metabolic programming by prenatal dexamethasone is characterized by an unexpected hepatic TG accumulation during prolonged fasting. The underlying mechanism may depend on increased hepatic glycolytic flux due to increased pkm2 expression and consequent conversion of pyruvate to non-esterified fatty acid synthesis due to increased CS activity and ACLY levels. Upregulation of AKT2 due to reduced miRNAs may serve as a permanent mechanism leading to increased pkm2 expression.


Assuntos
Dexametasona/farmacologia , Jejum/metabolismo , Fígado/metabolismo , Exposição Materna/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal , Triglicerídeos/metabolismo , Animais , Biomarcadores , Feminino , Glucose/metabolismo , Intolerância à Glucose , Testes de Função Hepática , Gravidez , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Fatores de Tempo
18.
Life Sci ; 127: 53-8, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25744409

RESUMO

AIMS: ARHGAP21 is a Rho GTPase-activating protein (RhoGAP) that associates with many proteins and modulates several cellular functions, including actin cytoskeleton rearrangement in different tissues. However, it is unknown whether ARHGAP21 is expressed in pancreatic beta cells and its function in these cells. Herein, we assess the participation of ARHGAP21 in insulin secretion. MAIN METHODS: Neonatal mice were treated with anti-sense oligonucleotide against ARHG AP21 (AS) for 2 days, resulting in a reduction of the protein's expression of about 60% in the islets. F-actin depolimerization, insulin secretion,mRNA level of genes involved in insulin secretion, maturation and proliferation were evaluated in islets from both control and AS-treated mice. KEY FINDINGS: ARHGAP21 co-localized with actin inMIN6 beta cells and with insulin in neonatal pancreatic islets. F-actin was reduced in AS-islets, as judged by lower phalloidin intensity. Insulin secretion was increased in islets from AS-treated mice, however no differences were observed in the GSIS (glucose-stimulated insulin secretion). In these islets, the pERK1/2 was increased, as well as the gene expressions of VAMP2 and SNAP25, proteins that are present in the secretory machinery. Maturation and cell proliferation were not affected in islets from AS-treated mice. SIGNIFICANCE: In conclusion, our data show, for the first time, that ARHGAP21 is expressed and participates in the secretory process of pancreatic beta cells. Its effect is probably via pERK1/2, which modulates the rearrangement of the cytoskeleton. ARHGAP21 also controls the expression of genes that encodes proteins of the secretory machinery.


Assuntos
Actinas/efeitos dos fármacos , Actinas/metabolismo , Proteínas Ativadoras de GTPase/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Animais , Animais Recém-Nascidos , DNA/biossíntese , DNA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Insulina/biossíntese , Insulina/genética , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos
19.
Mol Cell Endocrinol ; 404: 46-55, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25633666

RESUMO

Obesity is associated with insulin resistance and is known to be a risk factor for type-2 diabetes. In obese individuals, pancreatic beta-cells try to compensate for the increased insulin demand in order to maintain euglycemia. Most studies have reported that this adaptation is due to morphological changes. However, the involvement of beta-cell functional adaptations in this process needs to be clarified. For this purpose, we evaluated different key steps in the glucose-stimulated insulin secretion (GSIS) in intact islets from female ob/ob obese mice and lean controls. Obese mice showed increased body weight, insulin resistance, hyperinsulinemia, glucose intolerance and fed hyperglycemia. Islets from ob/ob mice exhibited increased glucose-induced mitochondrial activity, reflected by enhanced NAD(P)H production and mitochondrial membrane potential hyperpolarization. Perforated patch-clamp examination of beta-cells within intact islets revealed several alterations in the electrical activity such as increased firing frequency and higher sensitivity to low glucose concentrations. A higher intracellular Ca(2+) mobilization in response to glucose was also found in ob/ob islets. Additionally, they displayed a change in the oscillatory pattern and Ca(2+) signals at low glucose levels. Capacitance experiments in intact islets revealed increased exocytosis in individual ob/ob beta-cells. All these up-regulated processes led to increased GSIS. In contrast, we found a lack of beta-cell Ca(2+) signal coupling, which could be a manifestation of early defects that lead to beta-cell malfunction in the progression to diabetes. These findings indicate that beta-cell functional adaptations are an important process in the compensatory response to obesity.


Assuntos
Glicemia/metabolismo , Células Secretoras de Insulina/patologia , Insulina/metabolismo , Obesidade/fisiopatologia , Estado Pré-Diabético/fisiopatologia , Animais , Cálcio/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Resistência à Insulina , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Obesos , Obesidade/metabolismo , Estado Pré-Diabético/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA