Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38562689

RESUMEN

We previously showed that miR-146a-5p is upregulated in pancreatic islets treated with pro-inflammatory cytokines. Others have reported that miR-146a-5p overexpression is associated with ß cell apoptosis and impaired insulin secretion. However, the molecular mechanisms mediating these effects remain elusive. To investigate the role of miR-146a-5p in ß cell function, we developed stable MIN6 cell lines to either overexpress or inhibit the expression of miR-146a-5p. Monoclonal cell populations were treated with pro-inflammatory cytokines (IL-1ß, IFNγ, and TNFα) to model T1D in vitro. We found that overexpression of miR-146a-5p increased cell death under conditions of inflammatory stress, whereas inhibition of miR-146a-5p reversed these effects. Additionally, inhibition of miR-146a-5p increased mitochondrial DNA copy number, respiration rate, and ATP production. Further, RNA sequencing data showed enrichment of pathways related to insulin secretion, apoptosis, and mitochondrial function when the expression levels of miR-146a-5p were altered. Finally, a temporal increase in miR-146a-5p expression levels and a decrease in mitochondria function markers was observed in islets derived from NOD mice. Collectively, these data suggest that miR-146a-5p may promote ß cell dysfunction and death during inflammatory stress by suppressing mitochondrial function.

2.
Adv Exp Med Biol ; 1370: 267-278, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35882802

RESUMEN

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.


Asunto(s)
Islotes Pancreáticos , Taurina , Animales , Dieta Alta en Grasa/efectos adversos , Proteínas en la Dieta/metabolismo , Suplementos Dietéticos , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Taurina/metabolismo , Taurina/farmacología
3.
Life Sci ; 291: 120239, 2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-34942163

RESUMEN

Aim Investigate whether inheritance of improved skeletal muscle mitochondrial function and its association with glycemic control are multigenerational benefits of exercise. MAIN METHODS: Male Swiss mice were subjected to 8 weeks of endurance training and mated with untrained females. KEY FINDINGS: Trained fathers displayed typical endurance training-induced adaptations. Remarkably, offspring from trained fathers also exhibited higher endurance performance, mitochondrial oxygen consumption, glucose tolerance and insulin sensitivity. However, PGC-1α expression was not increased in the offspring. In the offspring, the expression of the co-repressor NCoR1 was reduced, increasing activation of PGC-1α target genes. These effects correlated with higher DNA methylation at the NCoR1 promoter in both, the sperm of trained fathers and in the skeletal muscle of their offspring. SIGNIFICANCE: Higher skeletal muscle mitochondrial function is inherited by epigenetic de-activation of a key PGC-1α co-repressor.


Asunto(s)
Mitocondrias/metabolismo , Condicionamiento Físico Animal/fisiología , Esfuerzo Físico/fisiología , Animales , Metilación de ADN , Epigénesis Genética/genética , Femenino , Masculino , Ratones , Mitocondrias/fisiología , Músculo Esquelético/fisiología , Co-Represor 1 de Receptor Nuclear/metabolismo , Consumo de Oxígeno/fisiología , Herencia Paterna/fisiología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Receptores Activados del Proliferador del Peroxisoma/fisiología , Condicionamiento Físico Animal/métodos , ARN Mensajero/genética
4.
Vitam Horm ; 115: 185-219, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33706949

RESUMEN

Aging is characterized by a progressive loss of physiological function leading to increase in the vulnerability to death. This deterioration process occurs in all living organisms and is the primary risk factor for pathological conditions including obesity, type 2 diabetes mellitus, Alzheimer's disease and cardiovascular diseases. Most of the age-related diseases have been associated with impairment of action of an important hormone, namely insulin. It is well-known that this hormone is a critical mediator of metabolism, growth, proliferation and differentiation. Insulin action depends on two processes that determine its circulating levels, insulin secretion and clearance, and insulin sensitivity in its target tissues. Aging has deleterious effects on these three mechanisms, impairing insulin action, thereby increasing the risk for diseases and death. Thus, improving insulin action may be an important strategy to have a healthier and longer life.


Asunto(s)
Diabetes Mellitus Tipo 2 , Resistencia a la Insulina , Envejecimiento/fisiología , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Insulina/metabolismo , Obesidad
5.
J Cell Physiol ; 234(5): 6313-6323, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30317568

RESUMEN

Nutrient malnutrition, during the early stages of development, may facilitate the onset of metabolic diseases later in life. However, the consequences of nutritional insults, such as a high-fat diet (HFD) after protein restriction, are still controversial. We assessed overall glucose homeostasis and molecular markers of mitochondrial function in the gastrocnemius muscle of protein-restricted mice fed an HFD until early adulthood. Male C57BL/6 mice were fed a control (14% protein-control diet) or a protein-restricted (6% protein-restricted diet) diet for 6 weeks. Afterward, mice received an HFD or not for 8 weeks (mice fed a control diet and HFD [CH] and mice fed a protein-restricted diet and HFD [RH]). RH mice showed lower weight gain and fat accumulation and did not show an increase in fasting plasma glucose and insulin levels compared with CH mice. RH mice showed higher energy expenditure, increased citrate synthase, peroxisome-proliferator-activated receptor gamma coactivator 1-alpha protein content, and higher levels of malate and α-ketoglutarate compared with CH mice. Moreover, RH mice showed increased AMPc-dependent kinase and acetyl coenzyme-A (CoA) carboxylase phosphorylation, lower intramuscular triacylglycerol content, and similar malonyl-CoA levels. In conclusion, protein undernourishment after weaning does not potentiate fat accumulation and insulin resistance in adult young mice fed an HFD. This outcome seems to be associated with increased skeletal muscle mitochondrial oxidative capacity and reduced lipids accumulation.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Glucosa/metabolismo , Homeostasis/fisiología , Músculo Esquelético/metabolismo , Deficiencia de Proteína/metabolismo , Animales , Metabolismo Energético/fisiología , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo
6.
Amino Acids ; 50(6): 765-774, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29556780

RESUMEN

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.


Asunto(s)
Glucemia/metabolismo , Homeostasis/efectos de los fármacos , Hipertrigliceridemia , Taurina/farmacología , Animales , Prueba de Tolerancia a la Glucosa , Hipertrigliceridemia/sangre , Hipertrigliceridemia/tratamiento farmacológico , Hipertrigliceridemia/patología , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Ratones , Ratones Obesos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología
7.
Sci Rep ; 7(1): 14876, 2017 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29093479

RESUMEN

Disruption of insulin secretion and clearance both contribute to obesity-induced hyperinsulinemia, though reduced insulin clearance seems to be the main factor. The liver is the major site for insulin degradation, a process mainly coordinated by the insulin-degrading enzyme (IDE). The beneficial effects of taurine conjugated bile acid (TUDCA) on insulin secretion as well as insulin sensitivity have been recently described. However, the possible role of TUDCA in insulin clearance had not yet been explored. Here, we demonstrated that 15 days treatment with TUDCA reestablished plasma insulin to physiological concentrations in high fat diet (HFD) mice, a phenomenon associated with increased insulin clearance and liver IDE expression. TUDCA also increased IDE expression in human hepatic cell line HepG2. This effect was not observed in the presence of an inhibitor of the hepatic membrane bile acid receptor, S1PR2, nor when its downstream proteins were inhibited, including IR, PI3K and Akt. These results indicate that treatment with TUDCA may be helpful to counteract obesity-induced hyperinsulinemia through increasing insulin clearance, likely through enhanced liver IDE expression in a mechanism dependent on S1PR2-Insulin pathway activation.


Asunto(s)
Insulina/farmacocinética , Insulisina/efectos de los fármacos , Hígado/enzimología , Ácido Tauroquenodesoxicólico/farmacología , Animales , Dieta Alta en Grasa , Células Hep G2 , Humanos , Hiperinsulinismo/tratamiento farmacológico , Insulisina/metabolismo , Hígado/metabolismo , Ratones , Ratones Obesos
8.
FASEB J ; 31(9): 4078-4087, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28572444

RESUMEN

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.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Proteínas en la Dieta/administración & dosificación , Insulina/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Deficiencia de Proteína/metabolismo , Taurina/farmacología , Animales , Línea Celular , Suplementos Dietéticos , Regulación de la Expresión Génica/fisiología , Islotes Pancreáticos , Masculino , Glicoproteínas de Membrana/genética , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Taurina/administración & dosificación
9.
Sci Rep ; 6: 33464, 2016 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-27633083

RESUMEN

Pancreatic beta cell (ß) dysfunction is an outcome of malnutrition. We assessed the role of the amplifying pathway (AMP PATH) in ß cells in malnourished obese mice. C57Bl-6 mice were fed a control (C) or a low-protein diet (R). The groups were then fed a high-fat diet (CH and RH). AMP PATH contribution to insulin secretion was assessed upon incubating islets with diazoxide and KCl. CH and RH displayed increased glucose intolerance, insulin resistance and glucose-stimulated insulin secretion. Only RH showed a higher contribution of the AMP PATH. The mitochondrial membrane potential of RH was decreased, and ATP flux was unaltered. In RH islets, glutamate dehydrogenase (GDH) protein content and activity increased, and the AMP PATH contribution was reestablished when GDH was blunted. Thus, protein malnutrition induces mitochondrial dysfunction in ß cells, leading to an increased contribution of the AMP PATH to insulin secretion through the enhancement of GDH content and activity.


Asunto(s)
Envejecimiento/patología , Insulina/metabolismo , Desnutrición Proteico-Calórica/metabolismo , Animales , Intolerancia a la Glucosa/complicaciones , Intolerancia a la Glucosa/metabolismo , Intolerancia a la Glucosa/patología , Glutamato Deshidrogenasa/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Ratones Endogámicos C57BL , Ratones Obesos , Mitocondrias/metabolismo , Desnutrición Proteico-Calórica/complicaciones , Desnutrición Proteico-Calórica/patología
10.
Metabolism ; 65(3): 54-63, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26892516

RESUMEN

OBJECTIVE: While bile acids are important for the digestion process, they also act as signaling molecules in many tissues, including the endocrine pancreas, which expresses specific bile acid receptors that regulate several cell functions. In this study, we investigated the effects of the conjugated bile acid TUDCA on glucose-stimulated insulin secretion (GSIS) from pancreatic ß-cells. METHODS: Pancreatic islets were isolated from 90-day-old male mice. Insulin secretion was measured by radioimmunoassay, protein phosphorylation by western blot, Ca(2+) signals by fluorescence microscopy and ATP-dependent K(+) (KATP) channels by electrophysiology. RESULTS: TUDCA dose-dependently increased GSIS in fresh islets at stimulatory glucose concentrations but remained without effect at low glucose levels. This effect was not associated with changes in glucose metabolism, Ca(2+) signals or KATP channel activity; however, it was lost in the presence of a cAMP competitor or a PKA inhibitor. Additionally, PKA and CREB phosphorylation were observed after 1-hour incubation with TUDCA. The potentiation of GSIS was blunted by the Gα stimulatory, G protein subunit-specific inhibitor NF449 and mimicked by the specific TGR5 agonist INT-777, pointing to the involvement of the bile acid G protein-coupled receptor TGR5. CONCLUSION: Our data indicate that TUDCA potentiates GSIS through the cAMP/PKA pathway.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , AMP Cíclico/fisiología , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Ácido Tauroquenodesoxicólico/farmacología , Animales , Señalización del Calcio/efectos de los fármacos , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Técnicas In Vitro , Células Secretoras de Insulina/metabolismo , Canales KATP/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/efectos de los fármacos
11.
Rev. bras. cineantropom. desempenho hum ; 18(1): 1-10, Jan.-Feb. 2016. tab
Artículo en Inglés | LILACS | ID: lil-778484

RESUMEN

Abstract Although it is a widely used resource for the treatment of musculoskeletal injuries, immobilization causes deleterious effects in muscle tissue after a short period of time. This study aimed to evaluate the gastrocnemius and tibialis anterior muscles of obese and protein malnourished animals under joint immobilization condition. Overall, 28 adult male mice were used (C57 / BL6), being divided into four groups (N = 7): Control Group (CG), Immobilized Control Group (ICG), Immobilized Obese Group (IOG) and Immobilized Malnourished Group (IMG). The immobilization protocol was performed by the use of adhesive tape and plaster. The conditions and obesity and protein malnutrition have been developed through the ingestion of diets specific for each group of animals. The histomorphometric analysis of muscles evaluated area and the diameter of muscle fibers. All immobilized groups showed reduction in the area and diameter of muscle fibers when compared to GC. Comparisons among immobilized groups showed that the area and diameter of muscle fibers of IOG and IMG were lower than ICG. The immobilization protocol caused reduction in muscle trophism in animals, and obese and malnourished animals suffered high losses under condition of muscle atrophy.


Resumo Embora seja um recurso muito utilizado para tratamento de lesões musculoesqueléticas, a imobilização causa efeitos deletérios no tecido muscular após curto período de tempo. O presente estudo teve como objetivo avaliar os músculos gastrocnêmio e tibial anterior de animais obesos e desnutridos proteicamente sob a condição de imobilização articular. Foram utilizados 28 camundongos (C57/BL6) machos adultos, distribuídos em quatro grupos (N=7): Grupo controle (GC), Grupo Controle Imobilizado (GCI), Grupo Obeso imobilizado (GOI) e Grupo Desnutrido Imobilizado (GDI). O protocolo de imobilização foi realizado por meio da utilização de tiras de esparadrapo e faixa gessada. As condições de obesidade e desnutrição proteica foram desenvolvidas por meio da ingestão de dietas específicas para cada grupo de animais. A análise histomorfométrica dos músculos avaliou a área e o diâmetro das fibras musculares. Todos os grupos imobilizados apresentaram redução na área e no diâmetro das fibras musculares quando comparados ao GC. As comparações entre os grupos imobilizados mostraram que os valores do diâmetro e área de fibras dos grupos GOI e GDI foram menores do que o GCI. O protocolo de imobilização provocou redução do trofismo muscular nos animais estudados e os animais obesos e desnutridos sofreram prejuízo elevado na condição de atrofia muscular.


Asunto(s)
Animales , Ratas , Músculo Esquelético/fisiopatología , Proteínas Musculares/deficiencia , Obesidad
13.
J Nutr Biochem ; 26(5): 556-62, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25736482

RESUMEN

Protein restriction in the early stages of life can result in several changes in pancreatic function. These alterations include documented reductions in insulin secretion and in cytoplasmic calcium concentration [Ca(2+)]i. However, the mechanisms underlying these changes have not been completely elucidated and may result, in part, from alterations in signaling pathways that potentiate insulin secretion in the presence of glucose. Our findings suggest that protein restriction disrupts the insulin secretory synergism between Cyclic adenosine monophosphate (cAMP)-dependent protein kinase (PKA) and Ca(2+)-dependent protein kinase C (PKC) in isolated islets. Western blot analysis demonstrated reduced levels of both phospho-cAMP response element-binding protein (phospho-CREB) at Ser-133 and substrates phosphorylated by PKCs (Phospho-(Ser) PKC substrate), suggesting that PKA and PKC activity was impaired in islets from rats fed a low-protein diet (LP). cAMP levels and global Ca(2+) entry were also reduced in LP islets. In summary, our findings showed that protein restriction altered the crosstalk between PKA and PKC signaling pathways, resulting in the alteration of secretory synergism in isolated islets.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Dieta con Restricción de Proteínas , Islotes Pancreáticos/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal , Animales , Técnicas In Vitro , Islotes Pancreáticos/enzimología , Masculino , Ratas , Ratas Wistar
14.
Cell Physiol Biochem ; 32(2): 310-21, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23942282

RESUMEN

BACKGROUND/AIMS: Metabolic syndrome has been identified as one of the most significant threats to human health in the 21(st) century. Exercise training has been shown to counteract obesity and metabolic syndrome. The present study aimed to investigate the effects of moderate exercise training on pancreatic beta-cell function and autonomic nervous system (ANS) activity in rats fed a high-fat diet (HFD). METHODS: Weaning rats were divided into four groups: rats fed a standard chow or HFD (sedentary, Control-SED and HFD-SED; or exercised, Control-EXE and HFD-EXE, respectively). Exercised rats ran (from 21- to 91-days-old) for 60 minutes (3 times/week) over a 10-week period. Glucose and insulin tolerance tests were performed. Pancreatic islets were isolated to study glucose-induced insulin secretion (GIIS). Parasympathetic and sympathetic nerve electrical signals were measured, and liver samples were processed and histologically analyzed. RESULTS: Exercise prevented obesity, insulin resistance, and liver steatosis as well as improved total cholesterol, ALT, and AST levels. Islets from HFD rats showed insulin hypersecretion which was ameliorated by exercise. Exercise decreased vagal nerve activity in the HFD-EXE group and increased the activity of the sympathetic nervous system in both exercised groups. CONCLUSION: Exercise prevents obesity and liver steatosis and restores pancreatic beta-cell function and ANS activity in HFD-obese rats.


Asunto(s)
Sistema Nervioso Autónomo/metabolismo , Dieta Alta en Grasa , Células Secretoras de Insulina/metabolismo , Condicionamiento Físico Animal , Animales , Células Cultivadas , Masculino , Obesidad/fisiopatología , Obesidad/terapia , Ratas , Ratas Wistar
15.
Endocrine ; 43(3): 571-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22983867

RESUMEN

The goal of the present study was to investigate changes on glucose homoeostasis and of the insulin receptor (IR) and insulin receptor substrate-1 (IRS-1) signalling in pancreatic islets from MSG-obese mice submitted to or not submitted to swim training. Swim training of 90-day-old MSG mice was used to evaluate whether signalling pathways of the IR and IRS-1 in islets are involved with the insulin resistance and glucose intolerance observed in this obese animal model. The results showed that IR tyrosine phosphorylation (pIR) was reduced by 42 % in MSG-obese mice (MSG, 6.7 ± 0.2 arbitrary units (a.u.); control, 11.5 ± 0.4 a.u.); on the other hand, exercise training increased pIR by 76 % in MSG mice without affecting control mice (MSG, 11.8 ± 0.3; control, 12.8 ± 0.2 a.u.). Although the treatment with MSG increased IRS-1 tyrosine phosphorylation (pIRS-1) by 96 % (MSG, 17.02 ± 0.6; control, 8.7 ± 0.2 a.u.), exercise training also increased it in both groups (control, 13.6 ± 0.1; MSG, 22.2 ± 1.1 a.u.). Current research shows that the practice of swim training increases the tyrosine phosphorylation of IRS-1 which can modulate the effect caused by obesity in insulin receptors.


Asunto(s)
Islotes Pancreáticos/metabolismo , Obesidad/metabolismo , Condicionamiento Físico Animal/fisiología , Receptor de Insulina/metabolismo , Natación/fisiología , Animales , Glucemia/metabolismo , Glucosa/farmacología , Insulina/metabolismo , Resistencia a la Insulina , Islotes Pancreáticos/efectos de los fármacos , Masculino , Ratones , Obesidad/inducido químicamente , Fosforilación/efectos de los fármacos , Glutamato de Sodio
16.
PLoS One ; 7(2): e30685, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22383969

RESUMEN

Pancreatic islets from adult rats whose mothers were protein restricted during lactation undersecrete insulin. The current work analyzes whether this secretory dysfunction can be improved when the pancreatic islets are grafted into hyperglycemic diabetic rats. Two groups of rats were used: the adult offspring from dams that received a low protein diet (4%) during the initial 2/3 of lactation (LP) and, as a control, the adult offspring from dams that consumed a normal protein diet (23%) during the entire period of lactation (NP). Islets from NP- and LP-rats were transplanted into diabetic recipient rats, which were generated by streptozotocin treatment. The islets were transplanted via the portal vein under anesthesia. The fed blood glucose levels were monitored during the 4 days post-transplantation. Transplanted islets from LP-rats (T LP) decreased the fed glucose levels of diabetic rats 34% (21.37 ± 0.24 mM, p<0.05); however, the levels still remained 2-fold higher than those of the sham-operated controls (6.88 ± 0.39 mM, p<0.05). Grafts with NP-islets (T NP) produced the same effect as the LP-islets in diabetic rats. The high fasting blood glucose levels of diabetic rats were improved by the transplantations. Islet grafts from both rat groups recovered 50% of the retroperitoneal fat mass of the diabetic rats (0.55 ± 0.08 g/100 g of body weight for T NP and 0.56 ± 0.07 g/100 g of body weight for T LP, p<0.05). Because pancreatic islets from both the NP- and LP-rats were able to regulate fasting blood glucose concentrations in hyperglycemic rats, we propose that the altered function of pancreatic islets from LP-rats is not permanent.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Insulina/metabolismo , Trasplante de Islotes Pancreáticos/métodos , Islotes Pancreáticos/metabolismo , Animales , Peso Corporal , Diabetes Mellitus Experimental/terapia , Dieta con Restricción de Proteínas , Femenino , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Homeostasis , Humanos , Exposición Materna , Modelos Biológicos , Embarazo , Preñez , Ratas , Ratas Wistar , Estreptozocina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...