Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Exp Physiol ; 100(1): 57-68, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25398717

RESUMEN

NEW FINDINGS: What is the central question of this study? Different nerve contributes periods of life are known for their differential sensitivity to interventions, and increased parasympathetic activity affects the development and maintenance of obesity. Thus, we evaluated the involvement of the vagus nerve by performing a vagotomy in young or adult rats that were offered an obesogenic high-fat diet. What is the main finding and its importance? Although the accumulation of adipose tissue decreased in both younger and older groups, the younger rats showed a greater response to the effects of vagotomy in general. In addition to the important role of the parasympathetic activity, we suggest that the vagus nerve contributes to the condition of obesity. Obesity has become a global problem, and this condition develops primarily because of an imbalance between energy intake and expenditure. The high complexity involved in the regulation of energy metabolism results from several factors besides endocrine factors. It has been suggested that obesity could be caused by an imbalance in the autonomous nervous system, which could lead to a condition of high parasympathetic activity in counterpart to low sympathetic tonus. High-fat (HF) diets have been used to induce obesity in experimental animals, and their use in animals leads to insulin resistance, hyperinsulinaemia and high parasympathetic activity, among other disorders. The aim of this work was to evaluate the effects of a vagotomy performed at the initiation of a HF diet at two different stages of life, weaning and adulthood. The vagotomy reduced parasympathetic activity (-32 and -51% in normal fat-fed rats and -43 and -55% in HF diet-fed rats; P < 0.05) and fat depots (-17 and -33%, only in HF diet-fed rats; P < 0.05). High-fat diet-fed rats exhibited fasting hyperinsulinaemia (fivefold higher in young rats and threefold higher in older rats; P < 0.05); however, vagotomy corrected it in younger rats only, and a similar effect was also observed during the glucose tolerance test. The insulin resistance exhibited by the HF diet-fed groups was not altered in the vagotomized rats. We suggest that the vagus nerve, in addition to the important role of parasympathetic activity, contributes to the condition of obesity, and that non-vagal pathways may be involved along with the imbalanced autonomic nervous system.


Asunto(s)
Dieta Alta en Grasa , Síndrome Metabólico/etiología , Obesidad/etiología , Nervio Vago/fisiopatología , Adiposidad , Factores de Edad , Animales , Biomarcadores/sangre , Glucemia/metabolismo , Modelos Animales de Enfermedad , Insulina/sangre , Resistencia a la Insulina , Masculino , Síndrome Metabólico/sangre , Síndrome Metabólico/fisiopatología , Síndrome Metabólico/prevención & control , Obesidad/sangre , Obesidad/fisiopatología , Obesidad/prevención & control , Ratas Wistar , Factores de Tiempo , Vagotomía , Nervio Vago/cirugía , Destete , Aumento de Peso
2.
Cell Physiol Biochem ; 33(4): 1075-86, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24732778

RESUMEN

BACKGROUND/AIMS: Impaired pancreatic beta cell function and insulin secretion/action are a link between obesity and type 2 diabetes, which are worldwide public health burdens. We aimed to characterize the muscarinic acetylcholine receptor (mAChR) M1-M4 subtypes in isolated pancreatic islets from pre-diabetic obese rats that had been treated neonatally with monosodium L-glutamate (MSG). METHODS: At 90 days of age, both the MSG and the control groups underwent biometric and biochemical evaluation. Anti-muscarinic drugs were used to study mAChR function either in vivo or in vitro. RESULTS: The results demonstrated that atropine treatment reduced insulin secretion in the MSG-treated and control groups, whereas treatment with an M2mAChR-selective antagonist increased secretion. Moreover, the insulinostatic effect of an M3mAChR-selective antagonist was significantly higher in the MSG-treated group. M1mAChR and M3mAChR expression was increased in the MSG-obese group by 55% and 73%, respectively. In contrast, M2mAChR expression decreased by 25% in the MSG group, whereas M4mAChR expression was unchanged. CONCLUSIONS: Functional changes in and altered content of the mAChR (M1-M4) subtypes are pivotal to the demand for high pancreatic beta cell insulin secretion in MSG-obese rats, which is directly associated with vagal hyperactivity and peripheral insulin resistance.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Obesidad/metabolismo , Receptores Muscarínicos/metabolismo , Glutamato de Sodio/farmacología , Animales , Glucemia/análisis , Prueba de Tolerancia a la Glucosa , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Masculino , Agonistas Muscarínicos/farmacología , Antagonistas Muscarínicos/farmacología , Obesidad/patología , Ratas , Ratas Wistar , Receptor Muscarínico M1/metabolismo , Receptor Muscarínico M2/metabolismo , Receptor Muscarínico M3/metabolismo , Receptor Muscarínico M4/metabolismo , Receptores Muscarínicos/química
3.
Cell Physiol Biochem ; 34(6): 1920-32, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25500480

RESUMEN

BACKGROUND/AIMS: The objective of the current work was to test the effect of metformin on the tumor growth in rats with metabolic syndrome. METHODS: We obtained pre-diabetic hyperinsulinemic rats by neonatal treatment with monosodium L-glutamate (MSG), which were chronically treated every day, from weaning to 100 day old, with dose of metformin (250 mg/kg body weight). After the end of metformin treatment, the control and MSG rats, treated or untreated with metformin, were grafted with Walker 256 carcinoma cells. Tumor weight was evaluated 14 days after cancer cell inoculation. The blood insulin, glucose levels and glucose-induced insulin secretion were evaluated. RESULTS: Chronic metformin treatment improved the glycemic homeostasis in pre-diabetic MSG-rats, glucose intolerance, tissue insulin resistance, hyperinsulinemia and decreased the fat tissue accretion. Meanwhile, the metformin treatment did not interfere with the glucose insulinotropic effect on isolated pancreatic islets. Chronic treatment with metformin was able to decrease the Walker 256 tumor weight by 37% in control and MSG rats. The data demonstrated that the anticancer effect of metformin is not related to its role in correcting metabolism imbalances, such as hyperinsulinemia. However, in morphological assay to apoptosis, metformin treatment increased programmed cell death. CONCLUSION: Metformin may have a direct effect on cancer growth, and it may programs the rat organism to attenuate the growth of Walker 256 carcinoma.


Asunto(s)
Carcinoma 256 de Walker/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Metformina/administración & dosificación , Neoplasias/tratamiento farmacológico , Animales , Glucemia , Carcinoma 256 de Walker/metabolismo , Carcinoma 256 de Walker/patología , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/patología , Intolerancia a la Glucosa/tratamiento farmacológico , Intolerancia a la Glucosa/patología , Hipoglucemiantes/administración & dosificación , Insulina/metabolismo , Resistencia a la Insulina/genética , Islotes Pancreáticos/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Ratas , Glutamato de Sodio/toxicidad
4.
Br J Nutr ; 111(2): 227-35, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-23841989

RESUMEN

Impaired pancreatic ß-cell function, as observed in the cases of early nutrition disturbance, is a major hallmark of metabolic diseases arising in adulthood. In the present study, we aimed to investigate the function/composition of the muscarinic acetylcholine receptor (mAChR) subtypes, M2 and M3, in the pancreatic islets of adult offspring of rats that were protein malnourished during lactation. Neonates were nursed by mothers that were fed either a low-protein (4 %, LP) or a normal-protein (23 %, NP) diet. Adult rats were pre-treated with anti-muscarinic drugs and subjected to the glucose tolerance test; the function and protein expression levels of M2mAChR and M3mAChR were determined. The LP rats were lean and hypoinsulinaemic. The selective M2mAChR antagonist methoctramine increased insulinaemia by 31 % in the NP rats and 155 % in the LP rats, and insulin secretion was increased by 32 % in the islets of the NP rats and 88 % in those of the LP rats. The selective M3mAChR antagonist 4-diphenylacetoxy-N-methylpiperidine methiodide decreased insulinaemia by 63 % in the NP rats and 40 % in the LP rats and reduced insulin release by 41 % in the islets of the NP rats and 28 % in those of the LP rats. The protein expression levels of M2mAChR and M3mAChR were 57 % higher and 53 % lower, respectively, in the islets of the LP rats than in those of the NP rats. The expression and functional compositions of M2mAChR and M3mAChR were altered in the islets of the LP rats, as a result of metabolic programming caused by the protein-restricted diet, which might be another possible effect involved in the weak insulin secretion ability of the islets of the programmed adult rats.


Asunto(s)
Alimentación Animal/análisis , Proteínas en la Dieta/administración & dosificación , Células Secretoras de Insulina/fisiología , Lactancia/fisiología , Receptores Muscarínicos/clasificación , Receptores Muscarínicos/metabolismo , Fenómenos Fisiológicos Nutricionales de los Animales , Animales , Glucemia , Dieta/veterinaria , Femenino , Glucosa/metabolismo , Intolerancia a la Glucosa , Prueba de Tolerancia a la Glucosa , Homeostasis , Masculino , Fenómenos Fisiologicos Nutricionales Maternos , Antagonistas Muscarínicos/farmacología , Embarazo , Ratas , Ratas Wistar
5.
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
6.
Endocr Res ; 36(2): 83-90, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21539446

RESUMEN

Low-intensity swimming training, started at an early age, was undertaken to observe glycemic control in hypothalamic obese mice produced by neonatal monosodium l-glutamate (MSG) treatment. Although swimming exercises by weaning pups inhibited hypothalamic obesity onset and recovered sympathoadrenal axis activity, this event was not observed when exercise training is applied to young adult mice. However, the mechanisms producing this improved metabolism are still not fully understood. Current work verifies whether, besides reducing fat tissue accumulation, low-intensity swimming in MSG-weaned mice also improves glycemic control. Although MSG and control mice swam for 15 min/day, 3 days a week, from the weaning stage up to 90 days old, sedentary MSG and normal mice did not exercise at all. After 14 h of fasting, animals were killed at 90 days of age. Retroperitonial fat accumulation was measured to estimate obesity. Fasting blood glucose and insulin concentrations were also measured. Mice were also submitted to ipGTT. MSG obese mice showed fasting hyperglycemia, hyperinsulinemia, and glucose intolerance and insulin resistance. However, the exercise was able to block MSG treatment effects. Higher total cholesterol and triglycerides observed in MSG mice were normalized by exercise after weaning. Exercised MSG animals had higher HDLc than the sedentary group. Data suggest that early exercise training maintains normoglycemia, insulin tissue sensitivity, and normal lipid profile in mice programmed to develop metabolic syndrome.


Asunto(s)
Glucemia/análisis , Enfermedades Hipotalámicas/complicaciones , Lípidos/sangre , Obesidad/sangre , Condicionamiento Físico Animal , Natación , Animales , Animales Recién Nacidos , Ayuno , Femenino , Intolerancia a la Glucosa , Homeostasis , Enfermedades Hipotalámicas/inducido químicamente , Insulina/sangre , Resistencia a la Insulina , Masculino , Ratones , Obesidad/etiología , Glutamato de Sodio
7.
Eur J Pharmacol ; 556(1-3): 223-8, 2007 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-17174301

RESUMEN

Hypothalamic obese rats, obtained by neonatal treatment with monosodium L-glutamate (MSG), are hyperinsulinemic, and secrete more insulin than lean ones do when stimulated by glucose, while acetylcholine insulinotropic effect decreases. The effect of acetylcholine on glucose-induced insulin secretion is attributed to muscarinic receptors of pancreatic beta cells, mainly to M(3) subtype. However, it has been observed that activation of M(2) or M(4) subtypes causes inhibition of glucose-induced insulin secretion in insulin secreting cell line. Insulin secretion was measured, stimulated by glucose in the presence of acetylcholine plus methoctramine, a muscarinic M(2) antagonist, on pancreatic islets isolated from MSG-obese and lean rats to investigate whether impairment of acetylcholine insulinotropic effect on pancreatic islets from MSG-obese rats has any relationship with muscarinic M(2) receptor function in beta cells. Insulin secretion stimulated by 8.3 mM glucose was higher in islets from obese rats than from lean ones. Insulinotropic effect of acetylcholine was reported in islets of both animals, albeit less than in obese ones. Blockage of muscarinic M(2) receptor, using methoctramine at 1; 5 and 10 microM, increased acetylcholine secretory response in islets of obese rats, while no effect has been observed in lean ones. Results demonstrate that muscarinic M(2) receptors are functioning in pancreatic islets of MSG-obese rats. The inhibitory action of muscarinic M(2) receptor may be a mechanism by which acetylcholine discloses weak insulinotropic effect in MSG-obese rats.


Asunto(s)
Hipotálamo/fisiopatología , Islotes Pancreáticos/fisiopatología , Obesidad/fisiopatología , Receptor Muscarínico M2/metabolismo , Acetilcolina/farmacología , Animales , Animales Recién Nacidos , Diaminas/farmacología , Prueba de Tolerancia a la Glucosa , Técnicas In Vitro , Resistencia a la Insulina , Masculino , Neostigmina/farmacología , Obesidad/inducido químicamente , Ratas , Ratas Wistar , Receptor Muscarínico M2/antagonistas & inhibidores , Glutamato de Sodio
8.
J Endocrinol ; 221(2): 285-95, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24599936

RESUMEN

Nutritional insults during developmental plasticity have been linked with metabolic diseases such as diabetes in adulthood. We aimed to investigate whether a low-protein (LP) diet at the beginning of adulthood is able to program metabolic disruptions in rats. While control rats ate a normal-protein (23%; NP group) diet, treated rats were fed a LP (4%; LP group) diet from 60 to 90 days of age, after which an NP diet was supplied until they were 150 days old. Plasma levels of glucose and insulin, autonomous nervous system (ANS), and pancreatic islet function were then evaluated. Compared with the NP group, LP rats exhibited unchanged body weight and reduced food intake throughout the period of protein restriction; however, after the switch to the NP diet, hyperphagia of 10% (P<0.05), and catch-up growth of 113% (P<0.0001) were found. The LP rats showed hyperglycemia, insulin resistance, and higher fat accretion than the NP rats. While the sympathetic tonus from LP rats reduced by 28%, the vagus tonus increased by 21% (P<0.05). Compared with the islets from NP rats, the glucose insulinotropic effect as well as cholinergic and adrenergic actions was unaltered in the islets from LP rats. Protein restriction at the beginning of adulthood induced unbalanced ANS activity and fat tissue accretion later in life, even without functional disturbances in the pancreatic islets.


Asunto(s)
Sistema Nervioso Autónomo/fisiopatología , Dieta con Restricción de Proteínas/efectos adversos , Islotes Pancreáticos/inervación , Islotes Pancreáticos/metabolismo , Animales , Peso al Nacer/efectos de los fármacos , Peso al Nacer/fisiología , Glucemia/metabolismo , Células Cultivadas , Proteínas en la Dieta/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Factores de Tiempo
9.
J Endocrinol ; 216(2): 195-206, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23151360

RESUMEN

Similar to gestation/lactation, puberty is also a critical phase in which neuronal connections are still being produced and during which metabolic changes may occur if nutrition is disturbed. In the present study we aimed to determine whether peripubertal protein restriction induces metabolic programming. Thirty-day-old male rats were fed either a low protein (LP group) diet (4% w/w protein) or a normal protein (NP group) diet (23%) until 60 days of age, when they received the NP diet until they were 120 days old. Body weight (BW), food intake, fat tissue accumulation, glucose tolerance, and insulin secretion were evaluated. The nerve electrical activity was recorded to evaluate autonomous nervous system (ANS) function. Adolescent LP rats presented hypophagia and lower BW gain during the LP diet treatment (P<0.001). However, the food intake and BW gain by the LP rats were increased (P<0.001) after the NP diet was resumed. The LP rats presented mild hyperglycemia, hyperinsulinemia, severe hyperleptinemia upon fasting, peripheral insulin resistance and increased fat tissue accumulation and vagus nerve activity (P<0.05). Glucose-induced insulin secretion was greater in the LP islets than in the NP islets; however, the cholinergic response was decreased (P<0.05). Compared with the islets from the NP rats, the LP islets showed changes in the activity of muscarinic receptors (P<0.05); in addition, the inhibition of glucose-induced insulin secretion by epinephrine was attenuated (P<0.001). Protein restriction during adolescence caused high-fat tissue accumulation in adult rats. Islet dysfunction could be related to an ANS imbalance.


Asunto(s)
Dieta con Restricción de Proteínas/efectos adversos , Islotes Pancreáticos/metabolismo , Pubertad/metabolismo , Animales , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Glucosa/farmacología , Hiperglucemia/metabolismo , Hiperinsulinismo/metabolismo , Insulina/metabolismo , Masculino , Ratas
10.
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
11.
Nutr Metab (Lond) ; 9(1): 80, 2012 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-22967456

RESUMEN

Protein restriction during lactation has been used as a rat model of metabolic programming to study the impact of perinatal malnutrition on adult metabolism. In contrast to protein restriction during fetal life, protein restriction during lactation did not appear to cause either obesity or the hallmarks of metabolic syndrome, such as hyperinsulinemia, when individuals reached adulthood. However, protein restriction provokes body underweight and hypoinsulinemia. This review is focused on the regulation of insulin secretion and the influence of the autonomic nervous system (ANS) in adult rats that were protein-malnourished during lactation. The data available on the topic suggest that the perinatal phase of lactation, when insulted by protein deficit, imprints the adult metabolism and thereby alters the glycemic control. Although hypoinsulinemia programs adult rats to maintain normoglycemia, pancreatic ß-cells are less sensitive to secretion stimuli, such as glucose and cholinergic agents. These pancreatic dysfunctions may be attributed to an imbalance of ANS activity recorded in adult rats that experienced maternal protein restriction.

12.
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
13.
Exp Diabetes Res ; 2011: 947917, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21860615

RESUMEN

The involvement of sympathoadrenal axis activity in obesity onset was investigated using the experimental model of treating neonatal rats with monosodium L-glutamate. To access general sympathetic nervous system activity, we recorded the firing rates of sympathetic superior cervical ganglion nerves in animals. Catecholamine content and secretion from isolated adrenal medulla were measured. Intravenous glucose tolerance test was performed, and isolated pancreatic islets were stimulated with glucose and adrenergic agonists. The nerve firing rate of obese rats was decreased compared to the rate for lean rats. Basal catecholamine secretion decreased whereas catecholamine secretion induced by carbachol, elevated extracellular potassium, and caffeine in the isolated adrenal medulla were all increased in obese rats compared to control. Both glucose intolerance and hyperinsulinaemia were observed in obese rats. Adrenaline strongly inhibited glucose-induced insulin secretion in obese animals. These findings suggest that low sympathoadrenal activity contributes to impaired glycaemic control in prediabetic obese rats.


Asunto(s)
Médula Suprarrenal/fisiopatología , Insulina/metabolismo , Obesidad/fisiopatología , Estado Prediabético/fisiopatología , Sistema Nervioso Simpático/fisiopatología , Médula Suprarrenal/química , Médula Suprarrenal/efectos de los fármacos , Animales , Glucemia/análisis , Cafeína/farmacología , Carbacol/farmacología , Catecolaminas/análisis , Catecolaminas/metabolismo , Prueba de Tolerancia a la Glucosa , Insulina/sangre , Secreción de Insulina , Masculino , Obesidad/inducido químicamente , Ratas , Ratas Wistar , Glutamato de Sodio
14.
Endocrine ; 36(3): 510-7, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19856134

RESUMEN

Current research employed electrical records from superior vagus and sympathetic nerve branch that supply fat retroperitoneal tissue (RS nerve) to investigate whether very moderate swim training in obese-programmed mice would change sympathetic and parasympathetic autonomic nervous system activities. Neonatal mice were treated with monosodium L: -glutamate (MSG), during their first 5 days of life, to induce obesity. Mice started training on weaning, comprising free swimming 3 days/week, 15 min/day for 10 weeks. After 12 h fasting, the nerve electrical signals of the 90-day-old mice were processed to obtain firing rates. Blood samples were collected to measure glucose and insulin levels. Adrenal catecholamine content was measured. MSG treatment caused obesity. Hyperglycemia and hyperinsulinemia in MSG-obese mice, without any change in food intake, were obtained. Vagus firing rates were higher in obese mice than those in lean ones. A decrease in RS nerve activity and lower adrenal catecholamine stores have been observed. Swimming normalized blood glucose and insulin levels and MSG-obesity onset was attenuated by exercise. Vagus activity from obese mice decreased, whereas RS nerve activity and adrenal catecholamine levels increased in trained ones. Results suggest that autonomic activity imbalance and metabolic dysfunctions observed in MSG-obese mice were inhibited by precocious and moderate exercise training.


Asunto(s)
Sistema Nervioso Autónomo/fisiología , Glucosa/metabolismo , Obesidad/inducido químicamente , Obesidad/fisiopatología , Condicionamiento Físico Animal/fisiología , Glutamato de Sodio , Médula Suprarrenal/efectos de los fármacos , Médula Suprarrenal/metabolismo , Animales , Animales Recién Nacidos , Sistema Nervioso Autónomo/efectos de los fármacos , Catecolaminas/metabolismo , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Femenino , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Insulina/metabolismo , Masculino , Ratones , Ratones Obesos , Obesidad/metabolismo , Condicionamiento Físico Animal/métodos , Natación/fisiología
15.
Nutr Neurosci ; 10(1-2): 79-87, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17539486

RESUMEN

Involvement of autonomic nervous system (ANS) neurotransmitters on insulin secretion in rats submitted to protein malnutrition during lactation was studied. During the first 2/3 of lactation, mothers received a 4% protein diet (LP). Control group received normal diet (23% protein) (NP). After protein restriction, mothers received normal diets. At 81 days rats were submitted to intravenous glucose tolerance tests (ivGTT). Plasma glucose and insulin concentration (PIC) were measured. Glucose-induced insulin secretion (GIIS) was tested in pancreatic islets. Fasting normoglycemia and hypoinsulinemia were observed in LP rats. Glucose intolerance and low PIC in LP group were detected during ivGTT. Acetylcholine (Ach) or blockage of alpha-adrenoceptors induced high PIC increment in LP rats; atropine or stimulation of alpha-adrenoceptors did not change PIC. Insulin secretion of LP rat islets showed low glucose and carbachol responses. Epinephrine-inhibited GIIS in both islet groups. Hypoinsulinemia observed in lactation-malnourished rats might be caused by alterations in GIIS regulation, including ANS modulation.


Asunto(s)
Antioxidantes/metabolismo , Sistema Nervioso Autónomo/fisiología , Dieta con Restricción de Proteínas , Glucosa/farmacología , Insulina/metabolismo , Lactancia/fisiología , Animales , Sistema Nervioso Autónomo/efectos de los fármacos , Sistema Nervioso Autónomo/crecimiento & desarrollo , Tamaño Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Femenino , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Modelos Animales , Ratas , Ratas Wistar
16.
Endocrine ; 31(2): 142-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17873325

RESUMEN

Hypothalamic MSG-obese rats show hyperinsulinemia and tissue insulin resistance, and they display intense parasympathetic activity. Current analysis investigates whether early subdiaphragmatic vagotomy prevents tissue insulin sensitivity impairment in adult obese MSG-rats. Hypothalamic obesity was induced by MSG (4 mg/g BW), daily, from birth up to 5 days. Control animals receiving saline solution. On the 30th day rats underwent bilateral subdiaphragmatic vagotomy or sham surgery. An intravenous glucose tolerance test (i.v.GTT) was performed when rats turned 90 days old. Total white fat tissue (WAT) from rat carcass was extracted and isolated; the interscapular brown fat tissue (IBAT) was weighed. Rather than blocking obesity, vagotomy reduced WAT and IBAT in MSG-obese rats when the latter were compared to sham MSG-rats. High blood fasting insulin and normal glucose levels were also observed in MSG-obese rats. Although glucose intolerance, high insulin secretion, and significant insulin resistance were recorded, vagotomy improved fasting insulinemia, glucose tolerance and insulin tissue sensitivity in MSG-obese rats. Results suggest that increased fat accumulation is caused, at least in part, by high blood insulin concentration, and enhanced parasympathetic activity on MSG-obese rats.


Asunto(s)
Tejido Adiposo/metabolismo , Enfermedades Hipotalámicas/metabolismo , Insulina/metabolismo , Obesidad/metabolismo , Nervio Vago/fisiopatología , Adiposidad/fisiología , Animales , Animales Recién Nacidos , Intolerancia a la Glucosa/etiología , Prueba de Tolerancia a la Glucosa , Crecimiento y Desarrollo/efectos de los fármacos , Enfermedades Hipotalámicas/inducido químicamente , Enfermedades Hipotalámicas/fisiopatología , Resistencia a la Insulina , Secreción de Insulina , Masculino , Obesidad/inducido químicamente , Obesidad/complicaciones , Obesidad/fisiopatología , Ratas , Ratas Wistar , Glutamato de Sodio , Vagotomía
17.
Endocrine ; 29(3): 445-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16943583

RESUMEN

Hyperinsulinemia in obesity has been attributed to insulin oversecretion by pancreatic beta-cells. Beta-cells are equipped with cholinergic and adrenergic receptors; whereas overall acetylcholine action is to potentiate, catecholamines' effect is to inhibit glucose-induced insulin release (GIIR) via alpha2-adrenoceptor. However, it has been shown that beta-adrenergic agonists potentiate glucose response. GIIR was studied in pancreatic islets from hyperinsulinemic adult obese rats, obtained by L-glutamate monosodium (MSG) neonatal treatment. Islets from MSG-rats were more glucose responsive than control ones. Isoproterenol, a beta-adrenergic agonist, inhibited the GIIR in islets from MSG-obese rats. Results indicate that MSG treatment causes alteration on function of beta-cell adrenoceptors.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Isoproterenol/farmacología , Obesidad/metabolismo , Animales , Animales Recién Nacidos , Glucemia/análisis , Hiperinsulinismo/inducido químicamente , Técnicas In Vitro , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Masculino , Obesidad/inducido químicamente , Ratas , Ratas Wistar , Glutamato de Sodio/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA