Physiology - Theses

Permanent URI for this collection

Search Results

Now showing 1 - 4 of 4
  • Item
    Thumbnail Image
    Exercise and adipose tissue GLUT4
    Flores Opazo, Marcelo Alejandro ( 2017)
    GLUT4 is the major insulin-sensitive glucose transporter expressed predominantly in skeletal & cardiac muscles, and adipose tissue (AT) and mediates insulin-stimulated glucose transport into these tissues. Due to the major role of skeletal muscle in glucose disposal, considerable attention has focused on this tissue in order to understand how glucose homeostasis is affected by changes in muscle glucose metabolism. However, AT insulin-mediated glucose uptake and GLUT4 expression have been shown to be of importance for both glucose homeostasis and whole body insulin action. Interestingly, reduced GLUT4 expression in AT is a common defect found in insulin resistant states, including obesity, metabolic syndrome, and diabetes, whereas muscle GLUT4 expression is unaltered. Exercise increases AT-GLUT4 expression in rodents, and our lab has previously shown that 4 weeks of exercise training normalizes the expression of GLUT4 in the AT of patients with T2DM. However, the mechanisms involved in exercise-induced up-regulation of AT-GLUT4 are unknown. The broad aim of my research project was to examine the effect of exercise on AT-GLUT4 expression. With this purpose, we conducted three interventions using mice, human primary adipocytes and human subjects, in an attempt to gain a better understanding of: 1) how a high fat diet (HFD) affects the expression of AT-GLUT4, 2) how exercise may be beneficial to prevent this effect; and 3) how this effect may occur. Results showed that AT-GLUT4 protein is rapidly reduced by a HFD, suggesting this could be an early defect contributing to HFD-induced insulin resistance; exercise training increases GLUT4 protein in HFD-fed mice, short-term (10 d) exercise training did not affect GLUT4 levels in human, subcutaneous AT; and serum from exercised subjects increased GLUT4 protein and mRNA in human primary adipocytes suggesting that circulating factor(s) may mediate exercise effects on AT GLUT4 expression. Adipose tissue (AT) glucose transporter GLUT4 is reduced in insulin resistance (IR). Exercise training (EX) (4 weeks) normalized AT-GLUT4 expression in diabetic patients. However, mechanisms involved are unknown. Results showed: 1) AT-GLUT4 is reduced by high fat diet (HFD), which may contributes IR; 2) EX increased GLUT in HFD-fed mice, 3) short-term (10 d) EX didn’t affect GLUT4 in human AT; and 4) exercise serum increased GLUT4 in human primary adipocytes, suggesting circulating factor(s) may mediate EX-effects on AT GLUT4.
  • Item
    Thumbnail Image
    Characterization of mechanisms of myocardial remodeling in genetic models of cardiac hypertrophy
    Domenighetti, Andrea A. ( 2005-12)
    Cardiac hypertrophy is clinically defined as a relative increase in heart size associated with a thickening of the ventricular wall. It is a common feature of individuals suffering from different cardio-vascular or metabolic conditions and leads to heart failure. The structural, functional and molecular mechanisms which induce hypertrophy independent of hemodynamic alterations are poorly characterized. In this study, questions about whether cardiac-specific neuro-endocrine activation or metabolic imbalance are sufficient to induce hypertrophic structural and functional remodeling are addressed using genetically manipulated mouse models of primary cardiac hypertrophy. (For complete abstract open document)
  • Item
    Thumbnail Image
    Effect of fasting on adipose tissue glycogen levels in the obese Zucker rat
    Flores Opazo, Marcelo Alejandro ( 2012)
    Current scientific investigations have shown that obesity is associated with several alterations affecting not only the metabolic state of adipose tissue, since it also interferes with functional outcomes in target organs including liver and skeletal muscle. Metabolic alterations linked with obesity in the adipose tissue include early development of insulin resistance, impaired glucose transport and increased adipogenesis. In this regards, glucose uptake is the rate limiting step for glucose metabolism in adipose tissue. Insulin-mediated glucose uptake provides the cells not only with glucose as an energy substrate towards glycolysis, but also participates in de novo synthesis of fatty acids, fatty acid re-esterification, and a small amount of the incoming glucose is stored as glycogen. Glycogen is the mechanism by which cells store and mobilize glucose to meet their energetic and synthetic demands. During feeding, high blood glucose and plasma insulin command glycogen synthesis. Conversely, glycogen degradation during fasting requires sequential inactivation of glycogen synthase and activation of glycogen phosphorylase – the glycogen-metabolising enzymes – both actions orchestrated by the upstream kinase PKA. Little is known about the regulatory mechanisms controlling glycogen metabolism in the adipose tissue and how it is affected by pathological states related to obesity. The aim of the project was to examine adipose glycogen metabolism and the effects of fasting on glycogen-metabolising enzymes in order to determine whether adipose glycogen metabolism is altered as consequence of the obesity-induced insulin resistance. It was hypothesised that obesity imposes a blunted response to fasting that is reflected by impaired glycogen mobilization. Data obtained in this study showed that lean Zucker rats activated the lipolytic pathway in response to fasting, and NEFA were released into circulation. In contrast, fed and fasted plasma NEFA and glycerol levels were elevated several fold in obese rats, although with a reduced response to fasting. Considering that lipolysis occurs in parallel with glycogen degradation, we then tested the hypothesis that the blunted fasting response was associated with altered glycogen metabolism. This study showed for the first time that glycogen is not mobilized by 24-h fasting in the adipose tissue of obese Zucker rats. It could be initially attributed to an impaired degradation due to the lack of GP activation, although the finding of an impaired GS inactivation could similarly contribute to higher glycogen content following fasting. Additionally, obesity induced a differential effect on the protein content of GS and GP; increasing GS whereas reducing GP proteins expression. Following fasting, protein contents of both enzymes were reduced which could be interpreted as an attempt to counterbalance the unaltered glycogen content during fasting. Accordingly, it can be argued that obesity in Zucker rats may be associated with a higher content of glycogen after 24-h of fasting mainly caused by obesity-induced dysregulation upon GS and GP protein expression, with a lack of GP activation and defective inactivation of GS. These latter findings led us questioning about the involvement of upstream steps into the activation and inactivation of glycogen-metabolising enzymes. Therefore we next examined the activity of the upstream kinase PKA. The PKA activity was increased by fasting in lean rats. However, PKA activity was reduced in obese fasted rats, which was associated with the observed high adipose tissue glycogen levels found among fasted animals. In order to confirm this finding, we tested the activation of CREB, the downstream effector of PKA signalling. CREB phosphorylation was increased 3-fold by fasting in the lean group in parallel with increase in PKA activity. Interestingly, a similar level of phosphorylation was found in obese fed rats when compared with lean fed rats, which implies basal activation of this transcription factor. This over-activated CREB could be explained considering the role of insulin in the activation of CREB. On the other hand, after 24-h fasting, the pCREB/totalCREB ratio was significantly reduced in the obese rats which means that CREB is less active during fasting in obese Zucker rats. Taking into consideration the repressive effect exerted by CREB on the expression of GLUT4, we analysed GLUT4 content in the adipose tissue. As anticipated, fasting reduced GLUT4 protein content in the adipose tissue of lean animals. In the obese rats, GLUT4 protein level was markedly reduced to one third of lean fed content, whereas fasting did not modify GLUT4 protein content in this group, which led us to consider that GLUT4 protein content in the obese was decreased secondary to obesity and that fasting did not induce any further repressive effect within the time course of 24-h of food deprivation. In sum, these novel findings have shed light on how obesity may alter adipose tissue glycogen metabolism impairing its degradation during fasting. Obesity impairs cAMP-PKA signalling leading to dysregulation of glycogen-metabolising enzymes. Additionally, the obesity-induced repressive effect on the protein contents of GLUT4 and GP in association with increased protein level of GS might be related to alterations in carbohydrate metabolism in the adipose tissue. Further analyses are required in order to investigate other potential mechanisms involved in the regulation of glycogen metabolism, impaired cAMP-PKA signalling and GLUT4 expression with obesity.
  • Item
    Thumbnail Image
    Exercise and GLUT4 expression in type 2 diabetes
    Hussey, Sophie Elizabeth ( 2010)
    Peripheral insulin resistance is characterised by reduced insulin-stimulated glucose uptake in skeletal muscle and adipose tissue, and the condition represents one of the earliest hallmarks in the development of type 2 diabetes (T2D). In patients with T2D, protein expression of the insulin-stimulated glucose transporter, GLUT4, is reduced in adipose tissue, but preserved in skeletal muscle. Transgenic studies in rodents provide evidence that overexpression of GLUT4 selectively in either skeletal muscle or adipose tissue enhances whole-body insulin action. Since skeletal muscle accounts for the majority of insulin-stimulated glucose disposal, the effect of adipose tissue GLUT4 on insulin sensitivity is thought to be secondary to an altered secretion of adipokines which affect insulin action in muscle, in the context of a ‘metabolic crosstalk’ between insulin sensitive tissues. Increasing GLUT4 expression in skeletal muscle and adipose tissue could be an effective therapy in the treatment of insulin resistance and T2D. Exercise training increases GLUT4 protein expression in skeletal muscle of patients with T2D. This adaptation occurs in the face of enhanced insulin sensitivity, and results from the cumulative and transient increase in GLUT4 mRNA following each acute exercise bout. Less is known regarding the regulation of skeletal muscle GLUT4 expression by a single bout of exercise in patients with T2D, or the effect of exercise training on GLUT4 expression in adipose tissue. The primary aim of the studies undertaken for this thesis was to enhance understanding of exercise-mediated GLUT4 expression in skeletal muscle and adipose tissue of patients with T2D. The first investigation determined the effect of a single bout of exercise on skeletal muscle GLUT4 mRNA, and the signalling pathways which regulate GLUT4 expression, in patients with T2D and healthy control volunteers, matched for age and BMI. Increased (p<0.05) expression of GLUT4 and PGC-1α mRNA, together with increased (p<0.05) phosphorylation of AMPK and p38 MAPK was observed following exercise in patients with T2D, to a similar extent as in age- and BMI-matched control subjects. These findings lead to the conclusion that exercise-mediated regulation of GLUT4 expression is normal in patients with T2D. The second investigation of this thesis sought to identify the effect of a 4 week exercise training program on skeletal muscle and adipose tissue GLUT4 expression in patients with T2D. It was found that exercise training increased (p<0.05) GLUT4 protein expression by ~36% and ~20% in adipose tissue and skeletal muscle, respectively. These adaptations occurred in the absence of changes in insulin sensitivity or plasma levels of adipokines, adiponectin and resistin. Accordingly, the third study of this thesis sought to identify novel adipokines that regulate peripheral glucose metabolism in an adipocyte model of GLUT4 overexpression. Amyloid precursor protein (APP) was reduced (p<0.05) in culture media of GLUT4 overexpressing adipocytes, and the APP cleavage product, amyloid-beta (Aβ), reduced (p<0.05) insulin-stimulated Akt phosphorylation in L6 myocytes in vitro. These observations lead to the conclusion that increased adipose tissue GLUT4 expression may influence whole body glucose metabolism through reduced levels of Aβ. The primary aim of the final study undertaken was to identify novel changes in the abundance of proteins in skeletal muscle following exercise training in patients with T2D, including proteins of glucose metabolism, which may regulate of GLUT4 expression. Exercise training altered the abundance of several proteins involved in energy metabolism, as well as some novel proteins which may play a role in cytoskeleton interactions with mitochondria. In summary, this thesis demonstrated that skeletal muscle from patients with T2D responds normally to an acute exercise bout in terms of increased GLUT4 mRNA expression. In addition, it was shown that exercise training increased GLUT4 protein expression, not only in skeletal muscle, but also in adipose tissue of patients with T2D. This is significant because adipose tissue GLUT4 overexpression enhances insulin sensitivity. Data from this thesis suggest that improvements in insulin sensitivity may be secondary to altered secretion of Aβ from adipose tissue. Collectively, the findings provide a number of therapeutic targets for the treatment of insulin resistance and T2D.