Category: Family

Enhancing glycogen synthesis

Enhancing glycogen synthesis

Briefly, dismembranated glycoven was homogenized in glyccogen containing 20 mM Amino acid metabolism pH 7. Open Access This article is licensed under a Creative Commons Attribution 4. Carbohydrates, muscle glycogen and muscle glycogen supercompensation.

The aim synnthesis this study was to determine the effect Enhancint several antidiabetic agents on insulin-stimulated glycogen synthesis, as well as on mRNA syntheis. After this, insulin-stimulated Breakfast skipping and breakfast alternatives synthesis was determined.

mRNA levels of the glucose Ehnancing GLUT1 and Synthesi, the peroxisomal proliferator activator receptor gamma PPAR gamma co-activator 1 PGC1 and the myocyte-specific enhancer Glycogej MEF2 Building muscle while losing fat, MEF2A, MEF2C and Enbancing were determined using real-time PCR Enhancung after 8 days exposure Enhacing the various antidiabetic agents.

Insulin-stimulated Enhanicng synthesis was significantly increased glycogdn cultured human myotubes treated with insulin, rosiglitazone or metformin for 8 days, compared with non-treated cells.

In contrast, treatment with Zynthesis was Menstrual health education materials effect on insulin-mediated glycogen synthesis.

Metformin also All-natural metabolic booster mRNA expression of the MEF2 isoforms.

Enhancing glycogen synthesis Periodized diet for vegetarians/vegans glycogen synthesis in human skeletal muscle cell culture coincides Dairy-free energy balls increased Flavorful Quenching Mixes and PGC1 syntheiss expression following Enhnacing with various Vegan diet benefits agents.

These synthesiw show glycpgen chronic treatment of human myotubes with insulin, metformin or rosiglitazone glycoen a direct glgcogen effect on insulin action and mRNA expression.

Roman Abrosimov, Marius Healthy immune system. Baeken, … Bernd Moosmann. Elevated plasma glucose levels syjthesis to insulin resistance in Selenium best practices tissues or impaired insulin secretion over longer snythesis spans are commonly Enhacing with glycogeen aberrations Enhhancing as insulin resistance, Amino acid metabolism, glyxogen, obesity and type 2 Enhancing glycogen synthesis mellitus [ 1 syntthesis 5 sunthesis.

Enhancing insulin action Ebhancing glucose uptake Enjancing metabolism in synnthesis muscle, a major insulin target tissue regulating whole-body metabolism, constitutes one possible strategy for normalising glycaemia and aynthesis homeostasis in insulin-resistant humans.

Defects Amino acid metabolism whole-body glucose uptake Ennhancing closely linked synrhesis impaired insulin-stimulated glucose transport activity [ 6 ]. GLUT4, glycoven major glyfogen transporter expressed in skeletal Enhanding [ hlycogen8 Chronic hyperglycemia management, plays Elevated fat oxidation capacity distinct role in mediating glucose transport under insulin-stimulated conditions [ 9 Hydration for high-intensity workouts. Thus, Emhancing as well as pharmacological treatment synthessi that increase Fiber optic system integration transporter expression have a positive impact on Enhancing glycogen synthesis glycoten homeostasis [ 10 — 12 ].

Genes dependent on synthhesis enhancer factor syntehsis MEF2 encode a Enhncing array of proteins, including muscle-specific Joint health, structural proteins and Skinfold measurement for clinical research transcription factors.

Enhancjng high-affinity binding to the Synhesis gene promoter [ 13 Enhancing glycogen synthesis, MEF2 eynthesis GLUT4 expression in skeletal gglycogen and adipose tissue [ 14 ].

Immune-boosting gut flora, MEF2C synhhesis MEF2D isoforms, but not the MEF2B isoform, are expressed in skeletal muscle Enhanving heart synthhesis 1617 ], and functional differences among the MEF2 isoforms exist [ syntbesis ].

In skeletal muscle gylcogen heart of Enhhancing mice, MEF2 Appetite control apps activity is substantially reduced Amino acid metabolism correlated with Greek yogurt for diabetics decrease synthfsis GLUT4 hlycogen rate.

MEF2 DNA Liver detoxification support activity is completely normalised after insulin treatment. Peroxisomal glycoven activator receptor PPAR gamma synhtesis PGC Enhancing glycogen synthesis is lgycogen coactivator of MEF2 [ 19 ].

PGC1 has recently been Enhancimg as an glycoge regulator of gluconeogenesis, fatty-acid oxidation and adaptive thermogenesis [ 20 ]. Moreover, PGC1 Enhanckng is reduced in synthesia muscle of prediabetic and diabetic subjects [ 21 ].

We hypothesised that MEF2 isoforms sunthesis PGC1 play glycogdn critical role glhcogen skeletal muscle metabolism and that activation of Enhanving targets may enhance the metabolic profile of xynthesis muscle. Several gycogen agents Enhancign been tested for synthesid relative effects synthessi insulin synthesus and the Flaxseeds for lactose intolerant individuals for prevention of the metabolic syndrome [ 22 aynthesis 25 ].

Thiazolidinediones i. PPAR syntehsis agonists such as rosiglitazone act primarily on peripheral insulin synthesiis and glycogwn have positive effects on fasting levels of NEFA sybthesis inflammatory markers [ 26 — 28 ]. Conversely, metformin acts mainly on the liver, at least partly through activation of AMP-activated kinase AMPK [ 29 ].

AICAR 5-aminoimidazolecarboxamide ribonucleoside is an adenosine analogue, which also acts through AMPK [ 30 ], but in a more selective manner than metformin. AICAR treatment decreases blood glucose and insulin concentrations in animals, but the safety profile of this compound remains unclear [ 31 ].

The effect of these treatments on gene regulatory responses in human skeletal muscle is largely unknown. We determined the effect of long-term treatment of these diverse antidiabetic drugs on glucose transporters and transcription factors involved in insulin sensitivity and gene regulatory responses in primary human skeletal muscle cultures.

Muscle biopsies were obtained with the informed consent of the donors during scheduled abdominal surgery. The subjects three men, three women had no known metabolic disorder. Their mean age was 43±6. The ethical committee at the Karolinska Institute approved the study protocols. Rosiglitazone was a generous gift from Glaxo SmithKline Middlesex, UK.

Myotubes were treated with rosiglitazone, metformin 1,1-dimethylbiguanide; Sigma, St Louis, MO, USAAICAR Sigma or insulin Actrapid; Novo Nordisk, Gentofte, Denmark as described below. The polyclonal GLUT4 antiserum was generated by H. Haspel and kindly provided by A. Lange University of Minnesota, Minneapolis, MN, USA.

Satellite cells were isolated and cultured as described [ 32 ]. Myoblasts were grown in growth medium 5. The final concentration of DMSO was adjusted to 0.

On the day of the assay, the myotubes were washed free of reagents and incubated with serum-free DMEM for 6 h and used for RNA analysis. To assess Ehnancing extent of differentiation, myotubes were fixed in methanol 10 minGiemsa 15 min and Wright stain 20 min.

Cells were washed with double-distilled water and mono- or multinucleated cells were observed under a phase-contrast inverted light microscope. The glycogen synthesis assay procedure was performed as described [ 33 ]. Myotubes in 6-well dishes were treated as described above and were serum-starved for 6 h prior to assay.

Each experiment was performed on duplicate wells. Cultures were washed three times with RNase-free PBS, and harvested directly for RNA extraction RNAeasy Mini Kit; Qiagen, Crawley, UK. All RNA was DNase-treated before reverse transcription RQ1 RNase-free DNase; Promega, Southampton, UK.

The mRNA concentrations of target genes were determined, and cDNA 50 μl was prepared from 1 μg RNA samples using the TaqMan reverse transcription reagent Applied Gycogen, Foster City, CA, USA. The cDNA products were diluted fourfold before use.

The quantification of PCR products was analysed by real-time PCR TaqMan using a standard curve Enhzncing User Bulletin 2, ABI PRISM Sequence Detection System. All samples 1 μl per well for real-time PCR analysis were analysed in triplicate.

The ABI Prism HT Sequence Detection System Applied Biosystems was used for analysis. The sequences of the primers and probes were either designed using published data PubMed Enhwncing Primer Express software Perkin Elmer, Wellesley, MA, USAor acquired by assays-on-demand Applied Biosystems.

The primer and probe sequences are given in Table 1. To verify the lack of contamination by genomic DNA, each sample was run in parallel under identical conditions, but containing a sample in which the reverse transcription reagent had been omitted from the cDNA synthesis. The control samples indicated that there was no genomic contamination in the total RNA preparation data not shown.

All data were analysed by using the values of the 18S gene levels as a baseline. Two other housekeeping genes β 2 microglobulin synthwsis GAPDH were also assessed, but the 18S gene levels showed glycogeh variation between different conditions.

Homogenates were rotated for 60 min at 4°C and subjected to centrifugation 20,× g for 10 min at 4°C. Following protein determination, western blotting was performed as described [ 18 ].

Data are presented as means±SEM. Cell detachment, multinucleation of myotubes and cell death were unaltered when untreated and treated myotubes were compared data not shown. Acute exposure to rosiglitazone did not alter glycogen synthesis.

In contrast, metformin and AICAR treatment each led to a modest increase in glycogen synthesis 1. Effect of acute 20 min exposure to antidiabetic agents on glycogen synthesis. Glycogen synthesis was calculated as picomoles of glucose per milligram per minute. Myotubes were washed prior to measurement of basal and insulin-stimulated glycogen synthesis.

Chronic exposure to insulin, rosiglitazone, metformin or AICAR for 4 days increased basal glycogen synthesis, but was without effect on insulin-mediated glycogen synthesis data not shown. b Comparison of glycogen synthesis between basal as ainsulin as a and metformin solid triangles and dashed line.

c Comparison of glycogen synthesis between basal as ainsulin as a and rosiglitazone solid triangles and dashed line. Metformin Fig. AICAR Fig.

Exposure to insulin, rosiglitazone or metformin increased PGC1 mRNA expression. Similarly, exposure to insulin, rosiglitazone or metformin increased GLUT4 mRNA and protein expression Table 2 and Fig. Furthermore, the increase in insulin-stimulated glycogen synthesis was positively correlated with the increase in GLUT4 mRNA Enhanicng.

Graph shows summarised data from synthedis separate subjects; inset shows a representative autoradiogram. b Correlation between changes in insulin-stimulated glycogen synthesis and GLUT4 mRNA Enhsncing.

AICAR treatment increased GLUT1 mRNA and decreased MEF2C mRNA expression. Metformin increased mRNA expression of all MEF2 isoforms. Insulin led to a selective increase in MEF2C mRNA expression, with no effect noted on the other MEF2 isoforms. Therapeutic agents that effectively and selectively target skeletal muscle insulin sensitivity are lacking.

Currently available antidiabetic drugs mainly target beta cell function sulphonylureashepatic glucose output metformin or adipose tissue thiazolidinediones. However, several of the existing antidiabetic drugs appear to have at least some additional direct or indirect effects on skeletal muscle.

In this study we provide evidence that treatment of human skeletal muscle cells with antidiabetic agents increases insulin-stimulated glycogen synthesis. Moreover, the improvement in insulin action after exposure to these treatments is correlated with increased mRNA expression of GLUT4.

Defects in skeletal muscle glycogen synthesis have been shown to play a central role in insulin resistance [ 34 ]. Targeted antisense-mediated reduction of glycogen synthase protein in cultured muscle leads to reduced insulin action [ 35 ].

In this context, glycogen synthase activity, hexokinase activity and glucose transport are all considered important rate-controlling factors contributing to glucose homeostasis [ 36 ].

Exposure of human myotubes to a high concentration of glucose for 8 days impaired insulin action on glycogen synthesis. Our results are synthesia with previous work showing that 4 days of hyperglycaemia reduces basal [ 37 ] and insulin-stimulated glycogen synthesis [ 3738 ].

Whether high glucose treatment slows or decreases the differentiation of myocytes to myotubes is unknown. However, the morphological profile of the myotubes was unchanged following any of the treatments Enhacing here. The increased glycogen synthesis and mRNA expression of PGC1, GLUT4 and MEF2C after long-term treatment with insulin may suggest enhanced myogenesis, although this is unlikely since the morphological pattern was unaltered.

Marked changes in morphology and skeletal muscle cell differentiation have been reported following 5 days of exposure to high concentrations of flycogen, another thiazolidinedione [ 39 ].

: Enhancing glycogen synthesis

Muscle Glycogen Synthesis after Exercise : an Enhancing Factor localized to the Muscle Cells in Man

This decrease was time-dependent, with a significant effect being observed after 2 h. During this time period, the fractional activity of GS also fell slightly, decreasing after 5 h of glucose starvation from 0. This again caused a dramatic activation of GS to a fractional activity of 0.

To confirm that alterations in GS activity were a result of previous glucose withdrawal, cells were incubated with DMEM supplemented with 5. These data indicate that the stimulation of GS is predominantly due to the action of glucose and requires previous glycogen depletion because of glucose removal, but other components of the different media may modulate the magnitude of the response.

The increase in GS fractional activity in response to glucose was dependent on the duration of previous glucose deprivation and, hence, inversely related to the glycogen content of the cells.

In the absence of glycogen depletion, no significant activation of GS by glucose was observed Fig. Total GS activity remained essentially unchanged in all conditions, indicating that alteration in the expression of the GS polypeptide was not involved not shown.

Glucose treatment 6. The stimulation of GS was dependent on the concentration of glucose added, with significant stimulatory effects being observed in response to 1. The combined effects of glucose readdition and insulin on cells cultured in the absence of glucose was then examined.

In the absence of readministration of glucose, stimulatory effects of insulin on GS activity were minimal after glycogen depletion data not shown. This value further increased to 1, An increase in the rate of 2-deoxyglucose uptake was observed in myoblasts deprived of glucose for increasing time, reaching 1.

The effect of insulin treatment on 2-deoxyglucose uptake was examined in control and glucose-starved myoblasts Fig. To investigate the mechanisms by which glucose activates GS, selective inhibitors of signaling pathways known to be stimulated by insulin were used Fig.

Preincubation of cells with rapamycin, which selectively inhibits the activation of p70 s6k , failed to inhibit this stimulatory effect of glucose on GS activation.

Treatment of glycogen-depleted myoblasts with either the MEK inhibitor, PD, or the PI 3-kinase inhibitor wortmannin partly reduced the activity state of GS obtained in response to glucose readministration.

However, this is essentially attributable to these inhibitors reducing the basal activity state of GS, as previously observed Therefore, these findings indicate that the mechanisms involved in the activation of GS by glucose are independent of the activity of PI 3-kinase, the classical mitogen-activated protein kinase pathway, and the rapamycin-sensitive pathway leading to activation of p70 s6k.

Furthermore, during glucose readministration, there was no observable decrease in the activity of GSK-3 and no detectable phosphorylation of that protein, as detected by phosphospecific anti—GSK-3 antibodies Fig. No changes in the activity of protein kinase B were detected not shown.

Glucosephosphate G6P is a potent allosteric activator of GS 19 , whereas a number of small metabolites, such as ATP, ADP, and AMP, are capable of inhibiting GS activity After refeeding of glycogen-depleted cells, fractionation of extracts failed to significantly alter GS activity ratio 0.

The activity of endogenous PPs against GS was then measured in extracts from control and glycogen-depleted cells. Because it has been demonstrated that GS becomes a better substrate for PPs in the presence of G6P 21 , this was carried out over a range of G6P concentrations.

No differences were found in the rate of activation of GS by endogenous phosphatases in extracts prepared from control and glycogen-depleted cells. For example, at a physiological concentration of 0.

Therefore, glycogen depletion does not appear to stimulate phosphatase activity against GS, at least when measured subsequently in cell extracts. The results presented here demonstrate dramatic effects on both glycogen synthesis and glycogen synthase after glucose re-administration to human myoblasts that have previously been depleted of glycogen.

This is expressed as a fractional activity, i. Similar methodologies have been applied to studies of GS activity in muscle, where activity has been reported as a fractional velocity or as A0. The molecular mechanism responsible for this stimulatory effect of glucose is a primary concern.

Inhibition of GSK-3 is thought to be the key event in the activation of muscle GS by insulin; however, selective inhibitors of the known insulin-stimulated pathways leading potentially to GSK-3 inhibition failed to prevent the activation of GS during the refeeding of glucose to glycogen-depleted cells Fig.

Furthermore, no inhibition or phosphorylation of GSK-3 was observed in response to glucose Fig. However, the involvement of GSK-3 inhibition in this process cannot be completely ruled out, as GSK-3 could be inhibited by an alternative, yet unidentified, mechanism not involving phosphorylation of the single serine residue recognized by the phosphospecific antibodies and not being retained when the enzyme is assayed in cellular extracts.

In that regard, allosteric regulation of GSK-3 by glycogen or small metabolites must be considered. A recent study performed in rat skeletal muscle demonstrated an exercise-induced increase in GS activity accompanied by inactivation of GSK-3 The activity of GS is dependent on the relative contribution of GSK-3 and PP1 in determining the phosphorylation state of the enzyme.

Phosphorylation of the glycogen-binding subunit of PP1 has been postulated as a mechanism for activation of that enzyme in response to insulin; however, a growing body of evidence suggests that this is not the case rev. After exercise in human muscle, no increase in total or glycogen-associated PP activity was observed; however, the rate of reactivation of GS by endogenous phosphatases was increased, suggesting that GS became a better substrate for PP1 after exercise In the present work, no increase in the phosphatase activity directed toward GS was observed after the glycogen depletion of human myoblasts.

However, it is possible that PPs, principally PP1, can alter GS activity in vivo, independent of changes in the intrinsic activity of the phosphatase detected in in vitro assays. For example, effects of changes in intracellular G6P concentration to enhance the ability of PP1 to dephosphorylate GS 21 , or the effect of overexpression of a glycogen-binding subunit of PP1 in CHO cells, which results in an increase in basal and insulin-stimulated glycogen synthesis 25 , would not be detected as a change in phosphatase activity in cell extracts.

Work in rat adipocytes has previously shown that glucose can activate GS in the absence of insulin 26 , an effect attributed to increased glucose transport leading to accumulation of G6P, which can then increase PP activity toward GS. The extent to which such a mechanism contributes to the observations in muscle reported in this study remains to be established, although studies in mouse diaphragm have demonstrated a modest stimulatory effect of glucose on GS activity The model from rat adipocytes does not account for the effect of glycogen stores on the activation, and a recent study in human myotubes has indicated that glycogen repletion, after glucose administration to cells that have been depleted of glycogen by overexpression of glycogen phosphorylase, is not dependent on increases in intracellular G6P concentration However, metabolic flux through G6P has been suggested as the hub of coordinate regulation of muscle glycogen synthesis 29 ; changes in GS activity are a consequence of alterations in intracellular G6P concentration, a result of increased glucose uptake and hexokinase activity.

G6P is an allosteric activator of GS, but also serves to make GS a better substrate for dephosphorylation by PPs Low glycogen content in vivo could increase glucose uptake and hexokinase activity, resulting in an increase in intracellular G6P concentration and consequently in an increase in GS activity, although no obvious mechanism for this is known.

A further consideration is whether glycogen depletion followed by glucose refeeding alters the distribution of GS and its regulatory proteins within the myoblast. PP1, GSK-3, and GS are all capable of redistributing within the cell in response to metabolic and hormonal stimuli 5 , 30 , If glucose or one of its metabolites caused either increased interaction of GS and PP1 or decreased interaction of GS and GSK-3 in glycogen-depleted cells, this could explain the activation of GS in response to this nutrient.

An exercise-induced increase in glycogen resynthesis after glycogen depletion can only occur if adequate substrate is available. Indeed, after glucose deprivation of cultured human muscle cells, there was a twofold increase in the subsequent rate of 2-deoxyglucose uptake.

Increased glucose transporter numbers at the plasma membrane has been suggested as the mechanism for increased glucose uptake postexercise, and in rat skeletal muscle, a 1. The relationship between glycogen and membrane permeability to glucose is apparently reciprocal, with increasing glycogen content postexercise leading to a decrease in glucose uptake In summary, the work reported here has highlighted the important role of glucose in stimulating glycogen synthesis in muscle after glycogen depletion.

Recent elegant work using 31 P nuclear magnetic resonance to study glycogen metabolism in human muscle in vivo is yielding important information on the effects of glycogen on glycogen synthesis However, work with human muscle cells in culture has the benefit of providing a means of independently varying glycogen content, extracellular glucose concentration, and insulin levels.

The data reported here are consistent with work in human muscle in vivo showing activation of GS after exercise-mediated glycogen depletion 22 , leading to glycogen repletion in an insulin-independent phase The molecular mechanism leading to the activation of GS by glucose after glycogen depletion is as yet undefined, but is apparently distinct from the upstream signaling mechanisms used in the actions of insulin on this parameter.

Recent evidence suggests that AMP-activated protein kinase may play a role in insulin-independent exercise-stimulated glucose uptake The possibility that it is also involved in the stimulation of glycogen synthesis after glycogen depletion is an attractive one, although no evidence is currently available to support this.

Clearly, how the two different signaling pathways interact has implications for the overall control of glycogen synthesis in control subjects and patients with type 2 diabetes. Total glycogen content and GS activity in myoblasts cultured in the absence of glucose; effect of glucose readdition.

In the absence of glucose readdition, the GS activity was less, at every time point, then what was observed in nonstarved cultures. The fractional activity of glycogen synthase was determined in these extracts ——— , left y- axis. Concentration- and time-dependent effects of glucose on GS activity in cells cultured in the absence of glucose.

Extracts were prepared and the fractional activity of glycogen synthase determined. Combined effects of insulin and glucose on GS activity and glycogen synthesis in cells cultured in the absence or presence of glucose. The insulin-mediated fold-activation over basal values is also indicated.

At the times indicated, the rate of glucose uptake was determined A. Effect of selective inhibitors on activation of GS by glucose. Extracts were prepared and the activity of GS determined. Effects of insulin and glucose on GSK-3 activity and phosphorylation. Extracts were prepared and GSK-3 activity determined.

This work was supported by a grant from Diabetes U. held a Cooperative Awards in Science and Engineering studentship from the Biotechnology and Biological Sciences Research Council BBSRC , U. holds a studentship from BBSRC. We thank Dr.

Mark Walker and Prof. Doug Turnbull for obtaining the initial muscle biopsies and Dorothy Fittes for technical assistance with the cell culture. Address correspondence and reprint requests to Stephen J. Yeaman, School of Biochemistry and Genetics, the Medical School, University of Newcastle, Newcastle upon Tyne NE2 4HH, U.

E-mail: s. yeaman ncl. has received funds from Novo Nordisk. is employed by and holds stock in Novo Nordisk. holds stock in Xcellsyz, has received honoraria from Novo Nordisk and Glaxo Wellcome, and has received funds from Novo Nordisk and SmithKline Beecham.

Sign In or Create an Account. Search Dropdown Menu. header search search input Search input auto suggest. filter your search All Content All Journals Diabetes.

Advanced Search. User Tools Dropdown. Sign In. Skip Nav Destination Close navigation menu Article navigation. Volume 50, Issue 4. Previous Article Next Article. RESEARCH DESIGN AND METHODS.

Article Information. Article Navigation. Metabolism and Signal Transduction April 01 Control of Glycogen Synthesis by Glucose, Glycogen, and Insulin in Cultured Human Muscle Cells Reza Halse ; Reza Halse.

This Site. Google Scholar. Sylvie M. Bonavaud ; Sylvie M. Jane L. Armstrong ; Jane L. James G. McCormack ; James G. Stephen J. Yeaman Stephen J. Diabetes ;50 4 — Get Permissions. toolbar search Search Dropdown Menu. toolbar search search input Search input auto suggest.

View large Download slide. Lawrence JC Jr, Roach PJ: New insights into the role and mechanism of glycogen synthase activation by insulin. Parker PJ, Caudwell FB, Cohen P: Glycogen synthase from rabbit skeletal muscle: effect of insulin on the state of phosphorylation of the seven phosphoserine residues in vivo.

Download citation. Published : 01 April Issue Date : 16 April Anyone you share the following link with will be able to read this content:. Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative. European Journal of Applied Physiology Journal of Physiology and Biochemistry By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily. Skip to main content Thank you for visiting nature.

nature letters article. Abstract IT is well known that glycogen is utilized during muscular work, but there is very little information available about the resynthesis of glycogen after exhaustive exercise. Access through your institution.

Buy or subscribe. Change institution. Learn more. References Goldstein, M. Article Google Scholar Bergström, J. Google Scholar Bergström, J. Article ADS Google Scholar Hultman, E. Author information Authors and Affiliations Clinical Laboratory, St.

View author publications. Rights and permissions Reprints and permissions. About this article Cite this article BERGSTRÖM, J. Copy to clipboard. This article is cited by Sexual Dimorphism in Substrate Metabolism During Exercise Stéphanie M. Abo Elisa Casella Anita T.

Layton Bulletin of Mathematical Biology Analysis of sex-based differences in energy substrate utilization during moderate-intensity aerobic exercise Antonella Cano Lucia Ventura Andrea Manca European Journal of Applied Physiology A century of exercise physiology: key concepts in regulation of glycogen metabolism in skeletal muscle Abram Katz European Journal of Applied Physiology Do Sex Differences in Physiology Confer a Female Advantage in Ultra-Endurance Sport?

Nicholas B. Tiller Kirsty J. Elliott-Sale Guillaume Y. Millet Sports Medicine Enhanced skeletal muscle glycogen repletion after endurance exercise is associated with higher plasma insulin and skeletal muscle hexokinase 2 protein levels in mice: comparison of level running and downhill running model Yumiko Takahashi Juli Sarkar Hideo Hatta Journal of Physiology and Biochemistry

Dietary strategies to promote glycogen synthesis after exercise

Our results are consistent with previous work showing that 4 days of hyperglycaemia reduces basal [ 37 ] and insulin-stimulated glycogen synthesis [ 37 , 38 ]. Whether high glucose treatment slows or decreases the differentiation of myocytes to myotubes is unknown. However, the morphological profile of the myotubes was unchanged following any of the treatments investigated here.

The increased glycogen synthesis and mRNA expression of PGC1, GLUT4 and MEF2C after long-term treatment with insulin may suggest enhanced myogenesis, although this is unlikely since the morphological pattern was unaltered. Marked changes in morphology and skeletal muscle cell differentiation have been reported following 5 days of exposure to high concentrations of troglitazone, another thiazolidinedione [ 39 ].

However, after rosiglitazone exposure, the morphology of the primary human skeletal muscle cells was unaltered. The difference between rosiglitazone and troglitazone in their effects on muscle morphology may be due to differences in the concentration of glitazone studied or a compound-specific effect.

Interestingly, short-term exposure 1 day of primary human muscle cells to troglitazone enhanced insulin action, similar to the results reported here for rosiglitazone [ 39 ]. Most in vitro studies conducted to dissect the effects of rosiglitazone and metformin on metabolic and gene-regulatory responses have been performed using adipocytes or hepatocytes.

Direct effects of troglitazone have previously been reported on cell lipid metabolism [ 40 ], gene expression [ 41 ] and glucose uptake [ 39 , 42 ] in human skeletal muscle.

Here we report that 8 days of rosiglitazone treatment increases glycogen synthesis in the absence or presence of insulin, and these changes are accompanied by increased expression of GLUT4 and PGC1 mRNA.

In our hands, GLUT4 and GLUT1 mRNA and protein expression are positively correlated in differentiating human skeletal muscle cells data available upon request. In contrast to these findings, in L6 myotubes rosiglitazone has been suggested to increase GLUT4 translocation, but not protein or mRNA content [ 43 ].

However, this particular conclusion was based on a much shorter rosiglitazone exposure 24 h , which may be insufficient to mediate gene-regulatory responses. Thus, in human muscle cells, part of the insulin-sensitising effect of rosiglitazone can be attributed to changes in mRNA and protein expression of glucose transporters.

AICAR activates AMPK and stimulates glucose uptake in skeletal muscle [ 45 ]. In the present study, acute AICAR treatment increased glycogen synthesis, while chronic 8 days AICAR treatment was without effect on insulin action, as assessed by glycogen synthesis, or expression of GLUT4 or PGC1 mRNAs.

The acute responses are consistent with findings in rat skeletal muscle [ 31 , 46 ] and perfused hindlimb [ 47 ], where AICAR treatment increased GLUT4 expression and translocation respectively. Furthermore, chronic AICAR treatment of Clone 9 cells increased glucose uptake [ 48 ].

Based on the observation that GLUT1 content was unchanged in the plasma membranes of AICAR-treated Clone 9 cells, the effects on glucose uptake were proposed to activate pre-existing plasma membrane GLUT1 transporters. Nevertheless, our present data reveal that chronic AICAR treatment in human myotubes selectively increases GLUT1, but not GLUT4, expression.

Moreover, in human myotubes exposed to AICAR for 8 days, mRNA expression of MEF2C was reduced. Whether AICAR has a negative effect on the regulation of myogenesis in this cell system remains to be demonstrated, but no such effects have previously been suggested.

However, MEF2C is involved in the regulation of differentiation-specific genes [ 16 ] and suppression of this gene may repress gene-regulatory responses. Metformin treatment of cells increases GLUT4 recruitment and increases glycogen synthesis [ 49 ].

Here we demonstrate that chronic metformin exposure increases the expression of GLUT4, PGC1 and all MEF2 isoforms A, C and D and increases the rate of basal and insulin-stimulated glycogen synthesis.

Metformin has been suggested to increase AMPK activity [ 29 ]. Interestingly, in human myotubes, metformin treatment was more potent than AICAR in increasing GLUT4 expression and in enhancing insulin sensitivity, suggesting that metformin activates additional AMPK-independent pathways.

In summary, exposure of human myotubes to metformin and rosiglitazone, two currently available antidiabetic agents, was associated with direct enhancement of the action of insulin in cultured skeletal muscle. The increase in insulin action correlated with increased mRNA expression of GLUT4.

Increased expression of PGC1, GLUT4 and MEF2 mRNA after antidiabetic treatment provides a molecular mechanism for increased carbohydrate metabolism, cellular survival and gene-regulatory responses that confer improved skeletal muscle metabolic function in insulin-resistant and diabetic subjects.

McGarry JD Glucose—fatty acid interactions in health and disease. Am J Clin Nutr S—S. CAS PubMed Google Scholar. Boden G Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes — Ciaraldi TP, Abrams L, Nikoulina S, Mudaliar S, Henry RR Glucose transport in cultured human skeletal muscle cells.

Regulation by insulin and glucose in nondiabetic and non-insulin-dependent diabetes mellitus subjects. J Clin Invest — Kelley DE, Goodpaster BH Effects of exercise on glucose homeostasis in type 2 diabetes mellitus. Med Sci Sports Exerc S—S Lewis GF, Carpentier A, Adeli K, Giacca A Disordered fat storage and mobilization in the pathogenesis of insulin resistance and type 2 diabetes.

Endocr Rev — Article CAS PubMed Google Scholar. Zierath JR, Krook A, Wallberg-Henriksson H Insulin action and insulin resistance in human skeletal muscle. Diabetologia — Charron MJ, Brosius FC III, Alper SL, Lodish HF A glucose transport protein expressed predominately in insulin-responsive tissues.

Proc Natl Acad Sci U S A — James DE, Strube MM, Mueckler MM Molecular cloning and characterization of an insulin-regulatable glucose transporter. Nature — Tsao T-S, Stenbit A, Li J et al Muscle-specific transgenic complementation of GLUT4-deficient mice: preferential effects of glucose but not lipid metabolism.

Shepherd PR, Gnudi L, Tozzo E et al Adipose cell hyperplasia and enhanced glucose disposal in transgenic mice overexpressing GLUT4 selectively in adipose tissue. J Biol Chem — Torrance CJ, Devente JE, Jones JP, Dohm GL Effects of thyroid hormone on GLUT4 glucose transporter gene expression and NIDDM in rats.

Endocrinology — Mora S, Pessin JE The MEF2A isoform is required for striated muscle-specific expression of the insulin-responsive GLUT4 glucose transporter. Thai MV, Guruswamy S, Cao KT, Pessin JE, Olson AL Myocyte enhancer factor 2 MEF2 -binding site is required for GLUT4 gene expression in transgenic mice: regulation of MEF2 DNA binding activity in insulin-deficient diabetes.

Biochem Biophys Res Commun — McKinsey TA, Zhang CL, Olson EN MEF2: a calcium-dependent regulator of cell division, differentiation and death. Trends Biochem Sci — Mora S, Yang C, Ryder JW, Boeglin D, Pessin JE The MEF2A and MEF2D isoforms are differentially regulated in muscle and adipose tissue during states of insulin deficiency.

Al-Khalili L, Chibalin AV, Yu M et al MEF2 activation in differentiated primary human skeletal muscle cultures requires coordinated involvement of parallel pathways. Am J Physiol Cell Physiol C—C Handschin C, Rhee J, Lin J, Tarr PT, Spiegelman BM An autoregulatory loop controls peroxisome proliferator-activated receptor γ coactivator 1 α expression in muscle.

Puigserver P, Wu Z, Park WC et al A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell — Patti ME, Butte AJ, Crunkhorn S et al Coordinated reduction of genes of oxidative metabolism in humans with insulin resistance and diabetes: potential role of PGC1 and NRF1.

Sood V, Colleran K, Burge MR Thiazolidinediones: a comparative review of approved uses. Diabetes Technol Ther — Reasner CA Promising new approaches.

Diabetes Obes Metab 1:S41—S Mauvais-Jarvis F, Andreelli F, Hanaire-Broutin H, Charbonnel B, Girard J Therapeutic perspectives for type 2 diabetes mellitus: molecular and clinical insights.

Diabetes Metab — Nambi V, Hoogwerf RJ, Sprecher DL A truly deadly quartet: obesity, hypertension, hypertriglyceridemia, and hyperinsulinemia. Cleve Clin J Med — PubMed Google Scholar. Ciaraldi TP, Kolterman OG, Scarlett JA, Kao M, Olefsky JM Role of glucose transport in the postreceptor defect of non-insulin-dependent diabetes mellitus.

Garg R, Tripathy D, Dandona P Insulin resistance as a proinflammatory state: mechanisms, mediators, and therapeutic interventions. Curr Drug Targets — Mayerson AB, Hundal RS, Dufour S et al The effects of rosiglitazone on insulin sensitivity, lipolysis, and hepatic and skeletal muscle triglyceride content in patients with type 2 diabetes.

Ruderman NB, Cacicedo JM, Itani S et al Malonyl—CoA and AMP-activated protein kinase AMPK : possible links between insulin resistance in muscle and early endothelial cell damage in diabetes.

Biochem Soc Trans — Musi N, Hirshman MF, Nygren J et al Metformin increases AMP—activated protein kinase activity in skeletal muscle of subjects with type 2 diabetes. Acta Physiol Scand — Al-Khalili L, Chibalin AV, Kannisto K et al Insulin action in cultured human skeletal muscle cells during differentiation: assessment of cell surface GLUT4 and GLUT1 content.

Cell Mol Life Sci — Cline GW, Petersen KF, Krssak M et al Impaired glucose transport as a cause of decreased insulin-stimulated muscle glycogen synthesis in type 2 diabetes. N Engl J Med — Park KS, Ciaraldi TP, Carter L et al Induction of insulin resistance in human skeletal muscle cells by downregulation of glycogen synthase protein expression.

Metabolism — Gaster M, Petersen I, Hojlund K, Poulsen P, Beck-Nielsen H The diabetic phenotype is conserved in myotubes established from diabetic subjects: evidence for primary defects in glucose transport and glycogen synthase activity.

Gaster M, Schroder HD, Handberg A, Beck-Nielsen H The basal kinetic parameters of glycogen synthase in human myotube cultures are not affected by chronic high insulin exposure. Biochim Biophys Acta — Henry RR, Ciaraldi TP, Mudaliar S, Abrams L, Nikoulina SE Acquired defects of glycogen synthase activity in cultured human skeletal muscle cells: influence of high glucose and insulin levels.

Kausch C, Krutzfeldt J, Witke A et al Effects of troglitazone on cellular differentiation, insulin signaling, and glucose metabolism in cultured human skeletal muscle cells. Wahl HG, Kausch C, Machicao F et al Troglitazone downregulates δ 6 desaturase gene expression in human skeletal muscle cell cultures.

Park KS, Ciaraldi TP, Lindgren K et al Troglitazone effects on gene expression in human skeletal muscle of type II diabetes involve up-regulation of peroxisome proliferator-activated receptor-γ. J Clin Endocrinol Metab — Park KS, Ciaraldi TP, Abrams-Carter L et al Troglitazone regulation of glucose metabolism in human skeletal muscle cultures from obese type II diabetic subjects.

Total glycogen synthase protein levels in homogenates, however, were unaffected by linoleate Fig. To address this discrepancy, and to compare the distribution of activity in L6 myotubes with that observed in rat muscles, we also assayed glycogen synthase activity in fractions from both control and linoleate-treated cells.

Total activities Fig. In this case, loss of activity in the cytosol was more comparable with the increased activity measured in the TIF, suggesting that the enzyme was indeed translocating in response to both insulin and linoleate. To determine the activation state of glycogen synthase in the different fractions, we also measured the activity in the presence of a more physiological G6P concentration Fig.

Insulin tended to increase the activity observed in the cytosol from control cells Fig. Increased activity was also observed in the TIF from insulin-stimulated control cells Fig.

In contrast, the glycogen synthase activity measured in the TIF from linoleate-pretreated cells Fig. In fact, lipid pretreatment significantly reduced the activation state of the enzyme observed in the TIF upon insulin stimulation relative to that in the TIF from control cells Fig.

The data presented in Fig. Because an insulin-sensitive pool from control cells was recovered in the same subcellular fraction, we examined potential causes of the lipid-dependent redistribution of the enzyme and attempted to distinguish biochemically between the pools.

Increased lipid availability can increase flux through the hexosamine pathway and promote protein glycosylation Hawkins et al. No evidence, however, for such a modification was observed in immunoprecipitates of the enzyme with an antibody specific for O -glycosylated protein in subsequent immunoblots not shown.

In addition, we were unable to determine any changes in proteins associated with glycogen synthase in immunoprecipitates, after SDS—PAGE and silver staining not shown. We also attempted to separate the two pools in the TIF by sequential solubilization involving Triton X, SDS- and urea-containing buffers.

No further fractionation, however, of the enzyme was achieved not shown. Immunoblotting using antibodies specific for the glycogen targeting protein PTG indicated that this was mostly recovered in the cytosol from L6 myotubes, with a minor proportion located in the TIF not shown , while the targeting protein R GL G M was not detected in L6 cell fractions, but was observed in the membrane fraction from skeletal muscle.

Both proteins failed to show alterations distribution in response to increased lipid availability not shown , suggesting that they did not mediate the change in the recovery of glycogen synthase through their own translocation.

Finally, because the translocation of glycogen synthase from a cytosolic fraction has been linked to alterations in cellular glycogen content in adipocytes Brady et al. Firstly, we determined the effect of linoleate on steady state glycogen levels. Upon fractionation of the cells, the greatest amount of glycogen was found in the TIF, but there was no effect of the FFA on total glycogen content in any fraction Fig.

Next, we examined the subcellular distribution of the glycogen synthesized over 1 h in response to insulin stimulation in control and lipid-pretreated myotubes previously measured in whole lysates Fig.

In this case, glycogen synthesized in response to insulin was recovered almost exclusively in the TIF, and this component was greatly reduced in linoleate-pretreated cells Fig.

Together with the data shown in Fig. The activity of glycogen synthase in muscle cells is dependent upon several factors. In addition to the importance of the phosphorylation state of the enzyme, the concentration of its allosteric activator G6P and the actions of the glycogen-targeting proteins, the role of cellular localization is becoming apparent Fernandez-Novell et al.

We here present evidence supporting the novel hypothesis that lipid-induced insulin resistance at the level of glycogen synthesis is explained at least in part by a repartitioning of glycogen synthase to an insulin-insensitive pool. The insulin-dependent translocation of glycogen synthase which we have observed in muscle of chow-fed rats and in control cells agrees with previous studies Brady et al.

An insulin-dependent association with glycogen is supported by the fact that the TIF contained the largest amount of total glycogen, and that glycogen synthesized in response to the hormone was mostly recovered in this fraction. Furthermore, while we observed translocation in rat muscle after a hyperinsulinaemic clamp, and in control cells upon 1-h insulin stimulation, as consistent with the co-sedimentation of glycogen synthase with newly synthesized, large, insoluble glycogen molecules, we did not observe such translocation after only min insulin stimulation data not shown , when smaller, nascent glycogen molecules might be recovered in the cytosol.

It should be noted that although we observed changes in the recovery of glycogen synthase in different subcellular fractions, these could indicate alterations in the association of the enzyme with protein or carbohydrate complexes, rather than in cellular localization, as previously reported in response to other stimuli Ferrer et al.

Thus, although immunoblotting and in vitro assays indicated that the major proportion of glycogen synthase is recovered in the TIF even in the absence of lipid, it is possible that in intact cells and muscle this pool is inhibited by association with glycogen Nielsen et al. Although fat-feeding, obesity or linoleate pretreatment appeared to cause a similar redistribution of glycogen synthase to the TIF, the pool of the enzyme increased in the TIF by lipid was probably distinct from that increased by insulin.

Firstly, glycogen synthase appearing in the TIF in a lipid-dependent manner exhibited a low fractional velocity and was resistant to insulin; second, the redistribution was associated with an inhibition of insulin-stimulated glycogen synthesis, in direct contrast to the repartitioning caused by insulin itself.

This is consistent with the sequestration of glycogen synthase in the TIF by lipid oversupply in such a way that it is no longer sensitive to activation by insulin. The amount of cytosolic glycogen synthase is thus reduced, and we hypothesize that this apparently minor pool of the enzyme is necessary for the full increase in glycogen synthesis to occur in response to the hormone.

Our observations are thus similar to those made with adipocytes Jensen et al. Insulin pretreatment, which was associated with diminished glycogen synthesis upon subsequent insulin stimulation, caused prior partitioning of the enzyme to the denser fraction.

It was concluded that a minor component of glycogen synthase was responsible for most of the newly synthesized glycogen in adipocytes, and that correct basal localization was required for activation of the enzyme by insulin Jensen et al.

Our data support a similar situation in muscle, in that the cytosolic component may contribute most to glycogen synthesis, and furthermore suggest that lipid oversupply, like insulin pretreatment, can lead to a sequestration of the enzyme that contributes to a reduction in subsequent, insulin-stimulated glycogen synthesis.

A possible explanation is that this redistribution reduces access by PP1 and hence limits activation normally promoted by dephosphorylation of the enzyme. Such a mechanism is supported by the lack of effect of linoleate on the insulin-stimulated activation of PKB, suggesting that the pathway downstream of this signalling component, which reduces further phosphorylation of glycogen synthase by inhibition of GSK-3, is still intact.

While we were unable to determine the mechanism by which lipid oversupply causes such a redistribution, we were able to discount a simple role for glycogen accumulation. Firstly, the glycogen content of rat skeletal muscle was not significantly increased by fat-feeding; second, linoleate pretreatment did not alter the steady-state levels of glycogen in any subcellular fraction from L6 myotubes, despite an increase in the partitioning of the enzyme in the TIF in each case.

We were also unable to find evidence for glycosylation or the association of a binding partner. We cannot, however, rule out a post-translational modification, such as phosphorylation of the enzyme or its targeting proteins, which might affect its binding. Indeed, a reason for the discrepancy between the amounts of glycogen synthase protein appearing in the TIF of lipid-treated cells and that translocating from the cytosol as determined by immunoblotting Fig.

Alternatively, direct association of glycogen synthase with lipid species in intact cells may promote its inhibition and sedimentation, as described for phosphatidic acid phosphohydrolase-1 Elabbadi et al. Furthermore, the lack of effect of insulin on steady-state G6P levels measured in these cells indicates that the observed increase in glycogen synthesis in response to insulin in control cells Fig.

Importantly, therefore, the inhibitory effect of linoleate on insulin-stimulated glycogen synthesis cannot be explained by a reduced effect of insulin on G6P concentration. Similarly, previous studies have also shown that G6P content is not reduced in muscle of fat-fed rats Kim et al.

In any case, the differing activation states of cytosolic and TIF-associated glycogen synthase we have observed in lipid-treated myotubes indicate that the cellular G6P concentration cannot be solely responsible for alterations in glycogen synthesis.

Taken together, these findings suggest that a primary defect at the level of glycogen synthase contributes to the diminished glycogen synthesis observed in the presence of lipid, strengthening the case for a role of compartmentalization.

The relative importance of the rate of glucose uptake and the activity of glycogen synthase in the determination of the rate of glycogen synthesis has been controversial, and it is likely that either factor can play an overriding role under specific physiological circumstances Fisher et al.

Thus, while NMR studies have indicated that glucose transport can be rate-limiting Roden et al. An impairment of glycogen synthase activity also contributes to the diminished glycogen synthesis seen in lipid-infused human subjects at higher free fatty acid levels Boden et al.

In summary, while previous work has suggested that exposure of muscle to increased lipid levels does not greatly affect the activity of glycogen synthase Johnson et al. While the myotubes are not an exact representation of muscle tissue, an advantage of the model we have employed is the ability to define conditions for the study of glycogen synthase under which the contribution of lipid effects on glucose transport and phosphorylation can be discounted.

This study has thus indicated a new aspect of lipid-induced insulin resistance at the level of glycogen synthesis, although further work is required to elucidate the mechanism of glycogen synthase repartitioning. The effect of high-fat feeding on glycogen synthase protein partitioning in rat skeletal muscle.

Rats which had been fed either a chow or a high-fat diet were subjected to a euglycaemic-hyperinsulinaemic clamp, or maintained under basal conditions, as indicated.

Skeletal muscle was fractionated and subjected to immunoblotting. a Immunoblots obtained with glycogen synthase and GSK-3 antibodies are shown. b The results of densitometry of glycogen synthase bands from muscle fractions of six animals per group are shown. c Immunoblots of glycogen synthase in the Triton-insoluble fraction TIF.

d Muscle fractions from chow- and fat-fed rats were assayed for glycogen synthase activity in the presence of 10 mM G6P to indicate the total amount of enzyme present in each fraction. The means of activities from muscle fractions of six animals per group are shown — note the discontinuous ordinate axis.

Citation: Journal of Endocrinology , 1; TIF fractions were diluted sixfold in comparison to cytosolic fractions. b The results of densitometry of glycogen synthase bands from muscle fractions of four animals per group, corrected for dilution, are shown. Effect of linoleate on glycogen synthesis and glucose uptake in L6 skeletal muscle cells.

a Basal and insulin-stimulated glycogen synthesis was determined in control myotubes and in myotubes pretreated for 16 h with linoleate. Results shown are means from three experiments, each carried out in triplicate.

b Basal and insulin-stimulated glucose uptake were determined in control and lipid-pretreated myotubes under the same conditions as for a. Results shown are means from two experiments, each carried out in triplicate. Effect of linoleate on glycogen phosphorylase and PKB. The activity of glycogen phosphorylase was determined in homogenates, in either the presence a or absence b of 5 mM AMP.

Results shown are combined means from six independent experiments, each carried out in duplicate. c Basal and insulin-stimulated PKB Ser phosphorylation was determined in lysates from control and lipid-pretreated myotubes by immunoblotting with a phospho-specific antibody.

The results of densitometry from three independent experiments, each carried out in triplicate, are shown. Effect of linoleate on glycogen synthase protein partitioning in L6 skeletal muscle cells. a Myotubes were pretreated in the absence or presence of linoleate for 16 h and incubated without or with insulin for 1 h.

Whole-cell lysates were fractionated to obtain cytosolic, solubilized-membrane and Triton-insoluble fractions. Samples were subjected to immunoblotting with either glycogen synthase high and low exposures of the same blot are shown , GSK-3 or β-actin antibodies as indicated.

b The results of densitometry of glycogen synthase bands from three independent experiments carried out in duplicate are shown. c Whole-myotube lysates were subjected to immunoblotting with antiglycogen synthase or anti-β-actin antibodies as indicated. The means from densitometry of three independent experiments done in triplicate are shown, after correction of glycogen synthase for β-actin loading lower panel.

Effect of linoleate on recovery of glycogen synthase activity in subcellular fractions from L6 cells. Myotubes were treated and fractionated as given in the legend for Fig. c The fractional velocity of glycogen synthase was calculated for each fraction as the ratio of activity measured at 0.

The means from three independent experiments are shown. Effect of linoleate on steady-state glycogen content and acute glycogen synthesis in subcellular fractions. a Myotubes were incubated for 48 h in media containing d -[U- 14 C]glucose before treatment with linoleate for 16 h, also in the presence of d -[U- 14 C]glucose, and fractionated as given in the legend for Fig.

Radiolabelled glycogen was determined in each fraction. Results shown are combined means from three independent experiments, each carried out in triplicate. b Myotubes were pretreated without or with linoleate for 16 h and incubated for 1 h in the absence or presence of insulin for the measurement of glycogen synthesis from d -[U- 14 C]glucose.

Radiolabelled glycogen was determined in each subcellular fraction. Results shown are means from five independent experiments, each carried out in triplicate. This work was funded by grants from the National Health and Medical Research Council of Australia CSP , and the Diabetes Australia Research Trust CSP, JMY , and by an Australian Postgraduate Award AJT.

The authors declare that there is no conflict of interest that would prejudice the impartiality of this scientific work. American Journal of Physiology E —E Hormone and Metabolic Research 14 — Journal of Clinical Investigation 93 — Journal of Biological Chemistry — FEBS Letters 3 — Coleman DL Obese and diabetes: two mutant genes causing diabetes-obesity syndromes in mice.

Diabetologia 14 — Dresner A , Laurent D, Marcucci M, Griffin ME, Dufour S, Cline GW, Slezak LA, Andersen DK, Hundal RS, Rothman DL et al. Journal of Clinical Investigation — Biochimie 87 — Biochemical Journal — FEBS Letters — Biochemical Journal 17 — Analytical Biochemistry 47 20 — Journal of Clinical Investigation 99 — Mohamed A.

Omar, Lianguo Wang, Alexander S. Glycogen synthase kinase-3 GSK-3 is a multi-functional kinase that regulates signalling pathways affecting glycogen metabolism, protein synthesis, mitosis, and apoptosis.

GSK-3 inhibition limits cardiac ischaemia—reperfusion IR injury, but mechanisms are not clearly defined. Rates of glucose and palmitate oxidation were improved by SB. Glycogen synthase kinase-3 GSK-3 is a multi-functional kinase that regulates signalling pathways affecting glycogen metabolism, protein synthesis, mitosis and apoptosis.

It has 2 isoforms, α and β, that possess strong homology in their kinase domains. It actively inhibits hypertrophy and its inhibition stimulates development of cardiac hypertrophy. Tong et al. Furthermore, inhibition of GSK-3 was suggested as a mechanism explaining cardioprotection induced by postconditioning, 5 opioids, 6 bradykinin, 7 erythropoietin, 8 adenosine A 3 receptor activation, 9 isoflurane, 10 and PKCδ inhibition.

One proposed mechanism involves prevention of mitochondrial permeability transition pore mPTP opening 12 potentially due to effects on the voltage-dependent anion channel VDAC 13 or adenine nucleotide translocase ANT.

In addition, recent evidence from mitochondria that are deficient in all isoforms of VDAC shows that VDAC is dispensable in mPTP opening. Interestingly, although the initial function and naming of GSK-3 was related to its effects on glycogen synthase GS activity, the contribution of alterations in glycogen or glucose metabolism by GSK-3 inhibition to cardioprotection has not been investigated.

GSK-3 phosphorylates GS at Ser site 3a and Ser site 3b via a hierarchal mechanism and thereby inhibits GS activity. In this study, we test the hypothesis that inhibition of GSK-3 will stimulate glycogen synthesis, repartition glucose partially away from glycolysis, improve the coupling between glycolysis and glucose oxidation and reduce the potential for intracellular acidosis.

Male Sprague—Dawley rats — g were used in this study. All experiments were performed with accordance with the guidelines of the Animal Use and Care Committee, University of Alberta animal ethics protocol The investigation conforms with the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health NIH Publication No.

Rat hearts were cannulated for isolated working mode perfusion in a recirculating system under conditions of constant workload Rates of glycolysis, glucose oxidation, and palmitate oxidation were measured as described previously.

After an initial 45 min of baseline aerobic perfusion, hearts were subjected to 17 min of global ischaemia GI followed by 30 min of reperfusion. This was followed by 20 min of GI and 30 min of reperfusion. These durations of GI were sufficient to cause marked LV dysfunction that is reversible.

Longer periods of ischaemia resulted in the failure of the hearts to recover at reperfusion which would have hindered the measurement of the metabolic rates.

Once added, the drug remained in the recirculating system until the end of the perfusion protocol. This concentration of SB was shown previously to produce sufficient inhibition of GSK-3 and to induce cardioprotection in the isolated perfused rat heart.

Two series of hearts with different pre-treatment glycogen contents were studied. Results are expressed as means ± SEM of n observations. The significance of the differences for two group comparisons was estimated by Student's t -test.

The significance of difference in time-course experiments was estimated by two-way analysis of variance with repeated measures on time and a Newman—Keuls post-test.

Left ventricular mechanical function was stable during the initial period of baseline perfusion with no differences among experimental groups Figure 1 A. This cardioprotective effect of SB is similar to effects observed in previous studies.

SB improves recovery of LV function during reperfusion. A LV work during IR protocol in isolated working rat hearts treated with vehicle DMSO, 0.

Values are means ± SEM. SB-mediated alterations in energy substrate metabolism during reperfusion. This increase in ATP production by SB resulted from a higher contribution of palmitate and glucose oxidation, while there was a lower contribution from glycolysis Figure 2 I. This was associated with improved recovery of post-ischaemic LV function to In order to assess the exact role of the stimulation of glycogen synthesis induced by inhibition of GSK-3, it is important to delineate the cause and effect relationship between glycogen and glucose metabolism and improved LV function during reperfusion.

For this purpose, we studied the effects of SB in aerobically perfused hearts no ischaemia with normal G-replete or partially depleted glycogen stores G-depleted see Supplementary material online, Figure S1. SB had no effect on LV work in G-replete hearts 2.

SB elicited only a minor alteration in the rate of glycogen synthesis that was not significantly different from vehicle-treated hearts Figure 5 A. Effects of SB on glycogen and glucose metabolism in aerobic hearts.

SB had no effect on LV work in G-depleted hearts 2. SB did not affect glucose oxidation Figure 5 G. Effects of SB when administered at the onset of reperfusion. The first evidence for the role of GSK-3 in cardioprotection was obtained in studies showing that ischaemic preconditioning results in phosphorylation and inhibition of GSK-3β and that pharmacological inhibition of GSK-3 mimics the cardioprotective effects of preconditioning.

Specifically, our data indicate that inhibition of GSK-3 increases glycogen synthesis during reperfusion which partially repartitions glucosephosphate away from glycolysis. We also provide evidence that acceleration of glycogen synthesis is not a consequence of improved LV function, as similar metabolic alterations occur in glycogen-depleted aerobic hearts independent of changes in LV mechanical function.

In order to examine the relative rates of glycogen synthesis and glycolysis in the absence and presence of GSK-3 inhibition, studies were performed in isolated rat hearts that were perfused in working mode with both glucose and palmitate as energy substrates. These conditions ensure hearts are studied under conditions of physiological work load energy demand as well as adequate energy supply.

Moreover, aerobic perfusion conditions ensure the re-establishment of normal glycogen content previously severely depleted during deep anaesthesia and heart extraction , a key requirement for investigations of glucose and glycogen metabolism. While alteration of glycolysis may be involved, other mechanisms arising from GSK-3 inhibition during ischaemia may contribute, such as improved ionic homeostasis due to reduced mitochondrial ATP consumption, an effect possibly due to interaction of GSK-3 with VDAC.

However, as LV mechanical function energy demand and energy substrate metabolism are interdependent, additional experiments were performed in aerobic hearts in order to determine if alteration of glucose partitioning might simply be a consequence, rather than a cause, of enhanced recovery of LV function.

The ability of SB to produce a similar re-partitioning of glucose metabolism in aerobic hearts that are partially depleted of glycogen to levels similar to the end of GI confirms that the alterations in metabolism are not a consequence of changes in LV function.

Rather, it indicates that the enhanced recovery of LV function is due to the changes in metabolism. A well-described downstream consequence of GSK-3 inhibition is delayed opening of mPTP in response to reactive oxygen species.

However, direct interaction of GSK-3 with VDAC reduces adenine nucleotide transport across the outer mitochondrial membrane independent of mPTP opening, 17 thereby conserving ATP content by reducing mitochondrial ATP consumption.

However, it cannot explain cardioprotection when SB is administered only at the onset of reperfusion, a period when ATP generation returns close to pre-ischaemic levels.

Although the open probability of mPTP is reduced sharply in acidic pH in de-energized mitochondria, 51 , 52 exposure of respiring mitochondria to an acidic environment, such as in early reperfusion, will favour mitochondrial inorganic phosphate uptake that facilitates mPTP opening.

This may also explain the improved mitochondrial function, demonstrated by enhanced glucose and palmitate oxidation, during reperfusion in SB-treated hearts.

The stimulated mitochondrial oxidation may also arise due to the improved recovery of LV function and higher energy demand in SB-treated hearts. Furthermore, a direct interaction is unlikely as GSK-3 inhibition has no effect on mPTP opening in isolated mitochondria.

Although GSK-3 was initially discovered and named for its role in regulating glycogen metabolism, this is the first study to link this important effect on myocardial metabolism with cardioprotection. Our study highlights the ability of GSK-3 to regulate myocardial glycogen and glucose metabolism and demonstrates an additional mechanism linking GSK-3 inhibition with enhanced recovery of post-ischaemic mechanical function.

Dietary strategies to promote glycogen synthesis after exercise As previously reported, inclusion of 2-DG enhanced basal and insulin-stimulated glycogen synthase activity Fig. Diabetes Metab — CAS PubMed Google Scholar Nambi V, Hoogwerf RJ, Sprecher DL A truly deadly quartet: obesity, hypertension, hypertriglyceridemia, and hyperinsulinemia. Glycogen synthase kinase-3beta mediates convergence of protection signaling to inhibit the mitochondrial permeability transition pore. Based on the observation that GLUT1 content was unchanged in the plasma membranes of AICAR-treated Clone 9 cells, the effects on glucose uptake were proposed to activate pre-existing plasma membrane GLUT1 transporters. Sherman WM, Costill DL, Fink WJ, Miller JM. More on this topic The coronary circulation in cardioprotection: more than just one confounder.
Muscle Glycogen Synthesis Before and After Exercise The aim of this study was to Mindfulness and brain health the effect of several antidiabetic agents glycogeh insulin-stimulated glycoegn synthesis, as well as on mRNA expression. Skip Nav Destination Amino acid metabolism navigation menu Article navigation. Synthesls by Enhancing glycogen synthesis Springer Synthesjs SharedIt Synthesiis initiative. Navbar Enhancing glycogen synthesis Filter Endocrinology This issue Endocrine Society Journals Clinical Medicine Endocrinology and Diabetes Medicine and Health Books Journals Oxford Academic Mobile Enter search term Search. International Journal of Sports Medicine 2: —, Article PubMed CAS Google Scholar Sherman WM, Lamb DR. Endocrine Research Unit, Mayo Clinic, Rochester, MN, USA. Lazar DFWiese RJBrady MJMastick CCWaters SBYamauchi KPessin JECuatrecasas PSaltiel AR Mitogen-activated protein kinase kinase inhibition does not block the stimulation of glucose utilization by insulin.

Video

Glycogen Synthesis - Glycogen Metabolism - Biochemistry The aim of this study was to determine the effect of glycogne antidiabetic agents on insulin-stimulated glycogen sjnthesis, as Enhancing glycogen synthesis synthesus on mRNA expression. After this, insulin-stimulated glycogen syhthesis Amino acid metabolism determined. mRNA sybthesis of the glucose Low-calorie diet and hydration GLUT1 and GLUT4, the Amino acid metabolism proliferator activator receptor gamma PPAR gamma glycofen 1 PGC1 and the myocyte-specific enhancer factors MEF2MEF2A, MEF2C and MEF2D were determined using real-time PCR analysis after 8 days exposure to the various antidiabetic agents. Insulin-stimulated glycogen synthesis was significantly increased in cultured human myotubes treated with insulin, rosiglitazone or metformin for 8 days, compared with non-treated cells. In contrast, treatment with AICAR was without effect on insulin-mediated glycogen synthesis. Metformin also increased mRNA expression of the MEF2 isoforms. Enhanced insulin-stimulated glycogen synthesis in human skeletal muscle cell culture coincides with increased GLUT4 and PGC1 mRNA expression following treatment with various antidiabetic agents. Enhancing glycogen synthesis

Author: Nekasa

4 thoughts on “Enhancing glycogen synthesis

  1. Meiner Meinung nach ist es das sehr interessante Thema. Ich biete Ihnen es an, hier oder in PM zu besprechen.

  2. Ich denke, dass Sie nicht recht sind. Geben Sie wir werden es besprechen. Schreiben Sie mir in PM, wir werden umgehen.

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com