Exercise in vivo marks human myotubes in vitro: Training-induced increase in lipid metabolism

Jenny Lund, Arild C Rustan, Nils G Løvsletten, Jonathan M Mudry, Torgrim M Langleite, Yuan Z Feng, Camilla Stensrud, Mari G Brubak, Christian A Drevon, Kåre I Birkeland, Kristoffer J Kolnes, Egil I Johansen, Daniel S Tangen, Hans K Stadheim, Hanne L Gulseth, Anna Krook, Eili T Kase, Jørgen Jensen, G Hege Thoresen, Jenny Lund, Arild C Rustan, Nils G Løvsletten, Jonathan M Mudry, Torgrim M Langleite, Yuan Z Feng, Camilla Stensrud, Mari G Brubak, Christian A Drevon, Kåre I Birkeland, Kristoffer J Kolnes, Egil I Johansen, Daniel S Tangen, Hans K Stadheim, Hanne L Gulseth, Anna Krook, Eili T Kase, Jørgen Jensen, G Hege Thoresen

Abstract

Background and aims: Physical activity has preventive as well as therapeutic benefits for overweight subjects. In this study we aimed to examine effects of in vivo exercise on in vitro metabolic adaptations by studying energy metabolism in cultured myotubes isolated from biopsies taken before and after 12 weeks of extensive endurance and strength training, from healthy sedentary normal weight and overweight men.

Methods: Healthy sedentary men, aged 40-62 years, with normal weight (body mass index (BMI) < 25 kg/m2) or overweight (BMI ≥ 25 kg/m2) were included. Fatty acid and glucose metabolism were studied in myotubes using [14C]oleic acid and [14C]glucose, respectively. Gene and protein expressions, as well as DNA methylation were measured for selected genes.

Results: The 12-week training intervention improved endurance, strength and insulin sensitivity in vivo, and reduced the participants' body weight. Biopsy-derived cultured human myotubes after exercise showed increased total cellular oleic acid uptake (30%), oxidation (46%) and lipid accumulation (34%), as well as increased fractional glucose oxidation (14%) compared to cultures established prior to exercise. Most of these exercise-induced increases were significant in the overweight group, whereas the normal weight group showed no change in oleic acid or glucose metabolism.

Conclusions: 12 weeks of combined endurance and strength training promoted increased lipid and glucose metabolism in biopsy-derived cultured human myotubes, showing that training in vivo are able to induce changes in human myotubes that are discernible in vitro.

Conflict of interest statement

Competing Interests: The authors report no conflicts of interests.

Figures

Fig 1. Effects of 12 weeks of…
Fig 1. Effects of 12 weeks of exercise on myotube fatty acid metabolism.
Satellite cells isolated from biopsies from m. vastus lateralis before and after 12 weeks of exercise were cultured and differentiated to myotubes. Oxidation, cell-associated (CA) radioactivity and lipid accumulation of [14C]oleic acid were measured, and total cellular uptake (CO2+CA), oxidation (CO2), fractional oxidation (CO2CO2+CA), and lipid accumulation were determined. (A) Lipid accumulation presented as cpm/μg protein. Values are presented as means ± SEM for all participants combined (n = 18). (B) Oleic acid oxidation and total cellular uptake presented as nmol/mg protein. Values are presented as means ± SEM for all participants combined (n = 18). (C) Fractional oleic acid oxidation. Values are presented as means ± SEM for all participants combined (n = 18). (D) Fatty acid metabolism relative to before exercise. Values are presented as means ± SEM for all participants combined (n = 18). (E) Lipid accumulation presented as cpm/μg protein in study group when separated by BMI. Values are presented as means ± SEM (n = 7 in the normal weight group and n = 11 in the overweight group). (F) Oleic acid oxidation and total cellular uptake presented as nmol/mg protein in study group when separated by BMI. Values are presented as means ± SEM (n = 7 in the normal weight group and n = 11 in the overweight group). (G) Fractional oleic acid oxidation in absolute values in study group when separated by BMI. Values are presented as means ± SEM (n = 7 in the normal weight group and n = 11 in the overweight group). (H) Fatty acid metabolism relative to before exercise in study group separated by BMI. Values are presented as means ± SEM (n = 7 in the normal weight group and n = 11 in the overweight group). *Statistically significant vs. before exercise (p < 0.05, linear mixed-model analysis, SPSS). #Statistically significant vs. normal weight group after exercise (p < 0.05, linear mixed-model analysis, SPSS). $Statistically significant vs. normal weight group (p < 0.05, linear mixed-model analysis, SPSS).
Fig 2. Effects of 12 weeks of…
Fig 2. Effects of 12 weeks of exercise on myotube glucose metabolism.
Satellite cells isolated from biopsies from m. vastus lateralis before and after 12 weeks of exercise were cultured and differentiated to myotubes. Oxidation and cell-associated (CA) radioactivity of [14C]glucose were measured, and total cellular uptake (CO2+CA), oxidation (CO2), and fractional oxidation (CO2CO2+CA) were determined. (A) Glucose oxidation and total cellular uptake presented as nmol/mg protein. Values are presented as means ± SEM for all participants combined (n = 18). (B) Fractional glucose oxidation. Values are presented as means ± SEM for all participants combined (n = 18). (C) Glucose metabolism relative to before exercise. Values are presented as means ± SEM for all participants combined (n = 18). *Statistically significant vs. before exercise (p < 0.05, linear mixed-model analysis, SPSS). (D) Glucose oxidation and total cellular uptake presented as nmol/mg protein in study group when separated by BMI. Values are presented as means ± SEM (n = 7 in the normal weight group and n = 11 in the overweight group). (E) Fractional glucose oxidation in absolute values in study group when separated by BMI. Values are presented as means ± SEM (n = 7 in the normal weight group and n = 11 in the overweight group). (F) Glucose metabolism relative to before exercise in study group when separated by BMI. Values are presented as means ± SEM (n = 7 in the normal weight group and n = 11 in the overweight group). *Statistically significant vs. before exercise (p < 0.05, linear mixed-model analysis, SPSS). $Statistically significant vs. normal weight group (p < 0.05, linear mixed-model analysis, SPSS). (G) Pearson’s test of correlation between exercise-induced changes in leg press and glucose oxidation in myotubes. Δ = after exercise–before exercise. Full line represents the regression line for all donors (n = 18, Pearson’s correlation coefficient, r = 0.52, and p = 0.03), whereas stapled line represents the regression line for the overweight group (n = 11, Pearson’s correlation coefficient, r = 0.68, and p = 0.02).
Fig 3. Effects of 12 weeks of…
Fig 3. Effects of 12 weeks of exercise on myotube AMPKα phosphorylation.
Satellite cells isolated from biopsies from m. vastus lateralis before and after 12 weeks of exercise were cultured and differentiated to myotubes. (A-C) AMPKα phosphorylation by immunoblotting. Protein was isolated and total AMPKα and pAMPKα expressions assessed by immunoblotting. A, one representative immunoblot. Bands selected from one membrane have been spliced together to show only relevant samples, as indicated by lines separating the spliced blots. B, quantified immunoblots for participants combined (n = 9) relative to before exercise. C, quantified immunoblots for study group when separated by BMI relative to normal weight before exercise (n = 5 in the normal weight group and n = 4 in the overweight group). Values are presented as means ± SEM. All samples were derived at the same time and processed in parallel.
Fig 4. Effects of 12 weeks of…
Fig 4. Effects of 12 weeks of exercise on mitochondria-related genes and proteins.
Satellite cells isolated from biopsies from m. vastus lateralis before and after 12 weeks of exercise were cultured and differentiated to myotubes. (A) mRNA expression of PPARGC1A, PDK4, CPT1A, and CYC1 after exercise relative to before exercise. mRNA was isolated and expression assessed by qPCR. All values were corrected for the housekeeping control GAPDH, and presented as means ± SEM for all participants combined (n = 18). (B) mRNA expression of PPARGC1A, PDK4, CPT1A, and CYC1 after exercise relative to before exercise in study group when separated by BMI. mRNA was isolated and expression assessed by qPCR. All values were corrected for the housekeeping control GAPDH, and presented as means ± SEM (n = 7 in the normal weight group and n = 11 in the overweight group). (C) Pearson’s test of correlation was performed between exercise-induced changes in visceral fat area and mRNA expression of PDK4 in myotubes. Δ = after exercise–before exercise. Full line represents the regression line for all donors (n = 18, Pearson’s correlation coefficient, r = -0.54, and p = 0.02), whereas stapled line represents the regression line for the overweight group (n = 11, Pearson’s correlation coefficient, r = -0.63, and p = 0.04). (D) DNA methylation of PPARGC1A, PDK4 and TFAM after exercise relative to before exercise. gDNA was isolated and bisulfite treated, and methylation assessed by immunoblotting. Values are presented as means ± SEM (n = 6). (E-G) OXPHOS complexes by immunoblotting. Protein was isolated and OXPHOS complexes assessed by immunoblotting. E, one representative immunoblot. F, quantified immunoblots of complex V for participants combined. All values were corrected for the housekeeping control α-tubulin, and presented as means ± SEM (n = 10). G, quantified immunoblots of complex V in study group when separated by BMI. All values were corrected for the housekeeping control α-tubulin, and presented as means ± SEM (n = 5 in each group).
Fig 5. Effects of 12 weeks of…
Fig 5. Effects of 12 weeks of exercise on myotube expression of lipid metabolism associated genes.
Satellite cells isolated from biopsies from m. vastus lateralis before and after 12 weeks of exercise were cultured and differentiated to myotubes. mRNA was isolated and expression assessed by qPCR. (A) mRNA expression after exercise relative to before exercise for all participants combined. All values were corrected for the housekeeping control GAPDH, and presented as means ± SEM (n = 18). (B) mRNA expression after exercise relative to before exercise for study group when separated by BMI. All values were corrected for the housekeeping control GAPDH, and presented as means ± SEM (n = 7 in the normal weight group and n = 11 in the overweight group).
Fig 6. Effects of 12 weeks of…
Fig 6. Effects of 12 weeks of exercise on myotube Akt phosphorylation, TBC1D4 phosphorylation and glycogen synthesis with or without 100 nmol/l insulin.
Satellite cells isolated from biopsies from m. vastus lateralis before and after 12 weeks of exercise were cultured and differentiated to myotubes. (A-C) Akt phosphorylation by immunoblotting. Protein was isolated and total Akt and pAkt expressions assessed by immunoblotting. A, one representative immunoblot. B, quantified immunoblots relative to basal before exercise for participants combined. Values are presented as means ± SEM (n = 9). C, quantified immunoblots relative to basal before exercise for study group when separated by BMI (n = 4 in the normal weight group and n = 5 in the overweight group). (A, D and E) TBC1D4 phosphorylation by immunoblotting. Protein was isolated and total TBC1D4 and pTBC1D4 expressions assessed by immunoblotting. A, one representative immunoblot. D, quantified immunoblots relative to basal before exercise for participants combined. Values are presented as means ± SEM (n = 10). E, quantified immunoblots relative to basal before exercise for study group when separated by BMI (n = 5 in both groups). All samples were derived at the same time and processed in parallel. (F) Glycogen synthesis relative to basal before exercise. Values are presented as means ± SEM (n = 5). Absolute values (range) representing 100%: Basal glycogen synthesis 3.9–15.4 nmol/mg protein. #Statistically significant vs. basal before exercise (p < 0.05, paired t test).
Fig 7. Effects of 12 weeks of…
Fig 7. Effects of 12 weeks of exercise on myotube IRS1 gene expression and IRS1 first exon DNA methylation.
(A)IRS1 mRNA expression after exercise relative to before exercise for participants combined. mRNA was isolated and expression assessed by qPCR. All values were corrected for the housekeeping control GAPDH, and presented as means ± SEM (n = 8). *Statistically significant vs. before exercise (p < 0.05, paired t test). (B)IRS1 mRNA expression after exercise relative to before exercise for study group when separated by BMI. mRNA was isolated and expression assessed by qPCR. All values were corrected for the housekeeping control GAPDH, and presented as means ± SEM (n = 3 in the normal weight group and n = 5 in the overweight group). *Statistically significant vs. before exercise (p < 0.05, paired t test). (C)IRS1 first exon DNA methylation after exercise relative to before exercise. gDNA was isolated and bisulfite treated, and methylation was assessed by pyrosequencing. Values are presented as means ± SEM (n = 6). *Statistically significant vs. before exercise (p < 0.05, paired t test). (D-F) IRS1 total protein expression. Protein was isolated and total IRS1 expression assessed by immunoblotting. D, one representative immunoblot. Bands selected from one membrane have been spliced together to show only relevant samples, as indicated by lines separating the spliced blots. E, quantified immunoblots relative to before exercise for participants combined. All values were corrected for the housekeeping control α-tubulin, and presented as means ± SEM (n = 9). G, quantified immunoblots relative to before exercise for study group when separated by BMI. All values were corrected for the housekeeping control α-tubulin, and presented as means ± SEM (n = 5 in the normal weight group and n = 4 in the overweight group). All samples were derived at the same time and processed in parallel.

References

    1. Ginter E, Simko V. Type 2 diabetes mellitus, pandemic in 21st century Diabetes: Springer; 2013. p. 42–50.
    1. Tuomilehto J, Lindström J, Eriksson JG, Valle TT, Hämäläinen H, Ilanne-Parikka P, et al. Prevention of type 2 diabetes mellitus by changes in lifestyle among subjects with impaired glucose tolerance. New England Journal of Medicine. 2001;344(18):1343–50. 10.1056/NEJM200105033441801
    1. Alberti KGM, Zimmet P, Shaw J. International Diabetes Federation: a consensus on Type 2 diabetes prevention. Diabetic Medicine. 2007;24(5):451–63. 10.1111/j.1464-5491.2007.02157.x
    1. James PT. Obesity: the worldwide epidemic. Clinics in dermatology. 2004;22(4):276–80. 10.1016/j.clindermatol.2004.01.010
    1. Smyth S, Heron A. Diabetes and obesity: the twin epidemics. Nature medicine. 2006;12(1):75–80. 10.1038/nm0106-75
    1. Turcotte LP, Fisher JS. Skeletal muscle insulin resistance: roles of fatty acid metabolism and exercise. Physical therapy. 2008;88(11):1279–96. 10.2522/ptj.20080018
    1. Kiens B. Skeletal muscle lipid metabolism in exercise and insulin resistance. Physiological Reviews. 2006;86(1):205–43. 10.1152/physrev.00023.2004
    1. Ramos-Jiménez A, Hernández-Torres RP, Torres-Durán PV, Romero-Gonzalez J, Mascher D, Posadas-Romero C, et al. The respiratory exchange ratio is associated with fitness indicators both in trained and untrained men: a possible application for people with reduced exercise tolerance. Clinical Medicine Insights Circulatory, Respiratory and Pulmonary Medicine. 2008;2:1.
    1. O'Gorman DJ, Krook A. Exercise and the treatment of diabetes and obesity. Endocrinology and metabolism clinics of North America. 2008;37(4):887–903. 10.1016/j.ecl.2008.07.006
    1. DeFronzo RA, Gunnarsson R, Björkman O, Olsson M, Wahren J. Effects of insulin on peripheral and splanchnic glucose metabolism in noninsulin-dependent (type II) diabetes mellitus. Journal of Clinical Investigation. 1985;76(1):149 10.1172/JCI111938
    1. Egan B, Zierath JR. Exercise metabolism and the molecular regulation of skeletal muscle adaptation. Cell metabolism. 2013;17(2):162–84. 10.1016/j.cmet.2012.12.012
    1. Mauro A. Satellite cell of skeletal muscle fibers. The Journal of biophysical and biochemical cytology. 1961;9(2):493–5.
    1. Blau HM, Webster C. Isolation and characterization of human muscle cells. Proceedings of the National Academy of Sciences. 1981;78(9):5623–7.
    1. Ling C, Groop L. Epigenetics: a molecular link between environmental factors and type 2 diabetes. Diabetes. 2009;58(12):2718–25. 10.2337/db09-1003
    1. Nitert MD, Dayeh T, Volkov P, Elgzyri T, Hall E, Nilsson E, et al. Impact of an exercise intervention on DNA methylation in skeletal muscle from first-degree relatives of patients with type 2 diabetes. Diabetes. 2012;61(12):3322–32. 10.2337/db11-1653
    1. Bourlier V, Saint-Laurent C, Louche K, Badin P-M, Thalamas C, de Glisezinski I, et al. Enhanced Glucose Metabolism Is Preserved in Cultured Primary Myotubes From Obese Donors in Response to Exercise Training. The Journal of Clinical Endocrinology & Metabolism. 2013;98(9):3739–47.
    1. Gaster M. Metabolic flexibility is conserved in diabetic myotubes. Journal of lipid research. 2007;48(1):207–17. 10.1194/jlr.M600319-JLR200
    1. Green C, Bunprajun T, Pedersen B, Scheele C. Physical activity is associated with retained muscle metabolism in human myotubes challenged with palmitate. The Journal of physiology. 2013;591(18):4621–35. 10.1113/jphysiol.2013.251421
    1. Aas V, Bakke SS, Feng YZ, Kase ET, Jensen J, Bajpeyi S, et al. Are cultured human myotubes far from home? Cell and tissue research. 2013;354(3):671–82. 10.1007/s00441-013-1655-1
    1. Kase ET, Feng YZ, Badin P-M, Bakke SS, Laurens C, Coue M, et al. Primary defects in lipolysis and insulin action in skeletal muscle cells from type 2 diabetic individuals. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids. 2015;1851(9):1194–201.
    1. Sharples AP, Stewart CE, Seaborne RA. Does skeletal muscle have an ‘epi’-memory? The role of epigenetics in nutritional programming, metabolic disease, aging and exercise. Aging cell. 2016.
    1. Ahlborg G, Felig P, Hagenfeldt L, Hendler R, Wahren J. Substrate turnover during prolonged exercise in man: splanchnic and leg metabolism of glucose, free fatty acids, and amino acids. Journal of Clinical Investigation. 1974;53(4):1080 10.1172/JCI107645
    1. Young D, Pelligra R, Shapira J, Adachi R, Skrettingland K. Glucose oxidation and replacement during prolonged exercise in man. Journal of applied physiology. 1967;23(5):734–41.
    1. Battaglia GM, Zheng D, Hickner RC, Houmard JA. Effect of exercise training on metabolic flexibility in response to a high-fat diet in obese individuals. American Journal of Physiology-Endocrinology and Metabolism. 2012;303(12):E1440–E5. 10.1152/ajpendo.00355.2012
    1. Berggren JR, Boyle KE, Chapman WH, Houmard JA. Skeletal muscle lipid oxidation and obesity: influence of weight loss and exercise. American Journal of Physiology-Endocrinology and Metabolism. 2008;294(4):E726–E32. 10.1152/ajpendo.00354.2007
    1. Langleite TM, Jensen J, Norheim F, Gulseth HL, Tangen DS, Kolnes KJ, et al. Insulin sensitivity, body composition and adipose depots following 12 w combined endurance and strength training in dysglycemic and normoglycemic sedentary men. Archives of Physiology and Biochemistry. 2016:1–13.
    1. Johansson L, Solvoll K, Opdahl S, Bjoerneboe G-EA, Drevon C. Response rates with different distribution methods and reward, and reproducibility of a quantitative food frequency questionnaire. European journal of clinical nutrition. 1997;51(6):346–53.
    1. Li Y, Lee S, Langleite T, Norheim F, Pourteymour S, Jensen J, et al. Subsarcolemmal lipid droplet responses to a combined endurance and strength exercise intervention. Physiological reports. 2014;2(11):e12187 10.14814/phy2.12187
    1. Henry RR, Abrams L, Nikoulina S, Ciaraldi TP. Insulin action and glucose metabolism in nondiabetic control and NIDDM subjects: comparison using human skeletal muscle cell cultures. Diabetes. 1995;44(8):936–46.
    1. Gaster M, Beck-Nielsen H, Schrøder H. Proliferation conditions for human satellite cells The fractional content of satellite cells. Apmis. 2001;109(11):726–34.
    1. Gaster M, Kristensen S, Beck-Nielsen H, Schrøder H. A cellular model system of differentiated human myotubes. Apmis. 2001;109(11):735–44.
    1. Wensaas A, Rustan A, Lövstedt K, Kull B, Wikström S, Drevon C, et al. Cell-based multiwell assays for the detection of substrate accumulation and oxidation. Journal of lipid research. 2007;48(4):961–7. 10.1194/jlr.D600047-JLR200
    1. Kase ET, Andersen B, Nebb HI, Rustan AC, Thoresen GH. 22-Hydroxycholesterols regulate lipid metabolism differently than T0901317 in human myotubes. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids. 2006;1761(12):1515–22.
    1. Hessvik NP, Bakke SS, Fredriksson K, Boekschoten MV, Fjørkenstad A, Koster G, et al. Metabolic switching of human myotubes is improved by n-3 fatty acids. Journal of lipid research. 2010;51(8):2090–104. 10.1194/jlr.M003319
    1. Hardie DG, Sakamoto K. AMPK: a key sensor of fuel and energy status in skeletal muscle. Physiology. 2006;21(1):48–60.
    1. McGee SL, Howlett KF, Starkie RL, Cameron-Smith D, Kemp BE, Hargreaves M. Exercise increases nuclear AMPK α2 in human skeletal muscle. Diabetes. 2003;52(4):926–8.
    1. Lira VA, Benton CR, Yan Z, Bonen A. PGC-1α regulation by exercise training and its influences on muscle function and insulin sensitivity. American Journal of Physiology-Endocrinology and Metabolism. 2010;299(2):E145–E61. 10.1152/ajpendo.00755.2009
    1. Koves TR, Sparks LM, Kovalik J, Mosedale M, Arumugam R, DeBalsi KL, et al. PPARγ coactivator-1α contributes to exercise-induced regulation of intramuscular lipid droplet programming in mice and humans. Journal of lipid research. 2013;54(2):522–34. 10.1194/jlr.P028910
    1. Baar K, Wende AR, Jones TE, Marison M, Nolte LA, Chen M, et al. Adaptations of skeletal muscle to exercise: rapid increase in the transcriptional coactivator PGC-1. The FASEB Journal. 2002;16(14):1879–86. 10.1096/fj.02-0367com
    1. Kerner J, Hoppel C. Fatty acid import into mitochondria. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids. 2000;1486(1):1–17.
    1. Ehrenborg E, Krook A. Regulation of skeletal muscle physiology and metabolism by peroxisome proliferator-activated receptor δ. Pharmacological Reviews. 2009;61(3):373–93. 10.1124/pr.109.001560
    1. Kulkarni SS, Salehzadeh F, Fritz T, Zierath JR, Krook A, Osler ME. Mitochondrial regulators of fatty acid metabolism reflect metabolic dysfunction in type 2 diabetes mellitus. Metabolism. 2012;61(2):175–85. 10.1016/j.metabol.2011.06.014
    1. Jäger S, Handschin C, Pierre JS-, Spiegelman BM. AMP-activated protein kinase (AMPK) action in skeletal muscle via direct phosphorylation of PGC-1α. Proceedings of the National Academy of Sciences. 2007;104(29):12017–22.
    1. Ljubicic V, Joseph A-M, Saleem A, Uguccioni G, Collu-Marchese M, Lai RY, et al. Transcriptional and post-transcriptional regulation of mitochondrial biogenesis in skeletal muscle: effects of exercise and aging. Biochimica et Biophysica Acta (BBA)-General Subjects. 2010;1800(3):223–34.
    1. Shaw CS, Sherlock M, Stewart PM, Wagenmakers AJ. Adipophilin distribution and colocalisation with lipid droplets in skeletal muscle. Histochemistry and cell biology. 2009;131(5):575–81. 10.1007/s00418-009-0558-4
    1. Bosma M, Hesselink MK, Sparks LM, Timmers S, Ferraz MJ, Mattijssen F, et al. Perilipin 2 improves insulin sensitivity in skeletal muscle despite elevated intramuscular lipid levels. Diabetes. 2012;61(11):2679–90. 10.2337/db11-1402
    1. Koonen DP, Glatz JF, Bonen A, Luiken JJ. Long-chain fatty acid uptake and FAT/CD36 translocation in heart and skeletal muscle. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids. 2005;1736(3):163–80.
    1. Zhang L, Keung W, Samokhvalov V, Wang W, Lopaschuk GD. Role of fatty acid uptake and fatty acid β-oxidation in mediating insulin resistance in heart and skeletal muscle. Biochimica et Biophysica Acta (BBA)-Molecular and Cell Biology of Lipids. 2010;1801(1):1–22.
    1. Feng YZ, Nikolić N, Bakke SS, Boekschoten MV, Kersten S, Kase ET, et al. PPARδ activation in human myotubes increases mitochondrial fatty acid oxidative capacity and reduces glucose utilization by a switch in substrate preference. Archives of physiology and biochemistry. 2014;120(1):12–21. 10.3109/13813455.2013.829105
    1. Kersten S, Mandard S, Tan NS, Escher P, Metzger D, Chambon P, et al. Characterization of the fasting-induced adipose factor FIAF, a novel peroxisome proliferator-activated receptor target gene. Journal of Biological Chemistry. 2000;275(37):28488–93. 10.1074/jbc.M004029200
    1. Mandard S, Zandbergen F, Tan NS, Escher P, Patsouris D, Koenig W, et al. The direct peroxisome proliferator-activated receptor target fasting-induced adipose factor (FIAF/PGAR/ANGPTL4) is present in blood plasma as a truncated protein that is increased by fenofibrate treatment. Journal of Biological Chemistry. 2004;279(33):34411–20. 10.1074/jbc.M403058200
    1. Staiger H, Haas C, Machann J, Werner R, Weisser M, Schick F, et al. Muscle-Derived Angiopoietin-Like Protein 4 Is Induced by Fatty Acids via Peroxisome Proliferator–Activated Receptor (PPAR)-δ and Is of Metabolic Relevance in Humans. Diabetes. 2009;58(3):579–89. 10.2337/db07-1438
    1. Boström PA, Graham EL, Georgiadi A, Ma X. Impact of exercise on muscle and nonmuscle organs. IUBMB life. 2013;65(10):845–50. 10.1002/iub.1209
    1. Feng YZ, Nikolić N, Bakke SS, Kase ET, Guderud K, Hjelmesæth J, et al. Myotubes from lean and severely obese subjects with and without type 2 diabetes respond differently to an in vitro model of exercise. American Journal of Physiology-Cell Physiology. 2015;308(7):C548–C56. 10.1152/ajpcell.00314.2014
    1. Nikolić N, Bakke SS, Kase ET, Rudberg I, Halle IF, Rustan AC, et al. Electrical pulse stimulation of cultured human skeletal muscle cells as an in vitro model of exercise. PLoS One. 2012;7(3).
    1. Perseghin G, Price TB, Petersen KF, Roden M, Cline GW, Gerow K, et al. Increased glucose transport–phosphorylation and muscle glycogen synthesis after exercise training in insulin-resistant subjects. New England Journal of Medicine. 1996;335(18):1357–62. 10.1056/NEJM199610313351804
    1. Wensaas AJ, Rustan AC, Just M, Berge RK, Drevon CA, Gaster M. Fatty Acid Incubation of Myotubes From Humans With Type 2 Diabetes Leads to Enhanced Release of β-Oxidation Products Because of Impaired Fatty Acid Oxidation. Diabetes. 2009;58(3):527–35. 10.2337/db08-1043
    1. Kase E, Thoresen G, Westerlund S, Højlund K, Rustan A, Gaster M. Liver X receptor antagonist reduces lipid formation and increases glucose metabolism in myotubes from lean, obese and type 2 diabetic individuals. Diabetologia. 2007;50(10):2171–80. 10.1007/s00125-007-0760-7
    1. Kase ET, Wensaas AJ, Aas V, Højlund K, Levin K, Thoresen GH, et al. Skeletal muscle lipid accumulation in type 2 diabetes may involve the liver X receptor pathway. Diabetes. 2005;54(4):1108–15.
    1. Bakke SS, Feng YZ, Nikolić N, Kase ET, Moro C, Stensrud C, et al. Myotubes from severely obese type 2 diabetic subjects accumulate less lipids and show higher lipolytic rate than myotubes from severely obese non-diabetic subjects. PloS one. 2015;10(3):e0119556 10.1371/journal.pone.0119556
    1. Gerhart-Hines Z, Rodgers JT, Bare O, Lerin C, Kim SH, Mostoslavsky R, et al. Metabolic control of muscle mitochondrial function and fatty acid oxidation through SIRT1/PGC-1α. The EMBO journal. 2007;26(7):1913–23. 10.1038/sj.emboj.7601633
    1. Wende AR, Huss JM, Schaeffer PJ, Giguere V, Kelly DP. PGC-1α coactivates PDK4 gene expression via the orphan nuclear receptor ERRα: a mechanism for transcriptional control of muscle glucose metabolism. Molecular and cellular biology. 2005;25(24):10684–94. 10.1128/MCB.25.24.10684-10694.2005
    1. Nikolić N, Rhedin M, Rustan AC, Storlien L, Thoresen GH, Strömstedt M. Overexpression of PGC-1α increases fatty acid oxidative capacity of human skeletal muscle cells. Biochemistry research international. 2011;2012.
    1. Reik W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature. 2007;447(7143):425–32. 10.1038/nature05918
    1. Barres R, Yan J, Egan B, Treebak JT, Rasmussen M, Fritz T, et al. Acute exercise remodels promoter methylation in human skeletal muscle. Cell metabolism. 2012;15(3):405–11. 10.1016/j.cmet.2012.01.001
    1. Jorge MLMP, de Oliveira VN, Resende NM, Paraiso LF, Calixto A, Diniz ALD, et al. The effects of aerobic, resistance, and combined exercise on metabolic control, inflammatory markers, adipocytokines, and muscle insulin signaling in patients with type 2 diabetes mellitus. Metabolism. 2011;60(9):1244–52. 10.1016/j.metabol.2011.01.006
    1. Stuart CA, South MA, Lee ML, McCurry MP, Howell ME, Ramsey MW, et al. Insulin responsiveness in metabolic syndrome after eight weeks of cycle training. Medicine and science in sports and exercise. 2013;45(11):2021 10.1249/MSS.0b013e31829a6ce8
    1. Nascimento EB, Riedl I, Jiang LQ, Kulkarni SS, Näslund E, Krook A. Enhanced glucose metabolism in cultured human skeletal muscle after Roux-en-Y gastric bypass surgery. Surgery for Obesity and Related Diseases. 2015;11(3):592–601. 10.1016/j.soard.2014.11.001
    1. DeFronzo RA. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes. 2009;58(4):773–95. 10.2337/db09-9028
    1. Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and missing links. Cell. 2012;148(5):852–71. 10.1016/j.cell.2012.02.017

Source: PubMed

3
Subskrybuj