Fuel for the Work Required: A Theoretical Framework for Carbohydrate Periodization and the Glycogen Threshold Hypothesis

Samuel G Impey, Mark A Hearris, Kelly M Hammond, Jonathan D Bartlett, Julien Louis, Graeme L Close, James P Morton, Samuel G Impey, Mark A Hearris, Kelly M Hammond, Jonathan D Bartlett, Julien Louis, Graeme L Close, James P Morton

Abstract

Deliberately training with reduced carbohydrate (CHO) availability to enhance endurance-training-induced metabolic adaptations of skeletal muscle (i.e. the 'train low, compete high' paradigm) is a hot topic within sport nutrition. Train-low studies involve periodically training (e.g., 30-50% of training sessions) with reduced CHO availability, where train-low models include twice per day training, fasted training, post-exercise CHO restriction and 'sleep low, train low'. When compared with high CHO availability, data suggest that augmented cell signalling (73% of 11 studies), gene expression (75% of 12 studies) and training-induced increases in oxidative enzyme activity/protein content (78% of 9 studies) associated with 'train low' are especially apparent when training sessions are commenced within a specific range of muscle glycogen concentrations. Nonetheless, such muscle adaptations do not always translate to improved exercise performance (e.g. 37 and 63% of 11 studies show improvements or no change, respectively). Herein, we present our rationale for the glycogen threshold hypothesis, a window of muscle glycogen concentrations that simultaneously permits completion of required training workloads and activation of the molecular machinery regulating training adaptations. We also present the 'fuel for the work required' paradigm (representative of an amalgamation of train-low models) whereby CHO availability is adjusted in accordance with the demands of the upcoming training session(s). In order to strategically implement train-low sessions, our challenge now is to quantify the glycogen cost of habitual training sessions (so as to inform the attainment of any potential threshold) and ensure absolute training intensity is not compromised, while also creating a metabolic milieu conducive to facilitating the endurance phenotype.

Conflict of interest statement

Conflict of Interest

Samuel Impey, Mark Hearris, Kelly Hammond, Jonathan Bartlett, Julien Louis, Graeme Close and James Morton declare that they have no conflicts of interest.

Funding

No sources of funding were used to assist in the preparation of this manuscript.

Figures

Fig. 1
Fig. 1
Schematic overview of the potential exercise-nutrient-sensitive cell signalling pathways regulating the enhanced mitochondrial adaptations associated with training with low CHO availability. (1) Reduced muscle glycogen enhances both AMPK and p38MAPK phosphorylation that results in (2) activation and translocation of PGC-1α and p53 to the mitochondria and nucleus. (3) Upon entry into the nucleus, PGC-1α co-activates additional transcription factors (i.e. NRF1/2) to increase the expression of COX subunits and Tfam, as well as autoregulating its own expression. In the mitochondria, PGC-1α co-activates Tfam to coordinate regulation of mtDNA, and induces expression of key mitochondrial proteins of the electron transport chain, e.g. COX subunits. Similar to PGC-1α, p53 also translocates to the mitochondria to modulate Tfam activity and mtDNA expression, and to the nucleus where it functions to increase expression of proteins involved in mitochondrial fission and fusion (DRP-1 and MFN-2) and electron transport chain proteins. (4) Exercising in conditions of reduced CHO availability increases adipose tissue and intramuscular lipolysis via increased circulating adrenaline concentrations. (5) The resulting elevation in FFA activates the nuclear transcription factor, PPARδ, to increase expression of proteins involved in lipid metabolism, such as CPT1, PDK4, CD36 and HSL. (6) However, consuming pre-exercise meals rich in CHO and/or CHO during exercise can downregulate lipolysis (thereby negating FFA-mediated signalling), as well as reducing both AMPK and p38MAPK activity, thus having negative implications for downstream regulators. (7) High-fat feeding can also modulate PPARδ signalling and upregulate genes with regulatory roles in lipid metabolism (and downregulate CHO metabolism), although high-fat diets may also reduce muscle protein synthesis via impaired mTOR-p70S6K signalling, despite feeding leucine-rich protein. 4EBP1 eukaryotic translation initiation factor 4E-binding protein 1, AMPK AMP-activated protein kinase, CHO carbohydrate, CD36 cluster of differentiation 36, COX cytochrome c oxidase, CPT1 carnitine palmitoyltransferase 1, Drp1 dynamin-related protein 1, FA fatty acid, FABP fatty acid binding protein, GLU glucose, GLUT4 glucose transporter type 4, HSL hormone-sensitive lipase, IMTG intramuscular triglycerides, LAT1 large neutral amino acid transporter, LEU leucine, Mfn2 mitofusion-2, mTORC1 mammalian target of rapamycin complex 1, p38MAPK p38 mitogen-activated protein kinase, p53 tumor protein 53,  p70S6K ribosomal protein S6 kinase, PDK4 pyruvate dehydrogenase kinase 4, PGC- peroxisome proliferator-activated receptor gamma coactivator 1-α, PPARδ peroxisome proliferator-activated receptor, Tfam mitochondrial transcription factor A
Fig. 2
Fig. 2
Overview of studies supporting the glycogen threshold hypothesis. Studies are categorized into those examining a cell signalling, b gene expression and c muscle contractile capacity and post-exercise signalling. In a and b, the green bars represent the trial within the specific study that has been completed with high muscle glycogen, and the red bars represent the trial completed with low muscle glycogen. The length of the bar in both instances corresponds to the pre- and post-exercise muscle glycogen concentration. Additionally, in studies from the authors’ laboratory (Bartlett et al. [38] and Impey et al. [48]), black and white circles represent individual subjects’ pre- and post-exercise muscle glycogen concentrations, respectively. In c, a variety of CHO manipulation protocols have been adopted to examine the effect of high (green bars) and low (red bars) muscle glycogen concentration on contractile properties and post-exercise cell signalling. The shaded area represents a potential muscle glycogen threshold in which exercise should be commenced (albeit specific to the training status of the participants studied in these investigations). AMPK AMP-activated protein kinase, ACC acetyl-CoA carboxylase, Ca2+ calcium, COX cytochrome c oxidase, p38MAPK p38 mitogen-activated protein kinase, p70S6K ribosomal protein S6 kinase, PDK4 pyruvate dehydrogenase kinase 4, PGC- peroxisome proliferator-activated receptor gamma coactivator 1-α, Tfam mitochondrial transcription factor A
Fig. 3
Fig. 3
Muscle glycogen utilization according to studies incorporating varied exercise intensity, duration, and pre-exercise muscle glycogen concentration. Such data illustrate how the pattern of glycogen use can vary (according to the interactive effects of the aforementioned parameters) and how this should be considered in relation to the proposed glycogen threshold (shaded area). Data represent a sampling from studies compiled from cycling exercise protocols only and represent glycogen use in the vastus lateralis muscle

References

    1. Thomas DT, Erdman KA, Burke LM. American college of sports medicine joint position statement. nutrition and athletic performance. Med Sci Sports Exerc. 2016;48:543–568.
    1. Bergstrom J, Hermansen L, Hultman E, Saltin B. Diet, muscle glycogen and physical performance. Acta Physiol Scand. 1967;71:140–150. doi: 10.1111/j.1748-1716.1967.tb03720.x.
    1. Bergstrom J, Hultman E. Muscle glycogen synthesis after exercise: an enhancing factor localized to the muscle cells in man. Nature. 1966;16:309–310. doi: 10.1038/210309a0.
    1. Bergstrom J, Hultman E. The effect of exercise on muscle glycogen and electrolytes in normals. Scand J Clin Lab Invest. 1966;18:16–20. doi: 10.3109/00365516609065602.
    1. Hermansen L, Hultman E, Saltin B. Muscle glycogen during prolonged severe exercise. Acta Physiol Scand. 1967;71:129–139. doi: 10.1111/j.1748-1716.1967.tb03719.x.
    1. Sherman WM, Costill DL, Fink WJ, Miller JM. Effect of exercise-diet manipulation on muscle glycogen and its subsequent utilization during performance. Int J Sports Med. 1981;2:114–118. doi: 10.1055/s-2008-1034594.
    1. Sherman WM, Wimer GS. Insufficient dietary carbohydrate during training? Does it impair athletic performance? Int J Sport Nutr. 1991;1:28–44. doi: 10.1123/ijsn.1.1.28.
    1. Sherman WM, Doyle JA, Lamb DR, Strauss RH. Dietary carbohydrate, muscle glycogen, and exercise performance during 7 days of training. Am J Clin Nutr. 1993;57:27–31. doi: 10.1093/ajcn/57.1.27.
    1. Hansen AK, Fischer CP, Plomgaard P, Andersen JL, Saltin B, Pedersen BK. Skeletal muscle adaptation: training twice every second day vs. training once daily. J Appl Physiol. 2005;98:93–99. doi: 10.1152/japplphysiol.00163.2004.
    1. Stellingwerff T. Case Study: nutrition and training periodization in three elite marathon runners. Int J Sport Nutri Exerc Metab. 2012;22:392–400. doi: 10.1123/ijsnem.22.5.392.
    1. Hawley JA, Morton JP. Ramping up the signal: promoting endurance training adaptation in skeletal muscle by nutritional manipulation. Clin Exp Pharmacol Physiol. 2014;41:608–613. doi: 10.1111/1440-1681.12246.
    1. Bartlett JD, Hawley JA, Morton JP. Carbohydrate availability and exercise training adaptation: too much of a good thing? Eur J Sport Sci. 2015;15:3–12. doi: 10.1080/17461391.2014.920926.
    1. Coyle EF, Coggan AR, Hemmert MK, Ivy JL. Muscle glycogen utilization during strenuous exercise when fed carbohydrate. J Appl Physiol. 1986;61:165–172. doi: 10.1152/jappl.1986.61.1.165.
    1. Wildrick JJ, Costill DL, Fink WJ, Hickey MS, McConnell GK, Tanaka H. Carbohydrate feedings and exercise performance: effect of initial muscle glycogen concentration. J Appl Physiol. 1993;74:2998–3005. doi: 10.1152/jappl.1993.74.6.2998.
    1. Pilegaard H, Keller C, Steensberg A, Helge JW, Pedersen BK, Saltin B, Neufer PD. Influence of pre-exercise muscle glycogen content on exercise-induced transcriptional regulation of metabolic genes. J Physiol. 2002;541:261–271. doi: 10.1113/jphysiol.2002.016832.
    1. Pilegaard H, Osada T, Andersen LT, Helge JW, Saltin B, Neufer PD. Substrate availability and transcriptional regulation of metabolic genes in human skeletal muscle during recovery from exercise. Metabolism. 2005;54:1048–1055. doi: 10.1016/j.metabol.2005.03.008.
    1. Yeo WK, Paton CD, Garnham AP, Burke LM, Carey AL, Hawley JA. Skeletal muscle adaptation and performance responses to once versus twice every second day endurance training regimens. J Appl Physiol. 2008;105:1462–1470. doi: 10.1152/japplphysiol.90882.2008.
    1. Morton JP, Croft L, Bartlett JD, MacLaren DP, Reilly T, Evans L, McArdle A, Drust B. Reduced carbohydrate availability does not modulate training-induced heat shock protein adaptations but does up regulate oxidative enzyme activity in human skeletal muscle. J Appl Physiol. 2009;106:1513–1521. doi: 10.1152/japplphysiol.00003.2009.
    1. Hulston CJ, Venables MC, Mann CH, Martin C, Philp A, Baar K, Jeukendrup AE. Training with low muscle glycogen enhances fat metabolism in well-trained cyclists. Med Sci Sports Exerc. 2010;42:2046–2055. doi: 10.1249/MSS.0b013e3181dd5070.
    1. Cochran AJ, Myslik F, Maclnnis MJ, Percival ME, Bishop D, Tarnopolsky MA, Gibala MJ. Manipulating carbohydrate availability between twice-daily sessions of high-intensity interval training over 2 weeks improves time-trial performance. Int J Sport Nutr Exerc Metab. 2015;25:463–470. doi: 10.1123/ijsnem.2014-0263.
    1. Steinberg GR, Watt MJ, McGee SL, Chan S, Hargreaves M, Febbraio MA, Stapleton D, Kemp BE. Reduced glycogen availability is associated with increased AMPKalpha2 activity, nuclear AMPKalpha2 protein abundance, and GLUT4 mRNA expression in contracting human skeletal muscle. Appl Physiol Nutr Metab. 2006;31:302–312. doi: 10.1139/h06-003.
    1. Cochran AJ, Little JP, Tarnopolsky MA, Gibala MJ. Carbohydrate feeding during recovery alters the skeletal muscle metabolic response to repeated sessions of high-intensity interval exercise in humans. J Appl Physiol. 2010;108:628–636. doi: 10.1152/japplphysiol.00659.2009.
    1. Yeo WK, McGee SL, Carey AL, Paton CD, Garnham AP, Hargreaves M, Hawley JA. Acute signalling responses to intense endurance training commenced with low or normal muscle glycogen. Exp Physiol. 2010;95:351–358. doi: 10.1113/expphysiol.2009.049353.
    1. Psilander N, Frank P, Flockhart M, Sahlin K. Exercise with low glycogen increases PGC-1a gene expression in human skeletal muscle. Eur J Appl Physiol. 2013;113:951–963. doi: 10.1007/s00421-012-2504-8.
    1. Horowitz JF, Mora-Rodriguez R, Byerley LO, Coyle EF. Lipolytic suppression following carbohydrate ingestion limits fat oxidation during exercise. Am J Physiol. 1997;273:768–775.
    1. Arkinstall MJ, Bruce CR, Nikolopoulos V, Garnham AP, Hawley JA. Effect of carbohydrate ingestion on metabolism during running and cycling. J Appl Physiol. 2001;91:2125–2134. doi: 10.1152/jappl.2001.91.5.2125.
    1. Akerstrom TCA, Birk JB, Klein DK, Erikstrup C, Plomgaard P, Pedersen BK, Wojtaszewski J. Oral glucose ingestion attenuates exercise-induced activation of 5′-AMP-activated protein kinase in human skeletal muscle. Biochem Biophys Res Commun. 2006;342:949–955. doi: 10.1016/j.bbrc.2006.02.057.
    1. Civitarese AE, Hesselink MK, Russell AP, Ravussin E, Schrauwen P. Glucose ingestion during exercise blunts exercise-induced gene expression of skeletal muscle fat oxidative genes. Am J Physiol Endocrinol Metab. 2005;289:1023–1029. doi: 10.1152/ajpendo.00193.2005.
    1. Cluberton LJ, McGee SL, Murphy RM, Hargreaves M. Effect of carbohydrate ingestion on exercise-induced alterations in metabolic gene expression. J Appl Physiol. 2005;99:1359–1363. doi: 10.1152/japplphysiol.00197.2005.
    1. Van Proeyen K, Szlufcik K, Nielens H, Ramaekers M, Hespel P. Beneficial metabolic adaptations due to endurance exercise training in the fasted state. J Appl Physiol. 2011;110:236–245. doi: 10.1152/japplphysiol.00907.2010.
    1. De Bock K, Derave W, Eijnde BO, Hesselink MK, Koninckx E, Rose AJ, Schrauwen P, Bonen A, Richter EA, Hespel P. Effect of training in the fasted state on metabolic responses during exercise with carbohydrate intake. J Appl Phyiol. 2008;104:1045–1055. doi: 10.1152/japplphysiol.01195.2007.
    1. Nybo L, Pedersen K, Christensen B, Aagaard P, Brandt N, Kiens B. Impact of carbohydrate supplementation during endurance training on glycogen storage and performance. Acta Physiol. 2009;197:117–127. doi: 10.1111/j.1748-1716.2009.01996.x.
    1. Impey SG, Smith D, Robinson AL, Owens DJ, Bartlett JD, Smith K, Limb M, Tang J, Fraser WD, Close GL, Morton JP. Leucine-enriched protein feeding does not impair exercise-induced free fatty acid availability and lipid oxidation: beneficial implications for training in carbohydrate-restricted states. Amino Acids. 2015;47:407–416. doi: 10.1007/s00726-014-1876-y.
    1. Taylor C, Bartlett JD, van de Graaf CS, Louhelainen J, Coyne V, Iqbal Z, MacLaren DP, Gregson W, Close GL, Morton JP. Protein ingestion does not impair exercise-induced AMPK signalling when in a glycogen-depleted state: implications for train-low compete-high. Eur J Appl Physiol. 2013;113:1457–1468. doi: 10.1007/s00421-012-2574-7.
    1. Hulston CJ, Wolsk E, Grøndahl TS, Yfanti C, Van Hall G. Protein intake does not increase vastus lateralis muscle protein synthesis during cycling. Med Sci Sports Exerc. 2011;43:1635–1642. doi: 10.1249/MSS.0b013e31821661ab.
    1. Wojtaszewski JF, MacDonald C, Nielsen JN, Hellsten Y, Hardie DG, Kemp BE, Kiens B, Richter EA. Regulation of 5′AMP-activated protein kinase activity and substrate utilization in exercising human skeletal muscle. Am J Physiol Endocrinol Metab. 2003;284:E812–E822. doi: 10.1152/ajpendo.00436.2002.
    1. Chan S, McGee SL, Watt MJ, Hargreaves M, Febbraio MA. Altering dietary nutrient intake that reduces glycogen content leads to phosphorylation of nuclear p38 MAP kinase in human skeletal muscle: association with IL-6 gene transcription during contraction. FASEB J. 2004;18:1785–1787. doi: 10.1096/fj.03-1039fje.
    1. Bartlett JD, Louhelainen J, Iqbal Z, Cochran AJ, Gibala MJ, Gregson W, Close GL, Drust B, Morton JP. Reduced carbohydrate availability enhances exercise-induced p53 signalling in human skeletal muscle: implications for mitochondrial biogenesis. Am J Physiol Regul Integr Comp Physiol. 2013;304:450–458. doi: 10.1152/ajpregu.00498.2012.
    1. Lane SC, Camera DM, Lassiter DG, Areta JL, Bird SR, Yeo WK, Jeacocke NA, Krook A, Zierath JR, Burke LM, Hawley JA. Effects of sleeping with reduced carbohydrate availability on acute training responses. J Appl Physiol. 2015;119:643–655. doi: 10.1152/japplphysiol.00857.2014.
    1. Marquet LA, Brisswalter J, Louis J, Tiollier E, Burke LM, Hawley JA, Hausswirth C. Enhanced endurance performance by periodization of carbohydrate intake: “Sleep low” strategy. Med Sci Sports Exerc. 2016;48:663–672. doi: 10.1249/MSS.0000000000000823.
    1. Marquet LA, Hausswirth C, Molle O, Hawley JA, Burke LM, Tiollier E, Brisswalter J. Periodization of carbohydrate intake: short-term effect on performance. Nutrients. 2016;8:755. doi: 10.3390/nu8120755.
    1. Zbinden-Foncea H, van Loon LJ, Raymackers JM, Francaux M, Deldicque L. Contribution of nonesterified fatty acids to mitogen-activated protein kinase activation in human skeletal muscle during endurance exercise. Int J Sport Nutr Exerc Metab. 2013;23:201–209. doi: 10.1123/ijsnem.23.3.201.
    1. Hammond KM, Impey SG, Currell K, Mitchell N, Shepherd SO, Jeromson S, Hawley JA, Close GL, Hamilton LD, Sharples AP, Morton JP. Postexercise high-fat feeding suppresses p70S6K1 activity in human skeletal muscle. Med Sci Sport Exerc. 2016;48:2108–2117. doi: 10.1249/MSS.0000000000001009.
    1. Stellingwerff T, Spriet LL, Watt MJ, Kimber NE, Hargreaves M, Hawley JA, Burke LM. Decreased PDH activation and glycogenolysis during exercise following fat adaptation with carbohydrate restoration. Am J Physiol Endocrinol Metab. 2006;290:380–388. doi: 10.1152/ajpendo.00268.2005.
    1. Burke LM, Ross ML, Garvican-Lewis LA, Welvaert M, Heikura IA, Forbes SG, Mirtschin JG, Cato LE, Strobel N, Sharma AP, Hawley JA. Low carbohydrate, high fat diet impairs exercise economy and negates the performance benefit from intensified training in elite race walkers. J Physiol. 2017;595:2785–2807. doi: 10.1113/JP273230.
    1. Stephens FB, Chee C, Wall BT, Murton AJ, Shannon CE, van Loon LJ, Tsintzas K. Lipid-induced insulin resistance is associated with an impaired skeletal muscle protein synthetic response to amino acid ingestion in healthy young men. Diabetes. 2015;64:1615–1620. doi: 10.2337/db14-0961.
    1. Vogt S, Heinrich L, Schumacher YO, Grosshauser M, Blum A, König D, Berg A, Schmid A. Energy intake and energy expenditure of elite cyclists during preseason training. Int J Sports Med. 2005;26:701–706. doi: 10.1055/s-2004-830438.
    1. Impey SG, Hammond KM, Shepherd SO, Sharples AP, Stewart C, Limb M, Smith K, Philp A, Jeromson S, Hamilton DL, Close GL, Morton JP. Fuel for the work required: a practical approach to amalgamating train-low paradigms for endurance athletes. Physiol Rep. 2016;4:e12803. doi: 10.14814/phy2.12803.
    1. Lee-Young RS, Palmer MJ, Linden KC, LePlastrier K, Canny BJ, Hargreaves M, Wadley GD, Kemp BE, McConell GK. Carbohydrate ingestion does not alter skeletal muscle AMPK signalling during exercise in humans. Am J Physiol Endocrinol Metab. 2006;291:566–573. doi: 10.1152/ajpendo.00023.2006.
    1. Ørtenblad N, Nielsen J, Saltin B, Holmberg H-C. Role of glycogen availability in sarcoplasmic reticulum Ca2+ kinetics in human skeletal muscle. J Physiol. 2011;589:711–25.
    1. Duhamel TA, Perco JG, Green HJ. Manipulation of dietary carbohydrates after prolonged effort modifies muscle sarcoplasmic reticulum responses in exercising males. Am J Physiol Regul Integr Comp Physiol. 2006;291:1100–1110. doi: 10.1152/ajpregu.00858.2005.
    1. Gejl KD, Hvid LG, Frandsen U, Jensen K, Sahlin K, Ørtenblad N. Muscle glycogen content modifies SR Ca2+ release rate in elite endurance athletes. Med Sci Sports Exerc. 2014;46:496–505. doi: 10.1249/MSS.0000000000000132.
    1. Costa RJ, Jones GE, Lamb KL, Coleman R, Williams JH. The effects of a high carbohydrate diet on cortisol and salivary immunoglobulin A (s-IgA) during a period of increase exercise workload amongst Olympic and Ironman triathletes. Int J Sports Med. 2005;26:880–886. doi: 10.1055/s-2005-837467.
    1. Jensen L, Gejl KD, Ørtenblad N, Nielsen JL, Bech RD, Nygaard T, Sahlin K, Frandsen U. Carbohydrate restricted recovery from long term endurance exercise does not affect gene responses involved in mitochondrial biogenesis in highly trained athletes. Physiol Rep. 2015;12:e121814.
    1. Gejl KD, Thams L, Hansen M, Rokkedal-Lausch T, Plomgaard P, Nybo L, Larsen FJ, Cardinale DA, Jensen K, Holmberg HC, Vissing K, Ørtenblad N. No superior adaptations to carbohydrate periodization in elite endurance athletes. Med Sci Sports Exerc. 2017;49(12):2486–97.
    1. Lane SC, Areta JL, Bird SR, Coffey VG, Burke LM, Desbrow B, Karagounis LG, Hawley JA. Caffeine ingestion and cycling power output in a low or normal muscle glycogen state. Med Sci Sports Exerc. 2013;45:1577–1584. doi: 10.1249/MSS.0b013e31828af183.
    1. Gollnick PD, Piehl K, Saltin B. Selective glycogen depletion pattern in human skeletal muscle fibres after exercise of varying intensity and at varying pedalling rates. J Physiol. 1974;241:45–57. doi: 10.1113/jphysiol.1974.sp010639.
    1. De Bock K., Derave W., Ramaekers M., Richter E. A., Hespel P. Fiber type-specific muscle glycogen sparing due to carbohydrate intake before and during exercise. Journal of Applied Physiology. 2007;102:183–188. doi: 10.1152/japplphysiol.00799.2006.

Source: PubMed

3
订阅