Skip to main content
Diabetes Technology & Therapeutics logoLink to Diabetes Technology & Therapeutics
editorial
. 2014 Jun 1;16(6):331–337. doi: 10.1089/dia.2014.0097

Exercise, Hypoglycemia, and Type 1 Diabetes

Rita Basu 1, Matthew L Johnson 1, Yogish C Kudva 1, Ananda Basu 1,
PMCID: PMC4029043  PMID: 24811269

Management of patients with type 1 diabetes (T1D) during and after exercise continues to be a challenge for care providers and patients alike in terms of approach both to insulin therapy and to hypoglycemia prevention and rescue. These issues pose significant limitations from the providers' standpoint of hypoglycemia prevention and from the patients' standpoint of seeking to achieve desirable level of fitness to improve general metabolic and cardiovascular health. The purpose of this editorial therefore is (1) to summarize available knowledge of carbohydrate physiology during and after exercise in healthy and T1D individuals, (2) to elucidate possible physiological mechanisms predisposing to hypoglycemia, (3) to describe current approaches to prevent and manage exercise-induced hypoglycemia in T1D, and (4) to identify knowledge gaps in our understanding of exercise physiology in T1D that require further study.

Physical Activity, Glucose Physiology, and Hypoglycemia

Blood glucose concentration at any given time point is a reflection of the net balance between the rates of glucose appearance (Ra) into and disappearance (Rd) from the circulation. Therefore, when blood glucose concentrations are rising (immediate postprandial state), by definition, Ra (glucose appearing from the meal+rates of endogenous glucose production [EGP]) must exceed Rd (glucose uptake into tissues such as liver, muscle, and fat cells). Likewise, when blood glucose concentrations are falling, by definition, Rd (glucose uptake) must exceed Ra (either from a meal and/or EGP). It is well established that exercise increases muscle glucose uptake through insulin-dependent and insulin-independent mechanisms and that EGP must increase to meet the increased metabolic demands of the exercising muscle to prevent hypoglycemia.1–4 These changes in glucose fluxes (Table 1) are facilitated by changes in (1) hormonal concentrations (i.e., falling insulin and rising glucagon and catecholamine levels during exercise5), (2) blood flow to exercising muscles,6 and (3) adaptations to intracellular pathways that facilitate glucose uptake.7–10

Table 1.

Effects of Exercise in Healthy Individuals

  Result Mechanism
During exercise
Muscle contraction Insulin-independent translocation of GLUT4, resulting in increased glucose uptake • Ca2+/CaMK activation
• AMPK activation
• PKC activation
Muscle energetic demands Increases glucose disposal • Increased glycolytic flux
Endogenous glucose production Increases blood glucose availability • Increased glucagon concentration
• Increased catecholamine concentration
Muscle blood flow Increases glucose delivery • Elevated cardiac output
• Increased capillary blood flow
• Skeletal muscle vasodilation
After exercise
Insulin sensitivity Leftward shift of insulin response curve for 24–48 h after cessation of exercise • Currently unknown, possibly involving phosphorylation by AMPK of TBC1D1 and AS160

Skeletal muscle glucose uptake increases dramatically in response to exercise together with a drop in plasma insulin concentrations in healthy individuals to overnight fasted levels. During exercise, the increase in glucose uptake is the result of muscle contraction that activates the Ca2+/calmodulin-activated protein kinase (CaMK) family, 5'-AMP-activated protein kinase (AMPK), and atypical protein kinase C (PKC). Exercise also increases the energetic demands of skeletal muscle, resulting in increased glycolytic flux and demand for glucose as an energy substrate. Endogenous glucose production increases, making blood glucose more available as a result of elevations in circulating glucagon and catecholamine concentrations. Finally, increased skeletal muscle blood flow as a result of elevations in cardiac output and increased capillary blood flow together with local vasodilation increase glucose delivery to active tissues. After exercise, a leftward shift of the insulin dose–response curve persists for 24–48 h after the cessation of exercise through a currently unknown mechanism that increases the sensitivity of skeletal muscle to plasma insulin concentration.

GLUT4, glucose transporter type 4.

The rise in EGP during exercise was observed to be due to increases in both gluconeogenesis and glycogenolysis in a series of elegant experiments in intact and pancreatectomized dogs.3,11–13 We have recently demonstrated14 that in recreationally active healthy individuals without diabetes, a 60-min bout of moderate-intensity exercise (50% of maximal oxygen uptake) 2 h after a mixed meal increased EGP eightfold, which was facilitated by a doubling of plasma glucagon concentrations. In this study, despite the fact that plasma insulin concentrations rapidly declined to fasting levels during exercise, eight of 12 subjects had biochemical but asymptomatic hypoglycemia within 15 min after starting exercise despite consuming a 600-kcal mixed meal for breakfast containing 75 g of carbohydrates, 2 h prior to the start of exercise. When these data are taken together, the falling glucose concentrations during exercise was due to the fact that the combined effect of meal glucose appearance and EGP (Ra) was unable to keep pace with exercise-induced changes in whole-body glucose uptake (Rd).

There have also been prior publications that have reported on the effect(s) of exercise of varying intensity on glucose physiology. These have included ground-breaking work by exercise physiologists using isotope dilution techniques and glucose clamps. In healthy adults, Friedlander et al.15,16 demonstrated that Rd was directly proportional to exercise intensity and that although training did not affect the magnitude of Rd when expressed as relative exercise intensity, it did decrease Rd for a given power output, implying improved training-induced efficiency of the working muscles. Furthermore, the increased efficiency of Rd was related to up-regulation of muscle glycogen synthesis secondary to increased stimulation of glucose transport across the skeletal muscle cell membrane.17

Interesting gender differences in glucose kinetics and hormonal responses during exercise have also been observed,16,18 with women shifting substrate oxidation from carbohydrates to lipids and showing lesser rises in post-exercise glucagon and epinephrine concentrations than men at submaximal workloads common to endurance levels and daily activity. Likewise, during endurance exercise and exercise training, women utilize lipids more than carbohydrates when compared at the same relative exercise intensity with equally trained men.19,20

In contrast, the effect size of menstrual cycle on exercise-related substrate metabolism is modest at best. In an early study, the effects of menstrual phases on glucose turnover during moderate- and high-intensity exercise were negligible in healthy young women.21 It is interesting that oral contraceptive use led to reduced rates of glucose turnover during moderate-intensity exercise without altering rates of carbohydrate and lipid oxidation in healthy young women.22 Further pioneering studies by the same investigative group demonstrated that although exercise-induced lipolysis, as measured by rate of appearance of glycerol, was not affected by menstrual cycles, use of oral contraceptives increased triglyceride mobilization via increased lipolysis during moderate-intensity exercise.23 Confirmation that these observations on gender differences of substrate metabolism during exercise were related to estrogen came from a fascinating study in men that revealed that 8 days of estrogen supplementation resulted in reductions in glucose turnover and respiratory exchange ratio during exercise, thus implying fuel shift from carbohydrate to lipid oxidation.24

Exercise and “Insulin Sensitivity”

Insulin sensitivity, in the purist term, is defined as the ability of insulin to stimulate uptake of glucose by target tissues (viz., muscle, liver, adipose tissue). Hence, by definition, an increase in insulin sensitivity implies a shift of the dose–response curve of insulin to the left such that a lower concentration of insulin results in an equivalent amount of glucose uptake by the target tissue(s). Although some studies had suggested that insulin was indeed required and had a permissive effect on muscle glucose uptake during and after exercise,25,26 others demonstrated similar findings on glucose uptake in the muscle both during and after exercise even in the absence of insulin.27–29 Thereafter, several studies have demonstrated that muscle contractions, per se, induce muscle glucose uptake even in the absence of insulin7,30–32 and that the effects of insulin and contractions on muscle glucose transport are additive.7,33–35 Table 1 summarizes some of the physiological adaptations to exercise as related to carbohydrate metabolism.

Mechanical factors that modulate muscle glucose uptake include, among others, tissue permeability to glucose and changes in blood flow to exercising muscles. In an elegant and pioneering series of experiments in isolated frog muscle, Hollozsy and Narahara27 demonstrated that both muscle contractions and insulin separately stimulated muscle glucose uptake, presumably through a common transport pathway. However, further mechanistic studies in isolated rat skeletal muscle revealed that effects of insulin and muscle contractions on glucose uptake were additive but caused glucose transporter type 4 (GLUT4) translocation via independent pathways in contracting skeletal muscle.7–10 It is noteworthy that although insulin effects on glucose transport showed a threshold and saturable kinetics, isometric exercise increased glucose transport in a linear fashion without a threshold effect.

A single endurance exercise bout increased insulin action on skeletal muscle36,37 predominately because of an increase in GLUT4 protein content.38–40 Consecutive exercise sessions significantly increased muscle GLUT4 content, and the increase was proportional to the increase in insulin sensitivity of the muscle.41 Exercise training also increased the amount of GLUT4 that translocated to the skeletal muscle surface in response to a given insulin dosage.42 These exercise-induced adaptations result in improved skeletal muscle insulin sensitivity in previously healthy, but sedentary, humans.43 Intracellular modulators of GLUT4 translocation in contracting human skeletal muscle include kinases (5'-AMP-activated protein kinase, Akt) and the Rab GTPase-activating proteins TBC1D1 and AS106.44,45 These mediators are also involved in the increase in post-exercise insulin-independent changes to glucose uptake that last for 24–48 h after an exercise bout.46 Taken together, it appears that an increase in insulin sensitivity that occurs in a trained muscle is related to training-induced changes in GLUT4 availability to the cell surface as a result of activating intracellular signaling pathways specific to muscle contraction, which further promotes muscle glucose uptake and shifts the insulin dose–response curve to the left.

Muscle blood flow, a factor beside blood glucose concentration that determines delivery to skeletal muscle, markedly increases during exercise. During light to moderate exercise, contracting skeletal muscle blood flow increases to 6–10 L/min in healthy adults, whereas vigorous activity demands over 12 L/min.6,47 This hemodynamic effect results in more glucose being delivered to active skeletal muscle together with a greater surface area for exchange. However, estimates are that the relative importance of blood flow alone (without skeletal muscle contraction) likely accounts for less than 30% of the increase in muscle glucose uptake during exercise.48

Strategies to prevent exercise-induced hypoglycemia could prolong exercise ability and delay exhaustion. In a series of experiments, various investigators49,50 demonstrated that carbohydrate feeding and water ingestion during prolonged exercise delay fatigue and increase neuromuscular power, at least in part by preventing hypoglycemia, by avoiding muscle glycogen depletion. A similar effect of carbohydrate ingestion on cycling time trial performance was also reported.51 Using the glucose clamp technique during intense exercise, Coyle et al.52 further demonstrated that although hyperglycemia did not alter muscle glycogen use, it increased muscle glucose uptake and oxidation. In contrast, recently, Fahey et al.53 demonstrated that a short sprint increased plasma glucose levels owing to a decline in Rd in both healthy and T1D individuals.

Intermittent Exercise and Glucose Physiology

In the free-living condition, humans frequently participate in sports activities (e.g., tennis, soccer, basketball, etc.) that involve intermittent, short bursts of moderate- to high-intensity exercise, interspersed with relative inactivity. Systematic assessments and comparisons of glucose turnover and insulin sensitivity during intermittent versus sustained exercise have been sparse in people with and without diabetes. Early investigations54 did not demonstrate differences between sustained and intermittent exercise on intramuscular glycogen content and lactate accumulation, but subsequent studies by the same investigators55 suggested lower rates of glycolysis with intermittent compared with sustained exercise in healthy adults. In an intriguing study,56,57 fat oxidation was threefold lower and carbohydrate oxidation was 1.2-fold higher during intermittent versus sustained exercise of equivalent energy expenditures. A recent study58 in type 2 diabetes subjects revealed improved insulin sensitivity, as measured by the homeostasis model of insulin resistance, during intermittent exercise performed with mild hypoxia. Unfortunately, this study did not compare intermittent versus sustained exercise at normoxia.

Physical Activity in T1D and Hypoglycemia

It is well known that physical activity (PA) influences glucose concentrations in patients with T1D not only during exercise but up to 15 h after exercise, leading to late evening and nocturnal hypoglycemia.59,60 Reports have also demonstrated that different types of exercise (i.e., resistance vs. aerobic) have contrasting effects on the duration and severity of acute61 and delayed62 post-exercise hypoglycemia. Although it has been suggested63 that there is sufficient evidence to recommend PA in the management of T1D, the duration, intensity, and form of exercise that should be recommended and whether such interventions would translate to better outcomes are presently unclear. A recent meta-analysis64 suggests benefits of regular aerobic training, interspersed with brief bouts of sprinting, on glycated hemoglobin concentrations and incidence of delayed hypoglycemia in T1D. Although the American Diabetes Association does not provide specific suggestions regarding PA in T1D, it recommends that adults with diabetes perform at least 150 min/week of moderate-intensity aerobic PA and, in those with type 2 diabetes without contraindications, resistance training at least twice a week.65 The American Diabetes Association also recommends ingestion of added carbohydrates if the pre-exercise glucose level is <100 mg/dL in insulin-treated individuals.

Systematic examinations of carbohydrate physiology and substrate metabolism during exercise are sparse in T1D. Increased risk of hypoglycemia during exercise in T1D individuals could be due to factors that either lower rates of EGP or increase rates of glucose uptake or a combination of both. Lower rates of EGP could be due to lower hepatic glycogen reserves, as has been demonstrated in the postprandial state in T1D subjects,66 accompanying α-cell dysfunction leading to impaired glucagon secretion during exercise, decreased hepatic glucagon sensitivity, deficient catecholamine response to exercise, or a combination thereof. In contrast, in a study comparing individuals with and without T1D during moderate- and high-intensity exercise, Petersen et al.,67 applying magnetic resonance technology, determined that compared with healthy individuals, those with T1D had higher rates of EGP that was entirely accounted for by increased rates of gluconeogenesis. However, to further complicate matters, defective activation of skeletal muscle and adipose tissue lipolysis during hypoglycemia in T1D subjects, as a result of insufficient catecholamine response,68 could potentially further prolong the duration and severity of exercise-induced hypoglycemia in these individuals.

PA in T1D: Management Strategies

Apart from applying common-sense tactics to prevent hypoglycemia during and after exercise, there have been few reports that have systematically examined therapeutic approaches to mitigate hypoglycemia during and after PA in T1D. An observational field study60 demonstrated the efficacy of real-time continuous glucose monitoring and carbohydrate loading in preventing post-exercise hypoglycemia in adolescents with T1D. Recently, short-term studies incorporating PA have shown improvements in glucose control in T1D patients. These studies have compared multiple daily insulin injections with open-loop continuous subcutaneous insulin infusion,69 open-loop continuous subcutaneous insulin infusion with single-hormone (insulin) closed-loop control,70 and open-loop continuous subcutaneous insulin infusion with single-hormone modular closed-loop control.71 Recent observational studies testing a dual-hormone (insulin and glucagon) closed-loop control system72 have shown low rates of hypoglycemia with moderate exercise.

Usual Versus Guideline-Based PA and Glucose Physiology

The recommendation of daily PA by the American Diabetes Association refers to all patients with diabetes. Specific PA recommendations for T1D are not available simply because of lack of good evidence of the effect of PA on glucose control or outcomes. Although there is evidence of benefits of standardized training (50–70% of maximal oxygen uptake) for 16–20 weeks on insulin sensitivity measured by the glucose clamp or intravenous glucose tolerance test in healthy lean and overweight adults,73 some of the beneficial effects on insulin sensitivity could have been attributed to accompanying weight loss. In an intriguing study74 conducted in healthy sedentary men, reductions in daily step count from approximately 10,000 steps to approximately 2,000 steps led to reductions in fat free mass with lowering of insulin sensitivity measured before and after a 2-week period.

Closed-Loop Strategies for Exercise

A recent review75 has identified exercise as a major obstacle in current closed-loop control efforts. Even if exercise is announced during closed-loop therapy, maintenance of optimal glucose control during and after exercise may be challenging despite dual-hormone systems without a flexible and learning program of concomitant carbohydrate ingestion. As elucidated above, even healthy trained and untrained individuals without diabetes need to resort to periodic carbohydrate ingestion during and after exercise to prevent exhaustion, fatigue, and symptomatic hypoglycemia and to improve performance, especially during prolonged, unusual, or intense physical activity or sports. Hence, it would be impractical and unnatural to expect to rely solely on closed-loop control (with dual-hormone insulin and glucagon) systems to prevent hypoglycemia without exogenous simple carbohydrate ingestion. Furthermore, the considerable intra- and interindividual variability of insulin sensitivity in T1D individuals, even in the absence of exercise under carefully controlled experimental conditions, as we have recently demonstrated,76 is expected to be far greater during exercise, posing additional challenges to a generic closed-loop control algorithm.

Summary and Conclusions

Physiological effects of exercise on glucose metabolism have been extensively studied in healthy humans, over the last several decades. However, detailed and systematic investigations into exercise effects on glucose homeostasis and resultant hypoglycemia, pertaining especially to T1D, are scarce. Studies exploring the effects of exercise of varying intensities and duration on counter-regulatory hormonal responses, substrate (carbohydrate and fat predominantly) metabolism, muscle and hepatic glucose, glycogen, and lipid kinetics, lactate threshold, etc., are necessary to fashion rational therapeutic approaches to prevent hypoglycemia, delay fatigue, and improve exercise performance and thereby health and fitness of our patients with T1D. The roles of short- and long-term glycemic control, duration of diabetes, and accompanying comorbidities and complications of diabetes on exercise physiology in T1D are also unknown. Such large and critical knowledge gaps need to be filled so that patients and their physicians are better informed to provide logical, evidence-based therapeutic options so that this patient population becomes fitter through regular exercise and training while simultaneously minimizing the constant and sometimes disabling fear of hypoglycemia that prevents these individuals from leading healthier and fitter lives. Such an understanding of physiological effects of exercise could also inform next-generation closed-loop control algorithms currently being designed for the artificial endocrine pancreas to treat T1D.

Acknowledgments

R.B. and A.B. wrote the manuscript. M.L.J. and Y.C.K. edited the manuscript.

R.B. is supported by grants DK 029953 and DK 090541. M.L.J. is supported by grant T32 DK 007198. Y.C.K. and A.B. are supported by grants DK 085516 and DK DP3 094331. All authors are supported by grant UL1 TR000135 from the National Center for Advancing Translational Science to Mayo Clinic.

Author Disclosure Statement

There are no competing financial interests to declare for any of the authors.

References

  • 1.Wahren J, Felig P, Ahlborg G, Jorfeldt L: Glucose metabolism during leg exercise in man. J Clin Invest 1971;50:2715–2725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Felig P, Wahren J: Fuel homeostasis in exercise. N Engl J Med 1975;293:1078–1084 [DOI] [PubMed] [Google Scholar]
  • 3.Wasserman DH, Williams PE, Lacy DB, Green DR, Cherrington AD: Importance of intrahepatic mechanisms to gluconeogenesis from alanine during exercise and recovery. Am J Physiol 1988;254:E518–E525 [DOI] [PubMed] [Google Scholar]
  • 4.Coggan AR, Swanson SC, Mendenhall LA, Habash DL, Kien CL: Effect of endurance training on hepatic glycogenolysis and gluconeogenesis during prolonged exercise in men. Am J Physiol 1995;268:E375–E383 [DOI] [PubMed] [Google Scholar]
  • 5.Wolfe RR, Nadel ER, Shaw JH, Stephenson LA, Wolfe MH: Role of changes in insulin and glucagon in glucose homeostasis in exercise. J Clin Invest 1986;77:900–907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Andersen P, Saltin B: Maximal perfusion of skeletal muscle in man. J Physiol 1985;366:233–249 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Nesher R, Karl IE, Kipnis DM: Dissociation of effects of insulin and contraction on glucose transport in rat epitrochlearis muscle. Am J Physiol 1985;249:C226–C232 [DOI] [PubMed] [Google Scholar]
  • 8.Yeh JI, Gulve EA, Rameh L, Birnbaum MJ: The effects of wortmannin on rat skeletal muscle. Dissociation of signaling pathways for insulin- and contraction-activated hexose transport. J Biol Chem 1995;270:2107–2111 [DOI] [PubMed] [Google Scholar]
  • 9.Lee AD, Hansen PA, Holloszy JO: Wortmannin inhibits insulin-stimulated but not contraction-stimulated glucose transport activity in skeletal muscle. FEBS Lett 1995;361:51–54 [DOI] [PubMed] [Google Scholar]
  • 10.Lund S, Holman GD, Schmitz O, Pedersen O: Contraction stimulates translocation of glucose transporter GLUT4 in skeletal muscle through a mechanism distinct from that of insulin. Proc Natl Acad Sci U S A 1995;92:5817–5821 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Wasserman DH, Johnson JL, Bupp JL, Lacy DB, Bracy DP: Regulation of gluconeogenesis during rest and exercise in the depancreatized dog. Am J Physiol 1993;265:E51–E60 [DOI] [PubMed] [Google Scholar]
  • 12.Wasserman DH, Mohr T, Kelly P, Lacy DB, Bracy D: Impact of insulin deficiency on glucose fluxes and muscle glucose metabolism during exercise. Diabetes 1992;41:1229–1238 [DOI] [PubMed] [Google Scholar]
  • 13.Wasserman DH, Williams PE, Lacy DB, Goldstein RE, Cherrington AD: Exercise-induced fall in insulin and hepatic carbohydrate metabolism during muscular work. Am J Physiol 1989;256:E500–E509 [DOI] [PubMed] [Google Scholar]
  • 14.Schiavon M, Hinshaw L, Mallad A, Dalla Man C, Sparacino G, Johnson M, Carter R, Basu R, Kudva Y, Cobelli C, Basu A: Postprandial glucose fluxes and insulin sensitivity during exercise: a study in healthy individuals. Am J Physiol Endocrinol Metab 2013;305:E557–E566 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Friedlander AL, Casazza GA, Horning MA, Huie MJ, Brooks GA: Training-induced alterations of glucose flux in men. J Appl Physiol 1997;82:1360–1369 [DOI] [PubMed] [Google Scholar]
  • 16.Friedlander AL, Casazza GA, Horning MA, Huie MJ, Piacentini MF, Trimmer JK, Brooks GA: Training-induced alterations of carbohydrate metabolism in women: women respond differently from men. J Appl Physiol 1998;85:1175–1186 [DOI] [PubMed] [Google Scholar]
  • 17.Perseghin G, Price TB, Petersen KF, Roden M, Cline GW, Gerow K, Rothman DL, Shulman GI: Increased glucose transport-phosphorylation and muscle glycogen synthesis after exercise training in insulin-resistant subjects. N Engl J Med 1996;335:1357–1362 [DOI] [PubMed] [Google Scholar]
  • 18.Horton TJ, Grunwald GK, Lavely J, Donahoo WT: Glucose kinetics differ between women and men, during and after exercise. J Appl Physiol 2006;100:1883–1894 [DOI] [PubMed] [Google Scholar]
  • 19.Tarnopolsky LJ, MacDougall JD, Atkinson SA, Tarnopolsky MA, Sutton JR: Gender differences in substrate for endurance exercise. J Appl Physiol 1990;68:302–308 [DOI] [PubMed] [Google Scholar]
  • 20.Friedlander AL, Casazza GA, Horning MA, Buddinger TF, Brooks GA: Effects of exercise intensity and training on lipid metabolism in young women. Am J Physiol 1998;275:E853–E863 [DOI] [PubMed] [Google Scholar]
  • 21.Suh SH, Casazza GA, Horning MA, Miller BF, Brooks GA: Luteal and follicular glucose fluxes during rest and exercise in 3-h postabsorptive women. J Appl Physiol 2002;93:42–50 [DOI] [PubMed] [Google Scholar]
  • 22.Suh SH, Casazza GA, Horning MA, Miller BF, Brooks GA: Effects of oral contraceptives on glucose flux and substrate oxidation rates during rest and exercise. J Appl Physiol 2003;94:285–294 [DOI] [PubMed] [Google Scholar]
  • 23.Casazza GA, Jacobs KA, Suh SH, Miller BF, Horning MA, Brooks GA: Menstrual cycle phase and oral contraceptive effects on triglyceride mobilization during exercise. J Appl Physiol 2004;97:302–309 [DOI] [PubMed] [Google Scholar]
  • 24.Devries MC, Hamadeh MJ, Graham TE, Tarnopolsky MA: 17β-Estradiol supplementation decreases glucose rate of appearance and disappearance with no effect on glycogen utilization during moderate intensity exercise in men. J Clin Endocrinol Metab 2005;90:6218–6225. Erratum in J Clin Endocrinol Metab 2006;91:1217. [DOI] [PubMed] [Google Scholar]
  • 25.Richter EA, Garetto LP, Goodman MN, Ruderman NB: Muscle glucose metabolism following exercise in the rat: increased sensitivity to insulin. J Clin Invest 1982;69:785–793 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Richter EA, Ruderman NB, Gavras H, Belur ER, Galbo H: Muscle glycogenolysis during exercise: dual control by epinephrine and contractions. Am J Physiol 1982;242:E25–E32 [DOI] [PubMed] [Google Scholar]
  • 27.Holloszy JO, Narahara HT: Studies of tissue permeability. X. Changes in permeability to 3-methylglucose associated with contraction of isolated frog muscle. J Biol Chem 1965;240:3493–3500 [PubMed] [Google Scholar]
  • 28.Ivy JL, Holloszy JO: Persistent increase in glucose uptake by rat skeletal muscle following exercise. Am J Physiol 1981;241:C200–C203 [DOI] [PubMed] [Google Scholar]
  • 29.Garetto LP, Richter EA, Goodman MN, Ruderman NB: Enhanced muscle glucose metabolism after exercise in the rat: the two phases. Am J Physiol 1984;246:E471–E475 [DOI] [PubMed] [Google Scholar]
  • 30.Richter EA, Garetto LP, Goodman MN, Ruderman NB: Enhanced muscle glucose metabolism after exercise: modulation by local factors. Am J Physiol 1984;246:E476–E482 [DOI] [PubMed] [Google Scholar]
  • 31.Holloszy JO, Coyle EF: Adaptations of skeletal muscle to endurance exercise and their metabolic consequences. J Appl Physiol 1984;56:831–838 [DOI] [PubMed] [Google Scholar]
  • 32.Wallberg-Henriksson H, Holloszy JO: Activation of glucose transport in diabetic muscle: responses to contraction and insulin. Am J Physiol 1985;24:C233–C237 [DOI] [PubMed] [Google Scholar]
  • 33.Constable SH, Favier RJ, Cartee GD, Young DA, Holloszy JO: Muscle glucose transport: interactions of in vitro contractions, insulin, and exercise. J Appl Physiol 1988;64:2329–2332 [DOI] [PubMed] [Google Scholar]
  • 34.Wallberg-Henriksson H, Constable SH, Young DA, Holloszy JO: Glucose transport into rat skeletal muscle: interaction between exercise and insulin. J Appl Physiol 1988;65:909–913 [DOI] [PubMed] [Google Scholar]
  • 35.Zorzano A, Balon TW, Goodman MN, Ruderman NB: Glycogen depletion and increased insulin sensitivity and responsiveness in muscle after exercise. Am J Physiol 1986;251:E664–E669 [DOI] [PubMed] [Google Scholar]
  • 36.Gulve EA, Cartee GD, Zierath JR, Corpus VM, Holloszy JO: Reversal of enhanced muscle glucose transport after exercise: roles of insulin and glucose. Am J Physiol 1990;259:E685–E691 [DOI] [PubMed] [Google Scholar]
  • 37.Greiwe JS, Hickner RC, Hansen PA, Racette SB, Chen MM, Holloszy JO: Effects of endurance exercise training on muscle glycogen accumulation in humans. J Appl Physiol 1999;87:222–226 [DOI] [PubMed] [Google Scholar]
  • 38.Friedman JE, Sherman WM, Reed MJ, Elton CW, Dohm GL: Exercise training increases glucose transporter protein GLUT-4 in skeletal muscle of obese Zucker (fa/fa) rats. FEBS Lett 1990;268:13–16 [DOI] [PubMed] [Google Scholar]
  • 39.Ploug T, Stallknecht BM, Pedersen O, Kahn BB, Ohkuwa T, Vinten J, Galbo H: Effect of endurance training on glucose transport capacity and glucose transporter expression in rat skeletal muscle. Am J Physiol 1990;259:E778–E786 [DOI] [PubMed] [Google Scholar]
  • 40.Rodnick KJ, Holloszy JO, Mondon CE, James DE: Effects of exercise training on insulin-regulatable glucose-transporter protein levels in rat skeletal muscle. Diabetes 1990;39:1425–1429 [DOI] [PubMed] [Google Scholar]
  • 41.Host HH, Hansen PA, Nolte LA, Chen MM, Holloszy JO: Glycogen supercompensation masks the effect of a traininginduced increase in GLUT-4 on muscle glucose transport. J Appl Physiol 1998;85:133–138 [DOI] [PubMed] [Google Scholar]
  • 42.Ren JM, Semenkovich CF, Gulve EA, Gao J, Holloszy JO: Exercise induces rapid increases in GLUT4 expression, glucose transport capacity, and insulin-stimulated glycogen storage in muscle. J Biol Chem 1994;269:14396–14401 [PubMed] [Google Scholar]
  • 43.Short KR, Vittone JL, Bigelow ML, Proctor DN, Rizza RA, Coenen-Schimke JM, Nair KS: Impact of aerobic exercise training on age-related changes in insulin sensitivity and muscle oxidative capacity. Diabetes 2003;52:1888–1896 [DOI] [PubMed] [Google Scholar]
  • 44.Treebak JT, Taylor EB, Witczak CA, An D, Toyoda T, Koh HJ, Xie J, Feener EP, Wojtaszewski JF, Hirshman MF, Goodyear LJ: Identification of a novel phosphorylation site on TBC1D4 regulated by AMP-activated protein kinase in skeletal muscle. Am J Physiol Cell Physiol 2010;298:C377–C385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Jessen N, An D, Lihn AS, Nygren J, Hirshman MF, Thorell A, Goodyear LJ: Exercise increases TBC1D1 phosphorylation in human skeletal muscle. Am J Physiol Endocrinol Metab 2011;301:E164–E171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Funai K, Schweitzer GG, Sharma N, Kanzaki M, Cartee GD: Increased AS160 phosphorylation, but not TBC1D1 phosphorylation, with increased postexercise insulin sensitivity in rat skeletal muscle. Am J Physiol Endocrinol Metab 2009;297:E242–E251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Proctor DN, Shen PH, Dietz NM, Eickhoff TJ, Lawler LA, Ebersold EJ, Loeffler DL, Joyner MJ: Reduced leg blood flow during dynamic exercise in older endurance-trained men. J Appl Physiol 1998;85:68–75 [DOI] [PubMed] [Google Scholar]
  • 48.Zinker BA, Lacy DB, Bracy DP, Wasserman DH: Role of glucose and insulin loads to the exercising limb in increasing glucose uptake and metabolism. J Appl Physiol 1993;74:2915–2921 [DOI] [PubMed] [Google Scholar]
  • 49.Coyle EF, Hagberg JM, Hurley BF, Martin WH, Ehsani AA, Holloszy JO: Carbohydrate feeding during prolonged strenuous exercise can delay fatigue. J Appl Physiol Respir Environ Exerc Physiol 1983;55:230–235 [DOI] [PubMed] [Google Scholar]
  • 50.Fritzsche RG, Switzer TW, Hodgkinson BJ, Lee SH, Martin JC, Coyle EF: Water and carbohydrate ingestion during prolonged exercise increase maximal neuromuscular power. J Appl Physiol 2000;88:730–737 [DOI] [PubMed] [Google Scholar]
  • 51.Febbraio MA, Chiu A, Angus DJ, Arkinstall MJ, Hawley JA: Effects of carbohydrate ingestion before and during exercise on glucose kinetics and performance. J Appl Physiol 2000;89:2220–2226 [DOI] [PubMed] [Google Scholar]
  • 52.Coyle EF, Hamilton MT, Alonso JG, Montain SJ, Ivy JL: Carbohydrate metabolism during intense exercise when hyperglycemic. J Appl Physiol 1991;70:834–840 [DOI] [PubMed] [Google Scholar]
  • 53.Fahey AJ, Paramalingam N, Davey RJ, Davis EA, Jones TW, Fournier PA: The effect of a short sprint on postexercise whole-body glucose production and utilization rates in individuals with type 1 diabetes mellitus. J Clin Endocrinol Metab 2012;97:4193–4200 [DOI] [PubMed] [Google Scholar]
  • 54.Essen B, Hagenfeldt L, Kaijser L: Utilization of blood-borne and intramuscular substrates during continuous and intermittent exercise in man. J Physiol 1977;265:489–506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Essen B, Kaijser L: Regulation of glycolysis in intermittent exercise in man. J Physiol 1978;281:499–511 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Christmass MA, Dawson B, Arthur PG: Effect of work and recovery duration on skeletal muscle oxygenation and fuel use during sustained intermittent exercise. Eur J Appl Physiol Occup Physiol 1999;80:436–447 [DOI] [PubMed] [Google Scholar]
  • 57.Christmass MA, Dawson B, Passeretto P, Arthur PG: A comparison of skeletal muscle oxygenation and fuel use in sustained continuous and intermittent exercise. Eur J Appl Physiol Occup Physiol 1999;80:423–435 [DOI] [PubMed] [Google Scholar]
  • 58.Mackenzie R, Maxwell N, Castle P, Elliott B, Brickley G, Watt P: Intermittent exercise with and without hypoxia improves insulin sensitivity in individuals with type 2 diabetes. J Clin Endocrinol Metab 2012;97:E546–E555 [DOI] [PubMed] [Google Scholar]
  • 59.Maran A, Pavan P, Bonsembiante B, Brugin E, Ermolao A, Avogaro A, Zaccaria M: Continuous glucose monitoring reveals delayed nocturnal hypoglycemia after intermittent high-intensity exercise in nontrained patients with type 1 diabetes. Diabetes Technol Ther 2010;12:763–768 [DOI] [PubMed] [Google Scholar]
  • 60.Riddell MC, Milliken J: Preventing exercise-induced hypoglycemia in type 1 diabetes using real-time continuous glucose monitoring and a new carbohydrate intake algorithm: an observational field study. Diabetes Technol Ther 2011;13:819–825 [DOI] [PubMed] [Google Scholar]
  • 61.Yardley JE, Kenny GP, Perkins BA, Riddell MC, Malcolm J, Boulay P, Khandwala F, Sigal RJ: Effects of performing resistance exercise before versus after aerobic exercise on glycemia in type 1 diabetes. Diabetes Care 2012;35:669–675 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yardley JE, Kenny GP, Perkins BA, Riddell MC, Balaa N, Malcolm J, Boulay P, Khandwala F, Sigal RJ: Resistance versus aerobic exercise: acute effects on glycemia in type 1 diabetes. Diabetes Care 2013;36:537–542 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Chimen M, Kennedy A, Nirantharakumar K, Pang TT, Andrews R, Narendran P: What are the health benefits of physical activity in type 1 diabetes mellitus? A literature review. Diabetologia 2012;55:542–551 [DOI] [PubMed] [Google Scholar]
  • 64.Tonoli C, Heyman E, Roelands B, Buyse L, Cheung SS, Berthoin S, Meeusen R: Effects of different types of acute and chronic (training) exercise on glycaemic control in type 1 diabetes mellitus: a meta-analysis. Sports Med 2012;42:1059–1080 [DOI] [PubMed] [Google Scholar]
  • 65.Standards of medical care in diabetes—2013. Diabetes Care 2013;36(Suppl 1):S11–S66 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hwang JH, Perseghin G, Rothman DL, Cline GW, Magnusson I, Petersen KF, Shulman GI: Impaired net hepatic glycogen synthesis in insulin-dependent diabetic subjects during mixed meal ingestion. A 13C nuclear magnetic resonance spectroscopy study. J Clin Invest 1995;95:783–787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Petersen KF, Price TB, Bergeron R: Regulation of net hepatic glycogenolysis and gluconeogenesis during exercise: impact of type 1 diabetes. J Clin Endocrinol Metab 2004;89:4656–4664 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Enoksson S, Caprio SK, Rife F, Shulman GI, Tamborlane WV, Sherwin RS: Defective activation of skeletal muscle and adipose tissue lipolysis in type 1 diabetes mellitus during hypoglycemia. J Clin Endocrinol Metab 2003;88:1503–1511 [DOI] [PubMed] [Google Scholar]
  • 69.Yardley JE, Iscoe KE, Sigal RJ, Kenny GP, Perkins BA, Riddell MC: Insulin pump therapy is associated with less post-exercise hyperglycemia than multiple daily injections: an observational study of physically active type 1 diabetes patients. Diabetes Technol Ther 2013;15:84–88 [DOI] [PubMed] [Google Scholar]
  • 70.Elleri D, Allen JM, Kumareswaran K, Leelarathna L, Nodale M, Caldwell K, Cheng P, Kollman C, Haidar A, Murphy HR, Wilinska ME, Acerini CL, Dunger DB, Hovorka R: Closed-loop basal insulin delivery over 36 hours in adolescents with type 1 diabetes: randomized clinical trial. Diabetes Care 2013;36:838–844 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Breton M, Farret A, Bruttomesso D, Anderson S, Magni L, Patek S, Dalla Man C, Place J, Demartini S, Del Favero S, Toffanin C, Hughes-Karvetski C, Dassau E, Zisser H, Doyle FJ, 3rd, De Nicolao G, Avogaro A, Cobelli C, Renard E, Kovatchev B; International Artificial Pancreas Study Group: Fully integrated artificial pancreas in type 1 diabetes: modular closed-loop glucose control maintains near normoglycemia. Diabetes 2012;61:2230–2237 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Russell SJ, El-Khatib FH, Nathan DM, Magyar KL, Jiang J, Damiano ER: Blood glucose control in type 1 diabetes with a bihormonal bionic endocrine pancreas. Diabetes Care 2012;35:2148–2155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Boulé NG, Weisnagel SJ, Lakka TA, Tremblay A, Bergman RN, Rankinen T, Leon AS, Skinner JS, Wilmore JH, Rao DC, Bouchard C; HERITAGE Family Study: Effects of exercise training on glucose homeostasis: the HERITAGE Family Study. Diabetes Care 2005;28:108–114 [DOI] [PubMed] [Google Scholar]
  • 74.Olsen RH, Krogh-Madsen R, Thomsen C, Booth FW, Pedersen BK: Metabolic responses to reduced daily steps in healthy nonexercising men. JAMA 2008;299:1261–1263 [DOI] [PubMed] [Google Scholar]
  • 75.van Bon AC, Verbitskiy E, von Basum G, Hoekstra JB, DeVries JH: Exercise in closed-loop control: a major hurdle. J Diabetes Sci Technol 2011;5:1337–1341 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hinshaw L, Man CD, Nandy DK, Saad A, Bharucha AE, Levine JA, Rizza RA, Basu R, Carter RE, Cobelli C, Kudva YC, Basu A: Diurnal pattern of insulin action in type 1 diabetes: implications for a closed loop system. Diabetes 2013;62:2223–2229 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Diabetes Technology & Therapeutics are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES