Effects of a low carbohydrate diet on energy expenditure during weight loss maintenance: randomized trial

Cara B Ebbeling, Henry A Feldman, Gloria L Klein, Julia M W Wong, Lisa Bielak, Sarah K Steltz, Patricia K Luoto, Robert R Wolfe, William W Wong, David S Ludwig, Cara B Ebbeling, Henry A Feldman, Gloria L Klein, Julia M W Wong, Lisa Bielak, Sarah K Steltz, Patricia K Luoto, Robert R Wolfe, William W Wong, David S Ludwig

Abstract

Objective: To determine the effects of diets varying in carbohydrate to fat ratio on total energy expenditure.

Design: Randomized trial.

Setting: Multicenter collaboration at US two sites, August 2014 to May 2017.

Participants: 164 adults aged 18-65 years with a body mass index of 25 or more.

Interventions: After 12% (within 2%) weight loss on a run-in diet, participants were randomly assigned to one of three test diets according to carbohydrate content (high, 60%, n=54; moderate, 40%, n=53; or low, 20%, n=57) for 20 weeks. Test diets were controlled for protein and were energy adjusted to maintain weight loss within 2 kg. To test for effect modification predicted by the carbohydrate-insulin model, the sample was divided into thirds of pre-weight loss insulin secretion (insulin concentration 30 minutes after oral glucose).

Main outcome measures: The primary outcome was total energy expenditure, measured with doubly labeled water, by intention-to-treat analysis. Per protocol analysis included participants who maintained target weight loss, potentially providing a more precise effect estimate. Secondary outcomes were resting energy expenditure, measures of physical activity, and levels of the metabolic hormones leptin and ghrelin.

Results: Total energy expenditure differed by diet in the intention-to-treat analysis (n=162, P=0.002), with a linear trend of 52 kcal/d (95% confidence interval 23 to 82) for every 10% decrease in the contribution of carbohydrate to total energy intake (1 kcal=4.18 kJ=0.00418 MJ). Change in total energy expenditure was 91 kcal/d (95% confidence interval -29 to 210) greater in participants assigned to the moderate carbohydrate diet and 209 kcal/d (91 to 326) greater in those assigned to the low carbohydrate diet compared with the high carbohydrate diet. In the per protocol analysis (n=120, P<0.001), the respective differences were 131 kcal/d (-6 to 267) and 278 kcal/d (144 to 411). Among participants in the highest third of pre-weight loss insulin secretion, the difference between the low and high carbohydrate diet was 308 kcal/d in the intention-to-treat analysis and 478 kcal/d in the per protocol analysis (P<0.004). Ghrelin was significantly lower in participants assigned to the low carbohydrate diet compared with those assigned to the high carbohydrate diet (both analyses). Leptin was also significantly lower in participants assigned to the low carbohydrate diet (per protocol).

Conclusions: Consistent with the carbohydrate-insulin model, lowering dietary carbohydrate increased energy expenditure during weight loss maintenance. This metabolic effect may improve the success of obesity treatment, especially among those with high insulin secretion.

Trial registration: ClinicalTrials.gov NCT02068885.

Conflict of interest statement

Competing interests: All authors have completed the ICMJE uniform disclosure form at www.icmje.org/coi_disclosure.pdf and declare: research support for the submitted work as described above; no financial relationships with any organisations that might have an interest in the submitted work in the previous three years; no other relationships or activities that could appear to have influenced the submitted work except as follows: CBE and DSL have conducted research studies examining the carbohydrate-insulin model funded by the National Institutes of Health and philanthropic organizations unaffiliated with the food industry; DSL received royalties for books on obesity and nutrition that recommend a low glycemic load diet.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://group.bmj.com/group/rights-licensing/permissions.

Figures

Fig 1
Fig 1
Study design
Fig 2
Fig 2
Participant flow (see supplementary figure for details of exclusions)
Fig 3
Fig 3
Change in total energy expenditure, the primary outcome, in intention-to-treat (top) and per protocol (bottom) analyses. Data are shown as mean change from start of test phase, with whiskers representing 1 standard error above and below the mean. P tests uniformity across diet groups for average of changes at midpoint and end of test phase
Fig 4
Fig 4
Effect modification by pre-weight loss insulin secretion (insulin concentration 30 minutes after oral glucose) in intention-to-treat and per protocol analyses. Pre-weight loss body weight differed by third (first third, 83.8 kg; second third, 92.8 kg; third third 98.4 kg, P

Fig 5

Biomeasures of compliance in intention-to-treat…

Fig 5

Biomeasures of compliance in intention-to-treat and per protocol analyses. Measures include 1,5-anhydroglucitol (upper),…

Fig 5
Biomeasures of compliance in intention-to-treat and per protocol analyses. Measures include 1,5-anhydroglucitol (upper), mean pre-weight loss value 17 μg/mL; triglycerides (middle), mean pre-weight loss value 78 mg/dL (retransformed); and high density lipoprotein cholesterol (lower), mean pre-weight loss value 48 mg/dL. Data are shown as mean change from start of test phase, with whiskers representing 1 standard error above and below the mean. P tests uniformity across diet groups for average of changes at midpoint and end of test phase. Left, intention-to-treat analysis; right, per-protocol analysis
Fig 5
Fig 5
Biomeasures of compliance in intention-to-treat and per protocol analyses. Measures include 1,5-anhydroglucitol (upper), mean pre-weight loss value 17 μg/mL; triglycerides (middle), mean pre-weight loss value 78 mg/dL (retransformed); and high density lipoprotein cholesterol (lower), mean pre-weight loss value 48 mg/dL. Data are shown as mean change from start of test phase, with whiskers representing 1 standard error above and below the mean. P tests uniformity across diet groups for average of changes at midpoint and end of test phase. Left, intention-to-treat analysis; right, per-protocol analysis

References

    1. Maclean PS, Bergouignan A, Cornier MA, Jackman MR. Biology’s response to dieting: the impetus for weight regain. Am J Physiol Regul Integr Comp Physiol 2011;301:R581-600. 10.1152/ajpregu.00755.2010
    1. Leibel RL, Rosenbaum M, Hirsch J. Changes in energy expenditure resulting from altered body weight. N Engl J Med 1995;332:621-8. 10.1056/NEJM199503093321001
    1. Ludwig DS. The glycemic index: physiological mechanisms relating to obesity, diabetes, and cardiovascular disease. JAMA 2002;287:2414-23. 10.1001/jama.287.18.2414
    1. Ludwig DS, Friedman MI. Increasing adiposity: consequence or cause of overeating? JAMA 2014;311:2167-8. 10.1001/jama.2014.4133
    1. Taubes G. The science of obesity: what do we really know about what makes us fat? An essay by Gary Taubes. BMJ 2013;346:f1050. 10.1136/bmj.f1050
    1. Ludwig DS, Ebbeling CB. The Carbohydrate-Insulin Model of Obesity: Beyond “Calories In, Calories Out”. JAMA Intern Med 2018;178:1098-103. 10.1001/jamainternmed.2018.2933
    1. Lennerz B, Lennerz JK. Food addiction, high-glycemic-index carbohydrates, and obesity. Clin Chem 2018;64:64-71. 10.1373/clinchem.2017.273532
    1. Ford ES, Dietz WH. Trends in energy intake among adults in the United States: findings from NHANES. Am J Clin Nutr 2013;97:848-53. 10.3945/ajcn.112.052662
    1. Ludwig DS. Lowering the bar on the low-fat diet. JAMA 2016;316:2087-8. 10.1001/jama.2016.15473
    1. Hall KD. A review of the carbohydrate-insulin model of obesity. Eur J Clin Nutr 2017;71:323-6. 10.1038/ejcn.2016.260
    1. Hall KD, Guo J. Obesity energetics: body weight regulation and the effects of diet composition. Gastroenterology 2017;152:1718-27.e3. 10.1053/j.gastro.2017.01.052
    1. Howell S, Kones R. “Calories in, calories out” and macronutrient intake: the hope, hype, and science of calories. Am J Physiol Endocrinol Metab 2017;313:E608-12. 10.1152/ajpendo.00156.2017
    1. Schwartz MW, Seeley RJ, Zeltser LM, et al. Obesity pathogenesis: an Endocrine Society scientific statement. Endocr Rev 2017;38:267-96. 10.1210/er.2017-00111
    1. Bosy-Westphal A, Hägele F, Nas A. Impact of dietary glycemic challenge on fuel partitioning. Eur J Clin Nutr 2017;71:327-30. 10.1038/ejcn.2016.230
    1. Owen OE, Caprio S, Reichard GA, Jr, Mozzoli MA, Boden G, Owen RS. Ketosis of starvation: a revisit and new perspectives. Clin Endocrinol Metab 1983;12:359-79. 10.1016/S0300-595X(83)80046-2
    1. Vazquez JA, Adibi SA. Protein sparing during treatment of obesity: ketogenic versus nonketogenic very low calorie diet. Metabolism 1992;41:406-14. 10.1016/0026-0495(92)90076-M
    1. Yang MU, Van Itallie TB. Composition of weight lost during short-term weight reduction. Metabolic responses of obese subjects to starvation and low-calorie ketogenic and nonketogenic diets. J Clin Invest 1976;58:722-30. 10.1172/JCI108519
    1. Phinney SD, Bistrian BR, Wolfe RR, Blackburn GL. The human metabolic response to chronic ketosis without caloric restriction: physical and biochemical adaptation. Metabolism 1983;32:757-68. 10.1016/0026-0495(83)90105-1
    1. Ebbeling CB, Klein GL, Luoto PK, et al. A randomized study of dietary composition during weight-loss maintenance: Rationale, study design, intervention, and assessment. Contemp Clin Trials 2018;65:76-86. 10.1016/j.cct.2017.12.004
    1. Wong JM, Bielak L, Eddy RG, et al. An academia-industry partnership for planning and executing a community-based feeding study. Curr Dev Nutr 2018;2:nzy060.
    1. Institute of Medicine Dietary Reference Intakes for Energy, Carbohydrate, Fiber, Fat, Fatty Acids, Cholesterol, Protein, and Amino Acids. The National Academies Press, 2002.
    1. Halton TL, Hu FB. The effects of high protein diets on thermogenesis, satiety and weight loss: a critical review. J Am Coll Nutr 2004;23:373-85. 10.1080/07315724.2004.10719381
    1. Frankenfield D, Roth-Yousey L, Compher C. Comparison of predictive equations for resting metabolic rate in healthy nonobese and obese adults: a systematic review. J Am Diet Assoc 2005;105:775-89. 10.1016/j.jada.2005.02.005
    1. Mifflin MD, St Jeor ST, Hill LA, Scott BJ, Daugherty SA, Koh YO. A new predictive equation for resting energy expenditure in healthy individuals. Am J Clin Nutr 1990;51:241-7. 10.1093/ajcn/51.2.241
    1. World Health Organization. Human Energy Requirements: Report of a Joint FAO/WHO/UNU Expert Consultation, Rome, Italy, 17-24 October 2001: United Nations, 2004:96.
    1. Wolever TM, Jenkins DJ. The use of the glycemic index in predicting the blood glucose response to mixed meals. Am J Clin Nutr 1986;43:167-72. 10.1093/ajcn/43.1.167
    1. Atkinson FS, Foster-Powell K, Brand-Miller JC. International tables of glycemic index and glycemic load values: 2008. Diabetes Care 2008;31:2281-3. 10.2337/dc08-1239
    1. Black AE, Prentice AM, Coward WA. Use of food quotients to predict respiratory quotients for the doubly-labelled water method of measuring energy expenditure. Hum Nutr Clin Nutr 1986;40:381-91.
    1. Eisenstein J, Roberts SB, Dallal G, Saltzman E. High-protein weight-loss diets: are they safe and do they work? A review of the experimental and epidemiologic data. Nutr Rev 2002;60:189-200. 10.1301/00296640260184264
    1. Ebbeling CB, Leidig MM, Feldman HA, Lovesky MM, Ludwig DS. Effects of a low-glycemic load vs low-fat diet in obese young adults: a randomized trial. JAMA 2007;297:2092-102. 10.1001/jama.297.19.2092
    1. Hron BM, Ebbeling CB, Feldman HA, Ludwig DS. Relationship of insulin dynamics to body composition and resting energy expenditure following weight loss. Obesity (Silver Spring) 2015;23:2216-22. 10.1002/oby.21213
    1. Lam YY, Ravussin E. Analysis of energy metabolism in humans: A review of methodologies. Mol Metab 2016;5:1057-71. 10.1016/j.molmet.2016.09.005
    1. Schoeller DA. Recent advances from application of doubly labeled water to measurement of human energy expenditure. J Nutr 1999;129:1765-8. 10.1093/jn/129.10.1765
    1. Wong WW. 90th Anniversary Commentary: Measurement of Energy Expenditure in Free-Living Humans by Using Doubly Labeled Water. J Nutr 2018;148:1660-2. 10.1093/jn/nxy107
    1. Wong WW, Roberts SB, Racette SB, et al. The doubly labeled water method produces highly reproducible longitudinal results in nutrition studies. J Nutr 2014;144:777-83. 10.3945/jn.113.187823
    1. Wong WW, Lee LS, Klein PD. Deuterium and oxygen-18 measurements on microliter samples of urine, plasma, saliva, and human milk. Am J Clin Nutr 1987;45:905-13. 10.1093/ajcn/45.5.905
    1. Ravussin E, Harper IT, Rising R, Bogardus C. Energy expenditure by doubly labeled water: validation in lean and obese subjects. Am J Physiol 1991;261:E402-9.
    1. Ravussin E, Bogardus C. Relationship of genetics, age, and physical fitness to daily energy expenditure and fuel utilization. Am J Clin Nutr 1989;49(Suppl):968-75. 10.1093/ajcn/49.5.968
    1. Ebbeling CB, Swain JF, Feldman HA, et al. Effects of dietary composition on energy expenditure during weight-loss maintenance. JAMA 2012;307:2627-34. 10.1001/jama.2012.6607
    1. Bender R, Lange S. Adjusting for multiple testing--when and how? J Clin Epidemiol 2001;54:343-9. 10.1016/S0895-4356(00)00314-0
    1. Sánchez-Peña MJ, Márquez-Sandoval F, Ramírez-Anguiano AC, Velasco-Ramírez SF, Macedo-Ojeda G, González-Ortiz LJ. Calculating the metabolizable energy of macronutrients: a critical review of Atwater’s results. Nutr Rev 2017;75:37-48. 10.1093/nutrit/nuw044
    1. Seaman SR, White IR. Review of inverse probability weighting for dealing with missing data. Stat Methods Med Res 2013;22:278-95. 10.1177/0962280210395740
    1. Chomistek AK, Yuan C, Matthews CE, et al. Physical activity assessment with the ActiGraph GT3X and doubly labeled water. Med Sci Sports Exerc 2017;49:1935-44. 10.1249/MSS.0000000000001299
    1. Troiano RP, Berrigan D, Dodd KW, Mâsse LC, Tilert T, McDowell M. Physical activity in the United States measured by accelerometer. Med Sci Sports Exerc 2008;40:181-8. 10.1249/mss.0b013e31815a51b3
    1. Baldwin KM, Joanisse DR, Haddad F, et al. Effects of weight loss and leptin on skeletal muscle in human subjects. Am J Physiol Regul Integr Comp Physiol 2011;301:R1259-66. 10.1152/ajpregu.00397.2011
    1. Goldsmith R, Joanisse DR, Gallagher D, et al. Effects of experimental weight perturbation on skeletal muscle work efficiency, fuel utilization, and biochemistry in human subjects. Am J Physiol Regul Integr Comp Physiol 2010;298:R79-88. 10.1152/ajpregu.00053.2009
    1. Park Y, Dodd KW, Kipnis V, et al. Comparison of self-reported dietary intakes from the Automated Self-Administered 24-h recall, 4-d food records, and food-frequency questionnaires against recovery biomarkers. Am J Clin Nutr 2018;107:80-93. 10.1093/ajcn/nqx002
    1. Holsen L, Cerit H, Lennerz B, et al. Hypothalamic and nucleus accumbens cerebral blood flow vary as a function of long-term carbohydrate-to-fat ratio diets [abstract]. Neuropsychopharmacology (American College of Neuropsychopharmacology 56th Annual Meeting, Palm Springs) 2017;42:S150-1.
    1. Shimy K, Feldman HA, Klein GL, et al. A mechanistic examination of dietary composition on metabolic fuel availability [abstract].. Hormone Research in Paediatrics (10th International Joint Meeting of Pediatric Endocrinology; Washington DC) 2017;88(Suppl 1):S337.
    1. Muller MJ, Geisler C, Heymsfield SB, et al. Recent advances in understanding body weight homeostasis in humans. F1000Res 2018;7:pii: F1000 Faculty Rev-1025.
    1. Hall KD, Chen KY, Guo J, et al. Energy expenditure and body composition changes after an isocaloric ketogenic diet in overweight and obese men. Am J Clin Nutr 2016;104:324-33. 10.3945/ajcn.116.133561
    1. Ludwig DS, Ebbeling CB. Raising the bar on the low-carbohydrate diet. Am J Clin Nutr 2016;104:1487-8. 10.3945/ajcn.116.142182
    1. Friedman MI, Appel S. Energy expenditure and body composition changes after an isocaloric ketogenic diet in overweight and obese men: a secondary analysis of energy expenditure and physical activity. bioRxiv. The Preprint Server for Biology, 2018, 10.1101/383752.
    1. Pawlak DB, Kushner JA, Ludwig DS. Effects of dietary glycaemic index on adiposity, glucose homoeostasis, and plasma lipids in animals. Lancet 2004;364:778-85. 10.1016/S0140-6736(04)16937-7
    1. Chaput JP, Tremblay A, Rimm EB, Bouchard C, Ludwig DS. A novel interaction between dietary composition and insulin secretion: effects on weight gain in the Quebec Family Study. Am J Clin Nutr 2008;87:303-9. 10.1093/ajcn/87.2.303
    1. Astley CM, Todd JN, Salem RM, et al. Genetic Evidence That Carbohydrate-Stimulated Insulin Secretion Leads to Obesity. Clin Chem 2018;64:192-200. 10.1373/clinchem.2017.280727
    1. Pittas AG, Das SK, Hajduk CL, et al. A low-glycemic load diet facilitates greater weight loss in overweight adults with high insulin secretion but not in overweight adults with low insulin secretion in the CALERIE Trial. Diabetes Care 2005;28:2939-41. 10.2337/diacare.28.12.2939
    1. Gardner CD, Trepanowski JF, Del Gobbo LC, et al. Effect of low-fat vs low-carbohydrate diet on 12-month weight loss in overweight adults and the association with genotype pattern or insulin secretion: The DIETFITS randomized clinical trial. JAMA 2018;319:667-79. 10.1001/jama.2018.0245
    1. Larsen TM, Dalskov SM, van Baak M, et al. Diet, Obesity, and Genes (Diogenes) Project Diets with high or low protein content and glycemic index for weight-loss maintenance. N Engl J Med 2010;363:2102-13. 10.1056/NEJMoa1007137
    1. Qi Q, Chu AY, Kang JH, et al. Sugar-sweetened beverages and genetic risk of obesity. N Engl J Med 2012;367:1387-96. 10.1056/NEJMoa1203039
    1. Levine JA, Eberhardt NL, Jensen MD. Role of nonexercise activity thermogenesis in resistance to fat gain in humans. Science 1999;283:212-4. 10.1126/science.283.5399.212
    1. Rosenbaum M, Goldsmith R, Bloomfield D, et al. Low-dose leptin reverses skeletal muscle, autonomic, and neuroendocrine adaptations to maintenance of reduced weight. J Clin Invest 2005;115:3579-86. 10.1172/JCI25977
    1. Rosenbaum M, Vandenborne K, Goldsmith R, et al. Effects of experimental weight perturbation on skeletal muscle work efficiency in human subjects. Am J Physiol Regul Integr Comp Physiol 2003;285:R183-92. 10.1152/ajpregu.00474.2002
    1. Cummings DE, Foster-Schubert KE, Overduin J. Ghrelin and energy balance: focus on current controversies. Curr Drug Targets 2005;6:153-69. 10.2174/1389450053174569
    1. Mihalache L, Gherasim A, Niţă O, et al. Effects of ghrelin in energy balance and body weight homeostasis. Hormones (Athens) 2016;15:186-96. 10.14310/horm.2002.1672
    1. Ye Z, Liu G, Guo J, Su Z. Hypothalamic endoplasmic reticulum stress as a key mediator of obesity-induced leptin resistance. Obes Rev 2018;19:770-85. 10.1111/obr.12673
    1. Crujeiras AB, Goyenechea E, Abete I, et al. Weight regain after a diet-induced loss is predicted by higher baseline leptin and lower ghrelin plasma levels. J Clin Endocrinol Metab 2010;95:5037-44. 10.1210/jc.2009-2566
    1. Erez G, Tirosh A, Rudich A, et al. Phenotypic and genetic variation in leptin as determinants of weight regain. Int J Obes (Lond) 2011;35:785-92. 10.1038/ijo.2010.217
    1. Mavri A, Stegnar M, Sabovic M. Do baseline serum leptin levels predict weight regain after dieting in obese women? Diabetes Obes Metab 2001;3:293-6. 10.1046/j.1463-1326.2001.00134.x
    1. Sacks FM, Bray GA, Carey VJ, et al. Comparison of weight-loss diets with different compositions of fat, protein, and carbohydrates. N Engl J Med 2009;360:859-73. 10.1056/NEJMoa0804748
    1. Hall KD, Guo J, Chen KY, et al. Potential bias of doubly labeled water for measuring energy expenditure differences between diets varying in carbohydrate. bioRxiv. The Preprint Server for Biology, 2018, 10.1101/403931.
    1. Haggarty P, McGaw BA, Fuller MF, Christie SL, Wong WW. Water hydrogen incorporation into body fat in pigs: effect on double/triple-labeled water method. Am J Physiol 1991;260:R627-34.
    1. Parks EJ, Krauss RM, Christiansen MP, Neese RA, Hellerstein MK. Effects of a low-fat, high-carbohydrate diet on VLDL-triglyceride assembly, production, and clearance. J Clin Invest 1999;104:1087-96. 10.1172/JCI6572
    1. Hudgins LC, Seidman CE, Diakun J, Hirsch J. Human fatty acid synthesis is reduced after the substitution of dietary starch for sugar. Am J Clin Nutr 1998;67:631-9. 10.1093/ajcn/67.4.631
    1. Schwarz JM, Neese RA, Turner S, Dare D, Hellerstein MK. Short-term alterations in carbohydrate energy intake in humans. Striking effects on hepatic glucose production, de novo lipogenesis, lipolysis, and whole-body fuel selection. J Clin Invest 1995;96:2735-43. 10.1172/JCI118342
    1. Strawford A, Antelo F, Christiansen M, Hellerstein MK. Adipose tissue triglyceride turnover, de novo lipogenesis, and cell proliferation in humans measured with 2H2O. Am J Physiol Endocrinol Metab 2004;286:E577-88. 10.1152/ajpendo.00093.2003
    1. Guo ZK, Cella LK, Baum C, Ravussin E, Schoeller DA. De novo lipogenesis in adipose tissue of lean and obese women: application of deuterated water and isotope ratio mass spectrometry. Int J Obes Relat Metab Disord 2000;24:932-7. 10.1038/sj.ijo.0801256
    1. Diraison F, Yankah V, Letexier D, Dusserre E, Jones P, Beylot M. Differences in the regulation of adipose tissue and liver lipogenesis by carbohydrates in humans. J Lipid Res 2003;44:846-53. 10.1194/jlr.M200461-JLR200
    1. Hudgins LC, Baday A, Hellerstein MK, et al. The effect of dietary carbohydrate on genes for fatty acid synthase and inflammatory cytokines in adipose tissues from lean and obese subjects. J Nutr Biochem 2008;19:237-45. 10.1016/j.jnutbio.2007.02.013
    1. Chong MF, Hodson L, Bickerton AS, et al. Parallel activation of de novo lipogenesis and stearoyl-CoA desaturase activity after 3 d of high-carbohydrate feeding. Am J Clin Nutr 2008;87:817-23. 10.1093/ajcn/87.4.817
    1. Letexier D, Pinteur C, Large V, Fréring V, Beylot M. Comparison of the expression and activity of the lipogenic pathway in human and rat adipose tissue. J Lipid Res 2003;44:2127-34. 10.1194/jlr.M300235-JLR200
    1. Viguerie N, Vidal H, Arner P, et al. Nutrient-Gene Interactions in Human Obesity--Implications for Dietary Guideline (NUGENOB) project Adipose tissue gene expression in obese subjects during low-fat and high-fat hypocaloric diets. Diabetologia 2005;48:123-31. 10.1007/s00125-004-1618-x
    1. Aarsland A, Chinkes D, Wolfe RR. Hepatic and whole-body fat synthesis in humans during carbohydrate overfeeding. Am J Clin Nutr 1997;65:1774-82. 10.1093/ajcn/65.6.1774
    1. Minehira K, Vega N, Vidal H, Acheson K, Tappy L. Effect of carbohydrate overfeeding on whole body macronutrient metabolism and expression of lipogenic enzymes in adipose tissue of lean and overweight humans. Int J Obes Relat Metab Disord 2004;28:1291-8. 10.1038/sj.ijo.0802760
    1. Rosenbaum M, Ravussin E, Matthews DE, et al. A comparative study of different means of assessing long-term energy expenditure in humans. Am J Physiol 1996;270:R496-504.
    1. Bueno NB, de Melo IS, de Oliveira SL, da Rocha Ataide T. Very-low-carbohydrate ketogenic diet v. low-fat diet for long-term weight loss: a meta-analysis of randomised controlled trials. Br J Nutr 2013;110:1178-87. 10.1017/S0007114513000548
    1. Johnston BC, Kanters S, Bandayrel K, et al. Comparison of weight loss among named diet programs in overweight and obese adults: a meta-analysis. JAMA 2014;312:923-33. 10.1001/jama.2014.10397
    1. Mancini JG, Filion KB, Atallah R, Eisenberg MJ. Systematic review of the mediterranean diet for long-term weight loss. Am J Med 2016;129:407-415.e4.
    1. Mansoor N, Vinknes KJ, Veierød MB, Retterstøl K. Effects of low-carbohydrate diets v. low-fat diets on body weight and cardiovascular risk factors: a meta-analysis of randomised controlled trials. Br J Nutr 2016;115:466-79. 10.1017/S0007114515004699
    1. Sackner-Bernstein J, Kanter D, Kaul S. Dietary intervention for overweight and obese adults: comparison of low-carbohydrate and low-fat diets. A meta-analysis. PLoS One 2015;10:e0139817. 10.1371/journal.pone.0139817
    1. Tobias DK, Chen M, Manson JE, Ludwig DS, Willett W, Hu FB. Effect of low-fat diet interventions versus other diet interventions on long-term weight change in adults: a systematic review and meta-analysis. Lancet Diabetes Endocrinol 2015;3:968-79. 10.1016/S2213-8587(15)00367-8

Source: PubMed

3
Prenumerera