Protocol for a single-centre, parallel-group, randomised, controlled, superiority trial on the effects of time-restricted eating on body weight, behaviour and metabolism in individuals at high risk of type 2 diabetes: the REStricted Eating Time (RESET) study

Jonas S Quist, Marie M Jensen, Kim K B Clemmensen, Hanne Pedersen, Natasja Bjerre, Joachim Størling, Martin B Blond, Nicolai J Wewer Albrechtsen, Jens J Holst, Signe S Torekov, Dorte Vistisen, Marit E Jørgensen, Satchidananda Panda, Christina Brock, Graham Finlayson, Kristine Færch, Jonas S Quist, Marie M Jensen, Kim K B Clemmensen, Hanne Pedersen, Natasja Bjerre, Joachim Størling, Martin B Blond, Nicolai J Wewer Albrechtsen, Jens J Holst, Signe S Torekov, Dorte Vistisen, Marit E Jørgensen, Satchidananda Panda, Christina Brock, Graham Finlayson, Kristine Færch

Abstract

Introduction: The aim of this study is to investigate the effects of time-restricted eating (TRE) on change in body weight and describe changes in behaviour and metabolism in individuals at high risk of type 2 diabetes.

Methods and analysis: The REStricted Eating Time (RESET) study is a randomised controlled parallel-group open-label trial. 100 women and men with (1) overweight (body mass index (BMI)≥25 kg/m2) and prediabetes (glycated haemoglobin 39-47 mmol/mol); or (2) obesity (BMI≥30 kg/m2) will be randomised to a control group (habitual living) or TRE (self-selected 10-hours eating window within the period from 06:00 to 20:00 in a 1:1 ratio. Testing is scheduled at baseline and after 6 weeks (mid-intervention), 3 months (post-intervention) and 6 months (follow-up). The primary outcome is change in body weight after 3 months of intervention. Secondary outcomes include changes in body composition; measures of glucose metabolism including glycaemic variability, hormones and metabolites; subjective and metabolic markers of appetite, food preferences and reward; dietary intake; physical activity, sleep, chronotype; gastric emptying, gastrointestinal transit time and motility; respiratory and glycolytic capacities; the plasma proteome and metabolome; blood pressure, resting heart rate and heart rate variability; and resting energy expenditure and substrate oxidation. Motivation and feasibility will be examined based on interviews at baseline and after 3 months. After the 3-month intervention, a 3-month follow-up period and subsequent testing are scheduled to assess maintenance and longer-term effects.

Ethics and dissemination: The study has been approved by the Ethics Committee of the Capital Region of Denmark (H-18059188) and the Danish Data Protection Agency. The study will be conducted in accordance with the Declaration of Helsinki. Results from the study will address whether TRE is effective and feasible in improving health outcomes in individuals at risk of lifestyle-related diseases and can potentially inform the design of feasible health recommendations.

Trial registration number: NCT03854656.

Keywords: diabetes & endocrinology; nutrition & dietetics.

Conflict of interest statement

Competing interests: Steno Diabetes Center Copenhagen is a hospital providing health services for the public healthcare system. Steno Diabetes Center Copenhagen is partly funded by the Novo Nordisk Foundation through unrestricted grants. The Novo Nordisk Foundation has no economic interests in the study. The Novo Nordisk Foundation will not have an influence on the study design, data collection, analysis, interpretation of data, the writing of the study report or any publication and the decision to submit the paper for publication. The investigators employed at Steno Diabetes Center Copenhagen will not benefit economically from conducting the study. HP is a coinvestigator on the project which is part of her Industrial PhD project in collaboration with iMotions A/S, where HP is employed. iMotions A/S is a collaborator on the project and gives advice for the use and analysis of biometric methods in the study design phase. SP has published a book, The Circadian Code, focusing on the concept of TRE.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Study design.

References

    1. Lee CMY, Colagiuri S, Woodward M, et al. . Comparing different definitions of prediabetes with subsequent risk of diabetes: an individual participant data meta-analysis involving 76 513 individuals and 8208 cases of incident diabetes. BMJ Open Diabetes Res Care 2019;7:1–10. 10.1136/bmjdrc-2019-000794
    1. Schmidt MI, Bracco PA, Yudkin JS, et al. . Intermediate hyperglycaemia to predict progression to type 2 diabetes (ELSA-Brasil): an occupational cohort study in Brazil. Lancet Diabetes Endocrinol 2019;7:267–77. 10.1016/S2213-8587(19)30058-0
    1. Vistisen D, Witte DR, Brunner EJ, et al. . Risk of cardiovascular disease and death in individuals with prediabetes defined by different criteria: the Whitehall II study. Diabetes Care 2018;41:899–906. 10.2337/dc17-2530
    1. Gummesson A, Nyman E, Knutsson M, et al. . Effect of weight reduction on glycated haemoglobin in weight loss trials in patients with type 2 diabetes. Diabetes Obes Metab 2017;19:1295–305. 10.1111/dom.12971
    1. le Roux CW, Astrup A, Fujioka K, et al. . 3 years of liraglutide versus placebo for type 2 diabetes risk reduction and weight management in individuals with prediabetes: a randomised, double-blind trial. Lancet 2017;389:1399–409. 10.1016/S0140-6736(17)30069-7
    1. American Diabetes Association 3. Prevention or delay of type 2 diabetes: Standards of Medical Care in diabetes-2019. Diabetes Care 2019;42:S29–33. 10.2337/dc19-S003
    1. Anderson JW, Konz EC, Frederich RC, et al. . Long-term weight-loss maintenance: a meta-analysis of US studies. Am J Clin Nutr 2001;74:579–84. 10.1093/ajcn/74.5.579
    1. Lemstra M, Bird Y, Nwankwo C, et al. . Weight loss intervention adherence and factors promoting adherence: a meta-analysis. Patient Prefer Adherence 2016;10:1547–59. 10.2147/PPA.S103649
    1. Melkani GC, Panda S. Time-restricted feeding for prevention and treatment of cardiometabolic disorders. J Physiol 2017;595:3691–700. 10.1113/JP273094
    1. Longo VD, Panda S. Fasting, circadian rhythms, and Time-Restricted feeding in healthy lifespan. Cell Metab 2016;23:1048–59. 10.1016/j.cmet.2016.06.001
    1. Gill S, Panda S. A smartphone APP reveals erratic diurnal eating patterns in humans that can be modulated for health benefits. Cell Metab 2015;22:789–98. 10.1016/j.cmet.2015.09.005
    1. Wittmann M, Dinich J, Merrow M, et al. . Social jetlag: misalignment of biological and social time. Chronobiol Int 2006;23:497–509. 10.1080/07420520500545979
    1. St-Onge M-P, Ard J, Baskin ML, et al. . Meal timing and frequency: implications for cardiovascular disease prevention: a scientific statement from the American heart association. Circulation 2017;135:e96–121. 10.1161/CIR.0000000000000476
    1. Arble DM, Bass J, Behn CD, et al. . Impact of sleep and circadian disruption on energy balance and diabetes: a summary of workshop discussions. Sleep 2015;38:1849–60. 10.5665/sleep.5226
    1. Garaulet M, Gómez-Abellán P. Timing of food intake and obesity: a novel association. Physiol Behav 2014;134:44–50. 10.1016/j.physbeh.2014.01.001
    1. Scheer FAJL, Hilton MF, Mantzoros CS, et al. . Adverse metabolic and cardiovascular consequences of circadian misalignment. Proc Natl Acad Sci U S A 2009;106:4453–8. 10.1073/pnas.0808180106
    1. Gonnissen HKJ, Hulshof T, Westerterp-Plantenga MS. Chronobiology, endocrinology, and energy- and food-reward homeostasis. Obes Rev 2013;14:405–16. 10.1111/obr.12019
    1. Chaix A, Manoogian ENC, Melkani GC, et al. . Time-Restricted eating to prevent and manage chronic metabolic diseases. Annu Rev Nutr 2019;39:12.1–12.5. 10.1146/annurev-nutr-082018-124320
    1. Sutton EF, Beyl R, Early KS, et al. . Early time-restricted feeding improves insulin sensitivity, blood pressure, and oxidative stress even without weight loss in men with prediabetes. Cell Metab 2018;27:1212–21. 10.1016/j.cmet.2018.04.010
    1. Jamshed H, Beyl RA, Della Manna DL, et al. . Early Time-Restricted feeding improves 24-hour glucose levels and affects markers of the circadian clock, aging, and autophagy in humans. Nutrients 2019;11:1234–16. 10.3390/nu11061234
    1. Hutchison AT, Regmi P, Manoogian ENC, et al. . Time-Restricted feeding improves glucose tolerance in men at risk for type 2 diabetes: a randomized crossover trial. Obesity 2019;27:724–32. 10.1002/oby.22449
    1. Ravussin E, Beyl RA, Poggiogalle E, et al. . Early time‐restricted feeding reduces appetite and increases fat oxidation but does not affect energy expenditure in humans. Obesity 2019;27:1244–54. 10.1002/oby.22518
    1. Gabel K, Hoddy KK, Haggerty N, et al. . Effects of 8-hour time restricted feeding on body weight and metabolic disease risk factors in obese adults: a pilot study. Nutr Healthy Aging 2018;4:345–53. 10.3233/NHA-170036
    1. Wilkinson MJ, Manoogian ENC, Zadourian A, et al. . Ten-Hour Time-Restricted eating reduces weight, blood pressure, and atherogenic lipids in patients with metabolic syndrome. Cell Metab 2020;31:92–104. 10.1016/j.cmet.2019.11.004
    1. Antoni R, Robertson TM, Robertson MD, et al. . A pilot feasibility study exploring the effects of a moderate time-restricted feeding intervention on energy intake, adiposity and metabolic physiology in free-living human subjects. J Nutr Sci 2018;7 10.1017/jns.2018.13
    1. Schulz KF, Altman DG, Moher D, et al. . CONSORT 2010 statement: updated guidelines for reporting parallel group randomised trials. BMJ 2010;340:c332 10.1136/bmj.c332
    1. Chan A-W, Tetzlaff JM, Gøtzsche PC, et al. . SPIRIT 2013 explanation and elaboration: guidance for protocols of clinical trials. BMJ 2013;346:e7586. 10.1136/bmj.e7586
    1. Ma C, Avenell A, Bolland M, et al. . Effects of weight loss interventions for adults who are obese on mortality, cardiovascular disease, and cancer: systematic review and meta-analysis. BMJ 2017;359:j4849. 10.1136/bmj.j4849
    1. Hulmán A, Færch K, Vistisen D, et al. . Effect of time of day and fasting duration on measures of glycaemia: analysis from the Whitehall II study. Diabetologia 2013;56:294–7. 10.1007/s00125-012-2770-3
    1. Weir JBDEB. New methods for calculating metabolic rate with special reference to protein metabolism. J Physiol 1949;109:1–9. 10.1113/jphysiol.1949.sp004363
    1. Frayn KN. Calculation of substrate oxidation rates in vivo from gaseous exchange. J Appl Physiol Respir Environ Exerc Physiol 1983;55:628–34. 10.1152/jappl.1983.55.2.628
    1. Geyer PE, Kulak NA, Pichler G, et al. . Plasma proteome profiling to assess human health and disease. Cell Syst 2016;2:185–95. 10.1016/j.cels.2016.02.015
    1. Galsgaard KD, Winther-Sørensen M, Ørskov C, et al. . Disruption of glucagon receptor signaling causes hyperaminoacidemia exposing a possible liver-alpha-cell axis. Am J Physiol Endocrinol Metab 2018;314:E93–103. 10.1152/ajpendo.00198.2017
    1. Jones N, Piasecka J, Bryant AH, et al. . Bioenergetic analysis of human peripheral blood mononuclear cells. Clin Exp Immunol 2015;182:69–80. 10.1111/cei.12662
    1. Flint A, Raben A, Blundell JE, et al. . Reproducibility, power and validity of visual analogue scales in assessment of appetite sensations in single test meal studies. Int J Obes 2000;24:38–48. 10.1038/sj.ijo.0801083
    1. Finlayson G, King N, Blundell JE. Is it possible to dissociate 'liking' and 'wanting' for foods in humans? A novel experimental procedure. Physiol Behav 2007;90:36–42. 10.1016/j.physbeh.2006.08.020
    1. Dalton M, Finlayson G. Psychobiological examination of liking and wanting for fat and sweet taste in trait binge eating females. Physiol Behav 2014;136:128–34. 10.1016/j.physbeh.2014.03.019
    1. Finlayson G, King N, Blundell J. The role of implicit wanting in relation to explicit liking and wanting for food: implications for appetite control. Appetite 2008;50:120–7. 10.1016/j.appet.2007.06.007
    1. Warde A. Consumption and theories of practice. J Consum Cult 2005;5:131–53. 10.1177/1469540505053090
    1. Malterud K. Systematic text condensation: a strategy for qualitative analysis. Scand J Public Health 2012;40:795–805. 10.1177/1403494812465030
    1. Maqbool S, Parkman HP, Friedenberg FK. Wireless capsule motility: comparison of the SmartPill Gi monitoring system with scintigraphy for measuring whole gut transit. Dig Dis Sci 2009;54:2167–74. 10.1007/s10620-009-0899-9
    1. Farmer AD, Wegeberg A-ML, Brock B, et al. . Regional gastrointestinal contractility parameters using the wireless motility capsule: inter-observer reproducibility and influence of age, gender and study country. Aliment Pharmacol Ther 2018;47:391–400. 10.1111/apt.14438
    1. Zarate N, Mohammed SD, O'Shaughnessy E, et al. . Accurate localization of a fall in pH within the ileocecal region: validation using a dual-scintigraphic technique. Am J Physiol Gastrointest Liver Physiol 2010;299:G1276–86. 10.1152/ajpgi.00127.2010
    1. The Danish Veterinary and Food Administration, Ministry of Environment and Food, Danish Dietary Recommendations [Internet]. Available:
    1. Jensen MD, Ryan DH, Apovian CM, et al. . 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults. Circulation 2014;129(25 Suppl:S102–38.
    1. Gabel K, Hoddy KK, Varady KA. Safety of 8-h time restricted feeding in adults with obesity. Appl Physiol Nutr Metab 2019;44:107–9. 10.1139/apnm-2018-0389
    1. Wing RR, Phelan S. Long-term weight loss maintenance. Am J Clin Nutr 2005;82:222S–5. 10.1093/ajcn/82.1.222S
    1. Wadden TA, Neiberg RH, Wing RR, et al. . Four-year weight losses in the look AHEAD study: factors associated with long-term success. Obesity 2011;19:1987–98. 10.1038/oby.2011.230
    1. Garcia Ulen C, Huizinga MM, Beech B, et al. . Weight regain prevention. Clin Diabetes 2008;26:100–13. 10.2337/diaclin.26.3.100

Source: PubMed

3
Subskrybuj