The influence of bright and dim light on substrate metabolism, energy expenditure and thermoregulation in insulin-resistant individuals depends on time of day

Jan-Frieder Harmsen, Jakob Wefers, Daniel Doligkeit, Luc Schlangen, Bas Dautzenberg, Pascal Rense, Dirk van Moorsel, Joris Hoeks, Esther Moonen-Kornips, Marijke C M Gordijn, Wouter D van Marken Lichtenbelt, Patrick Schrauwen, Jan-Frieder Harmsen, Jakob Wefers, Daniel Doligkeit, Luc Schlangen, Bas Dautzenberg, Pascal Rense, Dirk van Moorsel, Joris Hoeks, Esther Moonen-Kornips, Marijke C M Gordijn, Wouter D van Marken Lichtenbelt, Patrick Schrauwen

Abstract

Aims/hypothesis: In our modern society, artificial light is available around the clock and most people expose themselves to electrical light and light-emissive screens during the dark period of the natural light/dark cycle. Such suboptimal lighting conditions have been associated with adverse metabolic effects, and redesigning indoor lighting conditions to mimic the natural light/dark cycle more closely holds promise to improve metabolic health. Our objective was to compare metabolic responses to lighting conditions that resemble the natural light/dark cycle in contrast to suboptimal lighting in individuals at risk of developing metabolic diseases.

Methods: Therefore, we here performed a non-blinded, randomised, controlled, crossover trial in which overweight insulin-resistant volunteers (n = 14) were exposed to two 40 h laboratory sessions with different 24 h lighting protocols while staying in a metabolic chamber under real-life conditions. In the Bright day-Dim evening condition, volunteers were exposed to electric bright light (~1250 lx) during the daytime (08:00-18:00 h) and to dim light (~5 lx) during the evening (18:00-23:00 h). Vice versa, in the Dim day-Bright evening condition, volunteers were exposed to dim light during the daytime and bright light during the evening. Randomisation and allocation to light conditions were carried out by sequential numbering. During both lighting protocols, we performed 24 h indirect calorimetry, and continuous core body and skin temperature measurements, and took frequent blood samples. The primary outcome was plasma glucose focusing on the pre- and postprandial periods of the intervention.

Results: Spending the day in bright light resulted in a greater increase in postprandial triacylglycerol levels following breakfast, but lower glucose levels preceding the dinner meal at 18:00 h, compared with dim light (5.0 ± 0.2 vs 5.2 ± 0.2 mmol/l, n = 13, p=0.02). Dim day-Bright evening reduced the increase in postprandial glucose after dinner compared with Bright day-Dim evening (incremental AUC: 307 ± 55 vs 394 ± 66 mmol/l × min, n = 13, p=0.009). After the Bright day-Dim evening condition the sleeping metabolic rate was identical compared with the baseline night, whereas it dropped after Dim day-Bright evening. Melatonin secretion in the evening was strongly suppressed for Dim day-Bright evening but not for Bright day-Dim evening. Distal skin temperature for Bright day-Dim evening was lower at 18:00 h (28.8 ± 0.3°C vs 29.9 ± 0.4°C, n = 13, p=0.039) and higher at 23:00 h compared with Dim day-Bright evening (30.1 ± 0.3°C vs 28.8 ± 0.3°C, n = 13, p=0.006). Fasting and postprandial plasma insulin levels and the respiratory exchange ratio were not different between the two lighting protocols at any time.

Conclusions/interpretation: Together, these findings suggest that the indoor light environment modulates postprandial substrate handling, energy expenditure and thermoregulation of insulin-resistant volunteers in a time-of-day-dependent manner.

Trial registration: ClinicalTrials.gov NCT03829982.

Funding: We acknowledge the financial support from the Netherlands Cardiovascular Research Initiative: an initiative with support from the Dutch Heart Foundation (CVON2014-02 ENERGISE).

Keywords: Biological clock; Circadian rhythm; Glucose intolerance; Insulin resistance; Light at night; Light exposure; Melatonin; Postprandial metabolism; Sleeping metabolic rate.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Study scheme. Fasted blood samples were taken at 07:45 h on days 2 and 3 and at 17:45 h on day 2. Postprandial blood samples were taken for 4 h at 30 min intervals after both breakfasts and dinner. Slow stepping exercise for 30 min was performed at 12:30 h preceding lunch and at 15:30 h
Fig. 2
Fig. 2
Overview of different skin temperature outcomes averaged over 30 min intervals (n = 13). Tproximal (a), Tdistal (b) and DPG (c). The dashed lines with the light bulb indicate the time points when the light settings were changed. Data are presented as mean ± SEM
Fig. 3
Fig. 3
Plasma melatonin (n = 14) in the evening of day 2 upon Bright day–Dim evening (a) and Dim day–Bright evening (b). Lines represent individual data. Data points below the detection threshold of 1.9 pg/ml (8.18 pmol/l) are illustrated as 0 values. The horizontal dashed line indicates the DLMO threshold of 10 pg/ml (43.05 pmol/l). To convert melatonin values from pg/ml to pmol/l, please multiply by 4.305
Fig. 4
Fig. 4
Postprandial plasma responses for the two meals on day 2 (Breakfast1 [n = 13] and Dinner [n = 14]) and breakfast on day 3 (Breakfast2 [n = 13]). Blood glucose (a) and TG (d) for the first breakfast; blood glucose (b), TG (e) and insulin (g) for the dinner; blood glucose (c), TG (f) and insulin (h) for the second breakfast. Postprandial data were analysed using a generalised linear mixed model with time and light conditions and their interaction as fixed effects. Data are presented as mean ± SEM; ***p<0.001, *p<0.05 (note that there is a single * symbol in Fig. 4b, partially obscured by the y-axis)
Fig. 5
Fig. 5
Energy expenditure over the entire time spent in the respiration chamber (a) and SMR of both nights per condition (n = 13; Night1 refers to the first baseline night spent in the respiration chamber without any differences in light intervention; Night2 refers to the second night after the respective light intervention) (b); p values are based on paired t tests; **p<0.01. Data are presented as mean ± SEM

References

    1. Reinke H, Asher G. Crosstalk between metabolism and circadian clocks. Nat Rev Mol Cell Biol. 2019;20(4):227–241. doi: 10.1038/s41580-018-0096-9.
    1. Gutierrez-Monreal MA, Harmsen J-F, Schrauwen P, Esser KA. Ticking for metabolic health: the skeletal-muscle clocks. Obesity. 2020;28(Suppl 1):S46–S54.
    1. Mason IC, Qian J, Adler GK, Scheer FAJL. Impact of circadian disruption on glucose metabolism: implications for type 2 diabetes. Diabetologia. 2020;63(3):462–472. doi: 10.1007/s00125-019-05059-6.
    1. Albreiki MS, Middleton B, Hampton SM. A single night light exposure acutely alters hormonal and metabolic responses in healthy participants. Endocr Connect. 2017;6(2):100–110. doi: 10.1530/EC-16-0097.
    1. Gil-Lozano M, Hunter PM, Behan L-A, Gladanac B, Casper RF, Brubaker PL. Short-term sleep deprivation with nocturnal light exposure alters time-dependent glucagon-like peptide-1 and insulin secretion in male volunteers. Am J Physiol Endocrinol Metab. 2016;310(1):E41–E50. doi: 10.1152/ajpendo.00298.2015.
    1. Obayashi K, Yamagami Y, Kurumatani N, Saeki K. Bedroom lighting environment and incident diabetes mellitus: a longitudinal study of the HEIJO-KYO cohort. Sleep Med. 2020;65:1–3. doi: 10.1016/j.sleep.2019.07.006.
    1. Münch M, Wirz-Justice A, Brown SA, et al. The role of daylight for humans: gaps in current knowledge. Clocks & Sleep. 2020;2(1):61–85. doi: 10.3390/clockssleep2010008.
    1. Daugaard S, Markvart J, Bonde JP, et al. Light exposure during days with night, outdoor, and indoor work. Ann Work Expo Health. 2019;63(6):651–665. doi: 10.1093/annweh/wxy110.
    1. Versteeg RI, Stenvers DJ, Visintainer D, et al. Acute effects of morning light on plasma glucose and triglycerides in healthy men and men with type 2 diabetes. J Biol Rhythm. 2017;32(2):130–142. doi: 10.1177/0748730417693480.
    1. Melanson EL, Ritchie HK, Dear TB et al (2018) Daytime bright light exposure, metabolism, and individual differences in wake and sleep energy expenditure during circadian entrainment and misalignment. Neurobiol Sleep Circadian Rhythms 4:49–56. 10.1016/j.nbscr.2017.12.002
    1. Cheung IN, Zee PC, Shalman D, Malkani RG, Kang J, Reid KJ. Morning and evening blue-enriched light exposure alters metabolic function in Normal weight adults. PLoS One. 2016;11(5):e0155601. doi: 10.1371/journal.pone.0155601.
    1. Mari A, Pacini G, Murphy E, Ludvik B, Nolan JJ. A model-based method for assessing insulin sensitivity from the oral glucose tolerance test. Diabetes Care. 2001;24(3):539–548. doi: 10.2337/diacare.24.3.539.
    1. Schoffelen PF, Westerterp KR, Saris WH, Ten Hoor F. A dual-respiration chamber system with automated calibration. J Appl Physiol. 1997;83(6):2064–2072. doi: 10.1152/jappl.1997.83.6.2064.
    1. Weir JBDEB. New methods for calculating metabolic rate with special reference to protein metabolism. J Physiol. 1949;109(1–2):1–9. doi: 10.1113/jphysiol.1949.sp004363.
    1. ISO . ISO 9886: 2004. Ergonomics--evaluation of thermal strain by physiological measurements. Geneva: International Standards Organization; 2004.
    1. van Marken Lichtenbelt WD, Daanen HAM, Wouters L, et al. Evaluation of wireless determination of skin temperature using iButtons. Physiol Behav. 2006;88(4–5):489–497. doi: 10.1016/j.physbeh.2006.04.026.
    1. Plasqui G, Soenen S, Westerterp-Plantenga MS, Westerterp KR. Measurement of longitudinal changes in body composition during weight loss and maintenance in overweight and obese subjects using air-displacement plethysmography in comparison with the deuterium dilution technique. Int J Obes. 2011;35(8):1124–1130. doi: 10.1038/ijo.2010.250.
    1. van Faassen M, Bischoff R, Kema IP. Relationship between plasma and salivary melatonin and cortisol investigated by LC-MS/MS. Clin Chem Lab Med. 2017;55(9):1340–1348. doi: 10.1515/cclm-2016-0817.
    1. Kräuchi K, Cajochen C, Werth E, Wirz-Justice A. Functional link between distal vasodilation and sleep-onset latency? Am J Physiol Regul Integr Comp Physiol. 2000;278(3):R741–R748. doi: 10.1152/ajpregu.2000.278.3.R741.
    1. Berson DM, Dunn FA, Takao M. Phototransduction by retinal ganglion cells that set the circadian clock. Science. 2002;295(5557):1070–1073. doi: 10.1126/science.1067262.
    1. Barnard AR, Hattar S, Hankins MW, Lucas RJ. Melanopsin regulates visual processing in the mouse retina. Curr Biol. 2006;16(4):389–395. doi: 10.1016/j.cub.2005.12.045.
    1. Santhi N, Thorne HC, van der Veen DR, et al. The spectral composition of evening light and individual differences in the suppression of melatonin and delay of sleep in humans. J Pineal Res. 2012;53(1):47–59. doi: 10.1111/j.1600-079X.2011.00970.x.
    1. Wright KP, Jr, Gronfier C, Duffy JF, Czeisler CA. Intrinsic period and light intensity determine the phase relationship between melatonin and sleep in humans. J Biol Rhythm. 2005;20(2):168–177. doi: 10.1177/0748730404274265.
    1. Zerbini G, Winnebeck EC, Merrow M. Weekly, seasonal, and chronotype-dependent variation of dim-light melatonin onset. J Pineal Res. 2021;70(3):e12723. doi: 10.1111/jpi.12723.
    1. Wefers J, Connell NJ, Fealy CE, et al. Day-night rhythm of skeletal muscle metabolism is disturbed in older, metabolically compromised individuals. Mol Metab. 2020;41:101050. doi: 10.1016/j.molmet.2020.101050.
    1. Wefers J, van Moorsel D, Hansen J, et al. Circadian misalignment induces fatty acid metabolism gene profiles and compromises insulin sensitivity in human skeletal muscle. Proc Natl Acad Sci U S A. 2018;115(30):7789–779427. doi: 10.1073/pnas.1722295115.
    1. Refinetti R, Menaker M. The circadian rhythm of body temperature. Physiol Behav. 1992;51(3):613–637. doi: 10.1016/0031-9384(92)90188-8.
    1. Hood S, Amir S. The aging clock: circadian rhythms and later life. J Clin Invest. 2017;127(2):437–446. doi: 10.1172/JCI90328.
    1. Garaulet M, Qian J, Florez JC, Arendt J, Saxena R, Scheer FAJL. Melatonin effects on glucose metabolism: time to unlock the controversy. Trends Endocrinol Metab. 2020;31(3):192–204. doi: 10.1016/j.tem.2019.11.011.
    1. Morris CJ, Yang JN, Garcia JI, et al. Endogenous circadian system and circadian misalignment impact glucose tolerance via separate mechanisms in humans. Proc Natl Acad Sci U S A. 2015;112(17):E2225–E2234. doi: 10.1073/pnas.1418955112.
    1. Bonnefond A, Clément N, Fawcett K, et al. Rare MTNR1B variants impairing melatonin receptor 1B function contribute to type 2 diabetes. Nat Genet. 2012;44(3):297–301. doi: 10.1038/ng.1053.
    1. Tuomi T, Nagorny CLF, Singh P, et al. Increased melatonin signaling is a risk factor for type 2 diabetes. Cell Metab. 2016;23(6):1067–1077. doi: 10.1016/j.cmet.2016.04.009.
    1. Walford GA, Green T, Neale B, et al. Common genetic variants differentially influence the transition from clinically defined states of fasting glucose metabolism. Diabetologia. 2012;55(2):331–339. doi: 10.1007/s00125-011-2353-8.
    1. Cagnacci A, Arangino S, Renzi A, et al. Influence of melatonin administration on glucose tolerance and insulin sensitivity of postmenopausal women. Clin Endocrinol. 2001;54(3):339–346. doi: 10.1046/j.1365-2265.2001.01232.x.
    1. Sone Y, Hyun K-J, Nishimura S, Lee Y-A, Tokura H. Effects of dim or bright-light exposure during the daytime on human gastrointestinal activity. Chronobiol Int. 2003;20(1):123–133. doi: 10.1081/CBI-120017688.
    1. Nishimura S, Hyun K-J, Lee Y-A, Tokura H. Increase in parasympathetic nerve activity during the nighttime following bright light exposure during the daytime. Biol Rhythm Res. 2003;34(3):233–240. doi: 10.1076/brhm.34.3.233.18809.
    1. Tokura H, Gotoh K, Kondo M, Akimoto T, Kanikowska D. Salivary secretion under the influence of bright/dim light exposure in the morning and evening in humans. Biol Rhythm Res. 2002;33(2):129–140. doi: 10.1076/brhm.33.2.129.1316.
    1. Cajochen C, Münch M, Kobialka S, et al. High sensitivity of human melatonin, alertness, thermoregulation, and heart rate to short wavelength light. J Clin Endocrinol Metab. 2005;90(3):1311–1316. doi: 10.1210/jc.2004-0957.
    1. Lok R, van Koningsveld MJ, Gordijn MCM, Beersma DGM, Hut RA. Daytime melatonin and light independently affect human alertness and body temperature. J Pineal Res. 2019;67(1):e12583. doi: 10.1111/jpi.12583.
    1. Kräuchi K. How is the circadian rhythm of core body temperature regulated? Clin Auton Res. 2002;12(3):147–149. doi: 10.1007/s10286-002-0043-9.
    1. Kräuchi K, Gompper B, Hauenstein D, et al. Diurnal blood pressure variations are associated with changes in distal-proximal skin temperature gradient. Chronobiol Int. 2012;29(9):1273–1283. doi: 10.3109/07420528.2012.719961.
    1. Shea SA, Hilton MF, Hu K, Scheer FAJL. Existence of an endogenous circadian blood pressure rhythm in humans that peaks in the evening. Circ Res. 2011;108(8):980–984. doi: 10.1161/CIRCRESAHA.110.233668.
    1. Salles GF, Reboldi G, Fagard RH, et al. Prognostic effect of the nocturnal blood pressure fall in hypertensive patients. Hypertension. 2016;67(4):693–700. doi: 10.1161/HYPERTENSIONAHA.115.06981.
    1. Zitting K-M, Vujovic N, Yuan RK, et al. Human resting energy expenditure varies with circadian phase. Curr Biol. 2018;28(22):3685–3690. doi: 10.1016/j.cub.2018.10.005.
    1. Buxton OM, Cain SW, O’Connor SP, et al. Adverse metabolic consequences in humans of prolonged sleep restriction combined with circadian disruption. Sci Transl Med. 2012;4(129):129ra43. doi: 10.1126/scitranslmed.3003200.
    1. McHill AW, Melanson EL, Higgins J, et al. Impact of circadian misalignment on energy metabolism during simulated nightshift work. Proc Natl Acad Sci U S A. 2014;111(48):17302–17307. doi: 10.1073/pnas.1412021111.

Source: PubMed

3
订阅