Hostname: page-component-848d4c4894-4hhp2 Total loading time: 0 Render date: 2024-05-08T16:59:17.945Z Has data issue: false hasContentIssue false

Dietary palmitic acid to oleic acid ratio modulates energy metabolism and biological rhythms in young healthy Japanese males

Published online by Cambridge University Press:  14 August 2023

Katsuhiko Yajima*
Affiliation:
Laboratory of Nutritional Physiology, Faculty of Pharmaceutical Sciences, Josai University, Saitama, Japan
Shuto Chiba
Affiliation:
Laboratory of Nutritional Physiology, Faculty of Pharmaceutical Sciences, Josai University, Saitama, Japan
Insung Park
Affiliation:
International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
Hitomi Ogata
Affiliation:
Graduate School of Humanities and Social Sciences, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
Momoko Kayaba
Affiliation:
Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
Asuka Ishihara
Affiliation:
International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
Yoshiaki Tanaka
Affiliation:
International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
Zhang Simeng
Affiliation:
International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
Seol Jaehoon
Affiliation:
International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan Research Center for Overwork-Related Disorders, National Institute of Occupational Safety and Health, Japan (JNIOSH), Kanagawa, Japan R&D Center for Tailor-Made QOL, University of Tsukuba, Tsukuba, Japan
Masanori Katakura
Affiliation:
Laboratory of Nutritional Physiology, Faculty of Pharmaceutical Sciences, Josai University, Saitama, Japan
Kumpei Tokuyama
Affiliation:
International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
*
*Corresponding author: Katsuhiko Yajima, email k-yajima@josai.ac.jp

Abstract

The present study investigated the potential role of the composition of dietary fatty acids in the regulation of biological rhythms, such as the sleep architecture, core body temperature and leukocyte clock gene expression, in subjects fed meals rich in palmitic acid (PA) or oleic acid (OA). Eleven males participated in two sessions of indirect calorimetry in a whole-room metabolic chamber. In each session, subjects consumed three meals rich in PA (44·3 % of total fat as PA and 42·3 % as OA) or OA (11·7 % of total fat as PA and 59·3 % as OA) in the metabolic chamber. The ratio of PA to OA in plasma was significantly lower and fat oxidation was significantly higher during 24 h of indirect calorimetry in the session with meals rich in OA than in that with meals rich in PA. The duration of slow wave sleep (SWS) was shorter, the latency of SWS was longer and the nadir of core body temperature after bedtime was later in the session with meals rich in PA than in that with meals rich in OA. The peak in CRY1 gene expression was earlier and its amplitude was higher in the session with meals rich in PA than in that with meals rich in OA. In healthy young males, meals rich in PA decreased fat oxidation and disrupted biological rhythms, particularly the sleep architecture and core body temperature during sleep, more than meals rich in OA.

Type
Research Article
Copyright
© The Author(s), 2023. Published by Cambridge University Press on behalf of The Nutrition Society

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Jiang, P & Turek, FW (2018) Timing of meals: when is as critical as what and how much. Am J Physiol Endocrinol Metab 312, 369380.CrossRefGoogle Scholar
Canaple, L, Rambaud, J, Dkhissi-Benyahya, O, et al. (2006) Reciprocal regulation of brain and muscle Arnt-like protein 1 and peroxisome proliferator-activated receptor α defines a novel positive feedback loop in the rodent liver circadian clock. Mol Endocrinol 20, 17151727.CrossRefGoogle ScholarPubMed
Sonoda, J, Pei, L & Evans, RM (2008) Nuclear receptors: decoding metabolic disease. FEBS Lett 582, 29.CrossRefGoogle ScholarPubMed
Wang, N, Yang, G, Jia, Z, et al. (2008) Vascular PPARγ controls circadian variation in blood pressure and heart rate through Bmal1. Cell Metab 8, 482491.CrossRefGoogle ScholarPubMed
Scheer, FAJL, Hilton, MF, Mantzoros, CS, et al. (2009) Adverse metabolic and cardiovascular consequences of circadian misalignment. Proc Natl Acad Sci U S A 106, 44534458.CrossRefGoogle ScholarPubMed
Wefers, J, van Moorsel, D, Hansen, J, et al. (2018) Circadian misalignment induces fatty acid metabolism gene profiles and compromises insulin sensitivity in human skeletal muscle. Proc Natl Acad Sci U S A 115, 77897794.CrossRefGoogle ScholarPubMed
Morris, CJ, Purvis, TE, Hu, K, et al. (2016) Circadian misalignment increases cardiovascular disease risk factors in humans. Proc Natl Acad Sci U S A 113, E1402E1411.CrossRefGoogle ScholarPubMed
Astrup, A & Sci, M (2005) The role of dietary fat in obesity. Semin Vasc Med 5, 4047.CrossRefGoogle ScholarPubMed
Elmadfa, I & Kornsteiner, M (2009) Dietary fat intake - a global perspective. Ann Nutr Metab 54, 814.CrossRefGoogle ScholarPubMed
Astrup, A, Dyerberg, J, Elwood, P, et al. (2011) The role of reducing intakes of saturated fat in the prevention of cardiovascular disease: where does the evidence stand in 2010? Am J Clin Nutr 93, 684688.CrossRefGoogle ScholarPubMed
Kien, CL (2009) Dietary interventions for metabolic syndrome: role of modifying dietary fats. Curr Diab Rep 9, 4350.CrossRefGoogle ScholarPubMed
Kohsaka, A, Laposky, AD, Ramsey, KM, et al. (2007) High-fat diet disrupts behavioral and molecular circadian rhythms in mice. Cell Metab 6, 414421.CrossRefGoogle ScholarPubMed
Sartorius, T, Ketterer, C, Kullmann, S, et al. (2012) Monounsaturated fatty acids prevent the aversive effects of obesity on locomotion, brain activity, and sleep behavior. Diabetes 61, 16691679.CrossRefGoogle ScholarPubMed
Yuan, F, Wang, H, Tian, Y, et al. (2016) Fish oil alleviated high-fat diet-induced non-alcoholic fatty liver disease via regulating hepatic lipids metabolism and metaflammation: a transcriptomic study. Lipids Health Dis 15, 20.CrossRefGoogle ScholarPubMed
Barnea, M, Madar, Z & Froy, O (2009) High-fat diet delays and fasting advances the circadian expression of adiponectin signaling components in mouse liver. Endocrinology 150, 161168.CrossRefGoogle ScholarPubMed
Ding, L, Liu, J, Zhou, L, et al. (2022) A high-fat diet disrupts the hepatic and adipose circadian rhythms and modulates the diurnal rhythm of gut microbiota-derived short-chain fatty acids in gestational mice. Front Nutr 9, 925390.CrossRefGoogle ScholarPubMed
Popeijus, HE, van Otterdijk, SD, van der Krieken, SE, et al. (2014) Fatty acid chain length and saturation influences PPARα transcriptional activation and repression in HepG2 cells. Mol Nutr Food Res 58, 23422349.CrossRefGoogle ScholarPubMed
Gil-Lozano, M, Wu, WK, Martchenko, A, et al. (2016) High-fat diet and palmitate alter the rhythmic secretion of glucagon-like peptide-1 by the rodent L-cell. Endocrinology 157, 586599.CrossRefGoogle ScholarPubMed
Lago-Sampedro, A, Ho-Plagaro, A, Garcia-Serrano, S, et al. (2021) Oleic acid restores the rhythmicity of the disrupted circadian rhythm found in gastrointestinal explants from patients with morbid obesity. Clin Nutr 40, 43244333.CrossRefGoogle ScholarPubMed
Pivovarova, O, Jürchott, K, Rudovich, N, et al. (2015) Changes of dietary fat and carbohydrate content alter central and peripheral clock in humans. J Clin Endocrinol Metab 100, 22912302.CrossRefGoogle ScholarPubMed
Laura, X, Puig, S, Pillon, NJ, Näslund, E, et al. (2020) Influence of obesity, weight loss, and free fatty acids on skeletal muscle clock gene expression. Am J Physiol Endocrinol Metab 318, 110.Google Scholar
Yajima, K, Iwayama, K, Ogata, H, et al. (2018) Meal rich in rapeseed oil increases 24-h fat oxidation more than meal rich in palm oil. PLoS One 13, e0198858.CrossRefGoogle Scholar
Ministry of Health, Labour and Welfare of Japan (2020) Dietary Reference Intake for Japanese. Tokyo: Daiichi Shuppan Publishing Co, Ltd.Google Scholar
Tokuyama, K, Ogata, H, Katayose, Y, et al. (2009) Algorithm for transient response of whole body indirect calorimeter: deconvolution with a regularization parameter. J Appl Physiol 106, 640650.CrossRefGoogle ScholarPubMed
Ferrannini, E (1988) The theoretical bases of indirect calorimetry: a review. Metabolism 37, 287301.CrossRefGoogle ScholarPubMed
Ancoli-Israel, S, Cole, R, Alessi, C, et al. (2003) The role of actigraphy in the study of sleep and circadian rhythms. Sleep 26, 342392.CrossRefGoogle Scholar
Berry, RB, Budhiraja, R, Gottlieb, DJ, et al. (2012) Rules for scoring respiratory events in sleep: update of the 2007 AASM manual for the scoring of sleep and associated events. J Clin Sleep Med 8, 597619.CrossRefGoogle ScholarPubMed
Stein, PK & Kleiger, RE (1999) Insights from the study of heart rate variability. Annu Rev Med 50, 249261.CrossRefGoogle Scholar
Ogata, H, Horie, M, Kayaba, M, et al. (2020) Skipping breakfast for 6 d delayed the circadian rhythm of the body temperature without altering clock gene expression in human leukocytes. Nutrients 12, 118.CrossRefGoogle Scholar
Tanaka, Y, Ogata, H, Kayaba, M, et al. (2020) Effect of a single bout of exercise on clock gene expression in human leukocyte. J Appl Physiol 128, 847854.CrossRefGoogle ScholarPubMed
Muramatsu, H, Akimoto, N, Hashimoto, M, et al. (2021) Influence of polyunsaturated fatty acid intake on kidney functions of rats with chronic renal failure. Mar Drugs 19, 692.CrossRefGoogle ScholarPubMed
Bonsnes, RW & Taussky, HH (1945) On the colorimetric determination of creatinine by Jaffe reaction. J Biol Chem 158, 581591.CrossRefGoogle Scholar
Ishihara, A, Park, I, Suzuki, Y, et al. (2021) Metabolic responses to polychromatic LED and OLED light at night. Sci Rep 11, 12402.CrossRefGoogle ScholarPubMed
Minami, M, Takahashi, H, Inagaki, H, et al. (2009) Novel tryptamine-related substances, 5-sulphatoxydiacetyltryptamine, 5-hydroxydiacetyltryptamine, and reduced melatonin in human urine and the determination of those compounds, 6-sulphatoxymelatonin, and melatonin with fluorometric HPLC. J Chromatogr B Analyt Technol Biomed Life Sci 877, 814822.CrossRefGoogle ScholarPubMed
Kien, CL, Bunn, JY, Poynter, ME, et al. (2013) A lipidomics analysis of the relationship between dietary fatty acid composition and insulin sensitivity in young adults. Diabetes 62, 10541063.CrossRefGoogle ScholarPubMed
Dumas, JA, Bunn, JY, Nickerson, J, et al. (2016) Dietary saturated fat and monounsaturated fat have reversible effects on brain function and the secretion of pro-inflammatory cytokines in young women. Metabolism 65, 15821588.CrossRefGoogle ScholarPubMed
Rakhshandehroo, M, Knoch, B, Müller, M, et al. (2010) Peroxisome proliferator-activated receptor α target genes. PPAR Res 2010, 612089.CrossRefGoogle ScholarPubMed
Khodorova, NV, Rietman, A, Rutledge, DN, et al. (2021) Urinary medium-chained acyl-carnitines sign high caloric intake whereas short-chained acyl-carnitines sign high-protein diet within a high-fat, hypercaloric diet in a randomized crossover design dietary trial. Nutrients 13, 1191.CrossRefGoogle Scholar
Libert, R, van Hoof, F, Thillaye, M, et al. (2000) Identification of undescribed medium-chain acylcarnitines present in urine of patients with propionic and methylmalonic acidemias. Clin Chim Acta 295, 8796.CrossRefGoogle ScholarPubMed
Patterson, AD, Slanař, O, Krausz, KW, et al. (2009) Human urinary metabolomic profile of PPARRα induced fatty acid β-oxidation. J Proteome Res 8, 42934300.CrossRefGoogle ScholarPubMed
Stienstra, R, Duval, C, Müller, M, et al. (2007) PPARs, obesity, and inflammation. PPAR Res 2007, 95974.CrossRefGoogle ScholarPubMed
Dashti, HS, Follis, JL, Smith, CE, et al. (2015) Habitual sleep duration is associated with BMI and macronutrient intake and may be modified by CLOCK genetic variants. Am J Clin Nutr 101, 135143.CrossRefGoogle ScholarPubMed
St-Onge, MP, Roberts, A, Shechter, A, et al. (2016) Fiber and saturated fat are associated with sleep arousals and slow wave sleep. J Clin Sleep Med 12, 1924.CrossRefGoogle ScholarPubMed
Xu, W & Charoensuksai, P (2010) PPARs in rhythmic metabolic regulation and implications in health and disease. PPAR Res 2010, 243643.Google Scholar
Yang, X, Lamia, KA & Evans, RM (2007) Nuclear receptors, metabolism, and the circadian clock. Cold Spring Harb Symp Quant Biol 72, 387394.CrossRefGoogle ScholarPubMed
Reppert, SM & Weaver, DR (2002) Coordination of circadian timing in mammals. Nature 418, 935941.CrossRefGoogle ScholarPubMed
Chen, L & Yang, G (2014) PPARs integrate the mammalian clock and energy metabolism. PPAR Res 2014, 653017.CrossRefGoogle ScholarPubMed
Shirai, H, Oishi, K, Kudo, T, et al. (2007) PPARα is a potential therapeutic target of drugs to treat circadian rhythm sleep disorders. Biochem Biophys Res Commun 357, 679682.CrossRefGoogle ScholarPubMed
Chikahisa, S, Tominaga, K, Kawai, T, et al. (2008) Bezafibrate, a peroxisome proliferator-activated receptors agonist, decreases body temperature and enhances electroencephalogram delta-oscillation during sleep in mice. Endocrinology 149, 52625271.CrossRefGoogle ScholarPubMed
Griffin, EA Jr, Staknis, D & Weitz, CJ (1999) Light-independent role of CRY1 and CRY2 in the mammalian circadian clock. Science 286, 768771.CrossRefGoogle ScholarPubMed
Phillips, F, Crisp, AH, Mcguinness, B, et al. (1975) Isocaloric diet changes and electroencephalographic sleep. Lancet 2, 723725.CrossRefGoogle ScholarPubMed
Afaghi, A, O’Connor, H & Chow, CM (2008) Acute effects of the very low carbohydrate diet on sleep indices. Nutr Neurosci 11, 146154.CrossRefGoogle ScholarPubMed
Yajima, K, Seya, T, Iwayama, K, et al. (2014) Effects of nutrient composition of dinner on sleep architecture and energy metabolism during sleep. J Nutr Sci Vitaminol (Tokyo) 60, 114121.CrossRefGoogle ScholarPubMed
Kayaba, M, Park, I, Iwayama, K, et al. (2017) Energy metabolism differs between sleep stages and begins to increase prior to awakening. Metabolism 69, 1423.CrossRefGoogle ScholarPubMed
Begaye, B, Vinales, KL, Hollstein, T, et al. (2020) Impaired metabolic flexibility to high-fat overfeeding predicts future weight gain in healthy adults. Diabetes 69, 181192.CrossRefGoogle ScholarPubMed
Goodpaster, BH & Sparks, LM (2017) Metabolic flexibility in health and disease. Cell Metab 25, 10271036.CrossRefGoogle ScholarPubMed
Edmond, J, Robbins, RA, Bergstrom, JD, et al. (1987) Capacity for substrate utilization in oxidative metabolism by neurons, astrocytes, and oligodendrocytes from developing brain in primary culture. J Neurosci Res 18, 551561.CrossRefGoogle ScholarPubMed
Mak-Mccully, RA, Rolland, M, Sargsyan, A, et al. (2017) Coordination of cortical and thalamic activity during non-REM sleep in humans. Nat Commun 8, 15499.CrossRefGoogle ScholarPubMed
Nowak, N, Gaisl, T, Miladinovic, D, et al. (2021) Rapid and reversible control of human metabolism by individual sleep states. Cell Rep 37, 109903.CrossRefGoogle ScholarPubMed
Farhud, D & Aryan, Z (2018) Circadian rhythm, lifestyle and health: a narrative review. Iran J Public Health 47, 10681076.Google ScholarPubMed
Johnston, JD, Ordovás, JM, Scheer, FA, et al. (2016) Circadian rhythms, metabolism, and chrononutrition in rodents and humans. Adv Nutr 7, 399406.CrossRefGoogle ScholarPubMed
Almoosawi, S, Vingeliene, S, Karagounis, LG, et al. (2016) Chrono-nutrition: a review of current evidence from observational studies on global trends in time-of-day of energy intake and its association with obesity. Proc Nutr Soc 75, 487500.CrossRefGoogle ScholarPubMed
Baehr, EK, Revelle, W & Eastman, CI (2000) Individual differences in the phase and amplitude of the human circadian temperature rhythm: with an emphasis on morningness-eveningness. J Sleep Res 9, 117127.CrossRefGoogle ScholarPubMed
Archer, SN, Viola, AU, Kyriakopoulou, V, et al. (2008) Inter-individual differences in habitual sleep timing and entrained phase of endogenous circadian rhythms of BMAL1, PER2 and PER3 mRNA in human leukocytes. Sleep 31, 608617.CrossRefGoogle ScholarPubMed
Fukuya, H, Emoto, N, Nonaka, H, et al. (2007) Circadian expression of clock genes in human peripheral leukocytes. Biochem Biophys Res Commun 354, 924928.CrossRefGoogle ScholarPubMed
Kavcic, P, Rojc, B, Dolenc-Groselj, L, et al. (2011) The impact of sleep deprivation and nighttime light exposure on clock gene expression in humans. Croat Med J 52, 594603.CrossRefGoogle ScholarPubMed
Yoshida, C, Shikata, N, Seki, S, et al. (2012) Early nocturnal meal skipping alters the peripheral clock and increases lipogenesis in mice. Nutr Metab 9, 78.CrossRefGoogle ScholarPubMed
Shimizu, H, Hanzawa, F, Kim, D, et al. (2018) Delayed first active-phase meal, a breakfast skipping model, led to increased body weight and shifted the circadian oscillation of the hepatic clock and lipid metabolism-related genes in rats fed a high-fat diet. PLoS One 13, e0206669.CrossRefGoogle Scholar
Nováková, M, Polidarová, L, Sládek, M, et al. (2011) Restricted feeding regime affects clock gene expression profiles in the suprachiasmatic nucleus of rats exposed to constant light. Neurosci 197, 6571.CrossRefGoogle ScholarPubMed
Wehrens, SMT, Christou, S, Isherwood, C, et al. (2017) Meal timing regulates the human circadian system. Curr Biol 27, 17681775.CrossRefGoogle ScholarPubMed
Supplementary material: File

Yajima et al. supplementary material

Yajima et al. supplementary material 1

Download Yajima et al. supplementary material(File)
File 28.1 KB
Supplementary material: File

Yajima et al. supplementary material

Yajima et al. supplementary material 2

Download Yajima et al. supplementary material(File)
File 12.2 KB