Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Original Article
  • Published:

Adipocyte and Cell Biology

Regulation of the clock gene expression in human adipose tissue by weight loss

Abstract

Background:

The circadian clock coordinates numerous metabolic processes to adapt physiological responses to light–dark and feeding regimens and is itself regulated by metabolic cues. The implication of the circadian clock in the regulation of energy balance and body weight is widely studied in rodents but not in humans. Here we investigated (1) whether the expression of clock genes in human adipose tissue is changed by weight loss and (2) whether these alterations are associated with metabolic parameters.

Subjects/Methods:

Subcutaneous adipose tissue (SAT) samples were collected before and after 8 weeks of weight loss on an 800 kcal per day hypocaloric diet (plus 200 g per day vegetables) at the same time of the day. Fifty overweight subjects who lost at least 8% weight after 8 weeks were selected for the study. The expression of 10 clock genes and key metabolic and inflammatory genes in adipose tissue was determined by quantitative real-time PCR.

Results:

The expression of core clock genes PER2 and NR1D1 was increased after the weight loss. Correlations of PERIOD expression with body mass index (BMI) and serum total, high-density lipoprotein and low-density lipoprotein (LDL) cholesterol levels and of NR1D1 expression with total and LDL cholesterol were found that became non-significant after correction for multiple testing. Clock gene expression levels and their weight loss-induced changes tightly correlated with each other and with genes involved in fat metabolism (FASN, CPT1A, LPL, PPARG, PGC1A, ADIPOQ), energy metabolism (SIRT1), autophagy (LC3A, LC3B) and inflammatory response (NFKB1, NFKBIA, NLRP3, EMR1).

Conclusion:

Clock gene expression in human SAT is regulated by body weight changes and associated with BMI, serum cholesterol levels and the expression of metabolic and inflammatory genes. Our data confirm the tight crosstalk between molecular clock and metabolic and inflammatory pathways involved in adapting adipose tissue metabolism to changes of the energy intake in humans.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1
Figure 2

Similar content being viewed by others

References

  1. Asher G, Sassone-Corsi P . Time for food: the intimate interplay between nutrition, metabolism, and the circadian clock. Cell 2015; 161: 84–92.

    Article  CAS  Google Scholar 

  2. Huang W, Ramsey KM, Marcheva B, Bass J . Circadian rhythms, sleep, and metabolism. J Clin Invest 2011; 121: 2133–2141.

    Article  CAS  Google Scholar 

  3. Loboda A, Kraft WK, Fine B, Joseph J, Nebozhyn M, Zhang C et al. Diurnal variation of the human adipose transcriptome and the link to metabolic disease. BMC Med Genomics 2009; 2: 7.

    Article  Google Scholar 

  4. Eckel-Mahan KL, Patel VR, de Mateo S, Orozco-Solis R, Ceglia NJ, Sahar S et al. Reprogramming of the circadian clock by nutritional challenge. Cell 2013; 155: 1464–1478.

    Article  CAS  Google Scholar 

  5. Keller M, Mazuch J, Abraham U, Eom GD, Herzog ED, Volk HD et al. A circadian clock in macrophages controls inflammatory immune responses. Proc Natl Acad Sci USA 2009; 106: 21407–21412.

    Article  CAS  Google Scholar 

  6. Damiola F . Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev 2000; 14: 2950–2961.

    Article  CAS  Google Scholar 

  7. Hatori M, Vollmers C, Zarrinpar A, DiTacchio L, Bushong EA, Gill S et al. Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab 2012; 15: 848–860.

    Article  CAS  Google Scholar 

  8. Kohsaka A, Laposky AD, Ramsey KM, Estrada C, Joshu C, Kobayashi Y et al. High-fat diet disrupts behavioral and molecular circadian rhythms in mice. Cell Metab 2007; 6: 414–421.

    Article  CAS  Google Scholar 

  9. Pivovarova O, Jurchott K, Rudovich N, Hornemann S, Lu Y, Mockel S et al. Changes of dietary fat and carbohydrate content alter central and peripheral clock in humans. J Clin Endocrinol Metab 2015; 100: 2291–2302.

    Article  CAS  Google Scholar 

  10. Kumar Jha P, Challet E, Kalsbeek A . Circadian rhythms in glucose and lipid metabolism in nocturnal and diurnal mammals. Mol Cell Endocrinol 2015; 418: 74–88.

    Article  CAS  Google Scholar 

  11. Adamovich Y, Rousso-Noori L, Zwighaft Z, Neufeld-Cohen A, Golik M, Kraut-Cohen J et al. Circadian clocks and feeding time regulate the oscillations and levels of hepatic triglycerides. Cell Metab 2014; 19: 319–330.

    Article  CAS  Google Scholar 

  12. Benedict C, Shostak A, Lange T, Brooks SJ, Schioth HB, Schultes B et al. Diurnal rhythm of circulating nicotinamide phosphoribosyltransferase (Nampt/visfatin/PBEF): impact of sleep loss and relation to glucose metabolism. J Clin Endocrinol Metab 2012; 97: E218–E222.

    Article  CAS  Google Scholar 

  13. Gavrila A . Diurnal and ultradian dynamics of serum adiponectin in healthy men: comparison with leptin, circulating soluble leptin receptor, and cortisol patterns. J Clin Endocrinol Metab 2003; 88: 2838–2843.

    Article  CAS  Google Scholar 

  14. Turek FW . Obesity and metabolic syndrome in circadian clock mutant mice. Science 2005; 308: 1043–1045.

    Article  CAS  Google Scholar 

  15. Delezie J, Dumont S, Dardente H, Oudart H, Grechez-Cassiau A, Klosen P et al. The nuclear receptor REV-ERB alpha is required for the daily balance of carbohydrate and lipid metabolism. FASEB J 2012; 26: 3321–3335.

    Article  CAS  Google Scholar 

  16. Yang S, Liu A, Weidenhammer A, Cooksey RC, McClain D, Kim MK et al. The role of mPer2 clock gene in glucocorticoid and feeding rhythms. Endocrinology 2009; 150: 2153–2160.

    Article  CAS  Google Scholar 

  17. Paschos GK, Ibrahim S, Song WL, Kunieda T, Grant G, Reyes TM et al. Obesity in mice with adipocyte-specific deletion of clock component Arntl. Nat Med 2012; 18: 1768–1777.

    Article  CAS  Google Scholar 

  18. Garaulet M . PERIOD2 variants are associated with abdominal obesity, psycho-behavioral factors, and attrition in the dietary treatment of obesity. J Am Diet Assoc 2011; 110: 917–921.

    Article  Google Scholar 

  19. Garaulet M, Tardido AE, Lee YC, Smith CE, Parnell LD, Ordovas JM . SIRT1 and CLOCK 3111 T>C combined genotype is associated with evening preference and weight loss resistance in a behavioral therapy treatment for obesity. Int J Obesity 2012; 36: 1436–1441.

    Article  CAS  Google Scholar 

  20. Maury E, Ramsey KM, Bass J . Circadian rhythms and metabolic syndrome: from experimental genetics to human disease. Circ Res 2010; 106: 447–462.

    Article  CAS  Google Scholar 

  21. Ando H, Takamura T, Matsuzawa-Nagata N, Shima KR, Eto T, Misu H et al. Clock gene expression in peripheral leucocytes of patients with type 2 diabetes. Diabetologia 2009; 52: 329–335.

    Article  CAS  Google Scholar 

  22. Gomez-Abellan P, Hernandez-Morante JJ, Lujan JA, Madrid JA, Garaulet M . Clock genes are implicated in the human metabolic syndrome. Int J Obes (Lond) 2008; 32: 121–128.

    Article  CAS  Google Scholar 

  23. Gogebakan O, Kohl A, Osterhoff MA, van Baak MA, Jebb SA, Papadaki A et al. Effects of weight loss and long-term weight maintenance with diets varying in protein and glycemic index on cardiovascular risk factors: the diet, obesity, and genes (DiOGenes) study: a randomized, controlled trial. Circulation 2011; 124: 2829–2838.

    Article  Google Scholar 

  24. Rossmeislova L, Malisova L, Kracmerova J, Stich V . Adaptation of human adipose tissue to hypocaloric diet. Int J Obes (Lond) 2013; 37: 640–650.

    Article  CAS  Google Scholar 

  25. de Kreutzenberg SV, Ceolotto G, Papparella I, Bortoluzzi A, Semplicini A, Dalla Man C et al. Downregulation of the longevity-associated protein sirtuin 1 in insulin resistance and metabolic syndrome: potential biochemical mechanisms. Diabetes 2010; 59: 1006–1015.

    Article  Google Scholar 

  26. Kovsan J, Bluher M, Tarnovscki T, Kloting N, Kirshtein B, Madar L et al. Altered autophagy in human adipose tissues in obesity. J Clin Endocrinol Metab 2011; 96: E268–E277.

    Article  CAS  Google Scholar 

  27. Dasu MR, Devaraj S, Park S, Jialal I . Increased Toll-like receptor (TLR) activation and TLR ligands in recently diagnosed type 2 diabetic subjects. Diabetes Care 2010; 33: 861–868.

    Article  CAS  Google Scholar 

  28. Lumeng CN, Bodzin JL, Saltiel AR . Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 2007; 117: 175–184.

    Article  CAS  Google Scholar 

  29. Gomez-Santos C, Gomez-Abellan P, Madrid JA, Hernandez-Morante JJ, Lujan JA, Ordovas JM et al. Circadian rhythm of clock genes in human adipose explants. Obesity 2009; 17: 1481–1485.

    Article  CAS  Google Scholar 

  30. Garaulet M, Gomez-Abellan P, Alburquerque-Bejar JJ, Lee YC, Ordovas JM, Scheer FA . Timing of food intake predicts weight loss effectiveness. Int J Obes (Lond) 2013; 37: 604–611.

    Article  CAS  Google Scholar 

  31. Kahleova H, Belinova L, Malinska H, Oliyarnyk O, Trnovska J, Skop V et al. Eating two larger meals a day (breakfast and lunch) is more effective than six smaller meals in a reduced-energy regimen for patients with type 2 diabetes: a randomised crossover study. Diabetologia 2014; 57: 1552–1560.

    Article  Google Scholar 

  32. Li Y, Sato Y, Yamaguchi N . Shift work and the risk of metabolic syndrome: a nested case-control study. Int J Occup Environ Health 2011; 17: 154–160.

    Article  Google Scholar 

  33. Le Martelot G, Claudel T, Gatfield D, Schaad O, Kornmann B, Lo Sasso G et al. REV-ERBalpha participates in circadian SREBP signaling and bile acid homeostasis. PLoS Biol 2009; 7: e1000181.

    Article  Google Scholar 

  34. Grimaldi B, Bellet MM, Katada S, Astarita G, Hirayama J, Amin RH et al. PER2 controls lipid metabolism by direct regulation of PPARgamma. Cell Metab 2010; 12: 509–520.

    Article  CAS  Google Scholar 

  35. Soussi H, Reggio S, Alili R, Prado C, Mutel S, Pini M et al. DAPK2 down-regulation associates with attenuated adipocyte autophagic clearance in human obesity. Diabetes 2015; 64: 3452–3463.

    Article  CAS  Google Scholar 

  36. Ma D, Panda S, Lin JD . Temporal orchestration of circadian autophagy rhythm by C/EBPbeta. EMBO J 2011; 30: 4642–4651.

    Article  CAS  Google Scholar 

  37. Nakahata Y, Sahar S, Astarita G, Kaluzova M, Sassone-Corsi P . Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1. Science 2009; 324: 654–657.

    Article  CAS  Google Scholar 

  38. Lamia KA, Sachdeva UM, DiTacchio L, Williams EC, Alvarez JG, Egan DF et al. AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation. Science 2009; 326: 437–440.

    Article  CAS  Google Scholar 

  39. Ramsey KM, Yoshino J, Brace CS, Abrassart D, Kobayashi Y, Marcheva B et al. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science 2009; 324: 651–654.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank all study participants for their cooperation. We also thank Andreas Wagner, Melanie Hannemann and Anja Henkel for technical assistance. The study was supported by grants of the European Union Food Quality and Safety Priority of the Sixth Framework Program (FP6-2005-513946), of the European Community (contract no. FOOD-CT-2005-513946) and of the German Science Foundation (DFG grant KFO218 PF164/16-1 to OP, AK and AFHP).

Author contributions

AFHP, OG and OP designed the research; OP, SS, JG, VM and KK conducted the research; OP, MO and NR analyzed data and performed the statistical analysis; and OP, AK and AFHP wrote and edited the manuscript; OP had the primary responsibility for the final content. All authors read and approved the final manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to O Pivovarova.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies this paper on International Journal of Obesity website

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pivovarova, O., Gögebakan, Ö., Sucher, S. et al. Regulation of the clock gene expression in human adipose tissue by weight loss. Int J Obes 40, 899–906 (2016). https://doi.org/10.1038/ijo.2016.34

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ijo.2016.34

This article is cited by

Search

Quick links