Publication Cover
Chronobiology International
The Journal of Biological and Medical Rhythm Research
Volume 32, 2015 - Issue 5
647
Views
14
CrossRef citations to date
0
Altmetric
Original Article

Maternal obesity disrupts circadian rhythms of clock and metabolic genes in the offspring heart and liver

, , &
Pages 615-626 | Received 27 Nov 2014, Accepted 02 Mar 2015, Published online: 30 Apr 2015

REFERENCES

  • Asher G, Schibler U. (2011). Crosstalk between components of circadian and metabolic cycles in mammals. Cell Metab. 13:125–37
  • Ashino NG, Saito KN, Souza FD, et al. (2012). Maternal high-fat feeding through pregnancy and lactation predisposes mouse offspring to molecular insulin resistance and fatty liver. J Nutr Biochem. 23:341–8
  • Barker DJ. (2007). The origins of the developmental origins theory. J Intern Med. 261:412–17
  • Barker DJ, Gluckman PD, Godfrey KM, et al. (1993). Fetal nutrition and cardiovascular disease in adult life. Lancet 341:938–41
  • Bass J, Takahashi JS. (2010). Circadian integration of metabolism and energetics. Science 330:1349–54
  • Challier JC, Basu S, Bintein T, et al. (2008). Obesity in pregnancy stimulates macrophage accumulation and inflammation in the placenta. Placenta 29:274–81
  • Damiola F, Le Minh N, Preitner N, et al. (2000). Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev. 14:2950–61
  • Froy O. (2012). Circadian rhythms and obesity in mammals. ISRN Obesity 2012:1–12
  • Glatz JF, Luiken JJ, Bonen A. (2001). Involvement of membrane-associated proteins in the acute regulation of cellular fatty acid uptake. J Mol Neurosci. 16:123–32 (discussion 151–27)
  • Guillaumond F, Dardente H, Giguere V, Cermakian N. (2005). Differential control of Bmal1 circadian transcription by REV-ERB and ROR nuclear receptors. J Biol Rhythms. 20:391–403
  • Hara R, Wan K, Wakamatsu H, et al. (2001). Restricted feeding entrains liver clock without participation of the suprachiasmatic nucleus. Genes Cells 6:269–78
  • Hotamisligil GS. (2006). Inflammation and metabolic disorders. Nature 444:860–7
  • Howie GJ, Sloboda DM, Reynolds CM, Vickers MH. (2013). Timing of maternal exposure to a high fat diet and development of obesity and hyperinsulinemia in male rat offspring: same metabolic phenotype, different developmental pathways? J Nutr Metab. 2013:517384
  • Huang J, Lu C, Chen S, et al. (2010). Postnatal ontogenesis of clock genes in mouse suprachiasmatic nucleus and heart. Lipids Health Dis. 9:22
  • Huang W, Ramsey KM, Marcheva B, Bass J. (2011). Circadian rhythms, sleep, and metabolism. J Clin Invest. 121:2133–41
  • Jaaskelainen A, Pussinen J, Nuutinen O, et al. (2011). Intergenerational transmission of overweight among Finnish adolescents and their parents: A 16-year follow-up study. Int J Obes (Lond). 35:1289–94
  • Jones HN, Woollett LA, Barbour N, et al. (2009). High-fat diet before and during pregnancy causes marked up-regulation of placental nutrient transport and fetal overgrowth in C57/BL6 mice. FASEB J. 23:271–8
  • Kohsaka A, Laposky AD, Ramsey KM, et al. (2007). High-fat diet disrupts behavioral and molecular circadian rhythms in mice. Cell Metab. 6:414–21
  • Li M, Reynolds CM, Sloboda DM, et al. (2013). Effects of taurine supplementation on hepatic markers of inflammation and lipid metabolism in mothers and offspring in the setting of maternal obesity. PLoS One 8:e76961
  • Liu C, Li S, Liu T, et al. (2007). Transcriptional coactivator PGC-1α integrates the mammalian clock and energy metabolism. Nature 447:477–81
  • Livak KJ, Schmittgen TD. (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2−ΔΔCT method. Methods. 25:402–8
  • Luzzo KM, Wang Q, Purcell SH, et al. (2012). High fat diet induced developmental defects in the mouse: Oocyte meiotic aneuploidy and fetal growth retardation/brain defects. PLoS One 7:e49217
  • Melo AM, Benatti RO, Ignacio-Souza LM, et al. (2014). Hypothalamic endoplasmic reticulum stress and insulin resistance in offspring of mice dams fed high-fat diet during pregnancy and lactation. Metabolism 63:682–92
  • Neville MC, Allen JC, Archer PC, et al. (1991). Studies in human lactation: Milk volume and nutrient composition during weaning and lactogenesis. Am J Clin Nutr. 54:81–92
  • Nicholas KR, Hartmann PE. (1991). Milk secretion in the rat: Progressive changes in milk composition during lactation and weaning and the effect of diet. Comp Biochem Physiol A Comp Physiol. 98:535–42
  • Olson CM, Strawderman MS, Dennison BA. (2009). Maternal weight gain during pregnancy and child weight at age 3 years. Matern Child Health J. 13:839–46
  • Panda S, Antoch MP, Miller BH, et al. (2002). Coordinated transcription of key pathways in the mouse by the circadian clock. Cell 109:307–20
  • Park EJ, Lee JH, Yu GY, et al. (2010). Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 140:197–208
  • Portaluppi F, Smolensky MH, Touitou Y. (2010). Ethics and methods for biological rhythm research on animals and human beings. Chronobiol Int. 27:1911–29
  • Preitner N, Damiola F, Lopez-Molina L, et al. (2002). The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell 110:251–60
  • Reynolds CM, Li M, et al. (2013). Pre-weaning growth hormone treatment ameliorates bone marrow macrophage inflammation in adult male rat offspring following maternal undernutrition. PLoS One 8:e68262
  • Reynolds RM, Osmond C, Phillips DI, Godfrey KM. (2010). Maternal BMI, parity, and pregnancy weight gain: Influences on offspring adiposity in young adulthood. J Clin Endocrinol Metab. 95:5365–9
  • Richards J, Gumz ML. (2012). Advances in understanding the peripheral circadian clocks. FASEB J. 26:3602–13
  • Sakamoto K, Oishi K, Nagase T, et al. (2002). Circadian expression of clock genes during ontogeny in the rat heart. Neuroreport. 13:1239–42
  • Samuelsson AM, Matthews PA, Argenton M, et al. (2008). Diet-induced obesity in female mice leads to offspring hyperphagia, adiposity, hypertension, and insulin resistance: A novel murine model of developmental programming. Hypertension 51:383–92
  • Sandovici I, Hoelle K, Angiolini E, Constancia M. (2012). Placental adaptations to the maternal-fetal environment: Implications for fetal growth and developmental programming. Reprod Biomed Online 25:68–89
  • Sato TK, Panda S, Miraglia LJ, et al. (2004). A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron 43:527–37
  • Schibler U, Ripperger J, Brown SA. (2003). Peripheral circadian oscillators in mammals: Time and food. J Biol Rhythms 18:250–60
  • Segovia SA, Vickers MH, Gray C, Reynolds CM. (2014). Maternal obesity, inflammation, and developmental programming. BioMed Res Int. 2014:1–14
  • Shi SQ, Ansari TS, McGuinness OP, et al. (2013). Circadian disruption leads to insulin resistance and obesity. Curr Biol. 23:372–81
  • Silverman BL, Rizzo TA, Cho NH, Metzger BE. (1998). Long-term effects of the intrauterine environment: The Northwestern University Diabetes in Pregnancy Center. Diabetes Care 21:B142–9
  • Solt LA, Kojetin DJ, Burris TP. (2011). The REV-ERBs and RORs: Molecular links between circadian rhythms and lipid homeostasis. Future Med Chem. 3:623–38
  • Sookoian SC, Borengasser SJ, Kang P, et al. (2014). High fat diet and in utero exposure to maternal obesity disrupts circadian rhythm and leads to metabolic programming of liver in rat offspring. PLoS One 9:e84209
  • Sumova A, Bendova Z, Sladek M, et al. (2006). Setting the biological time in central and peripheral clocks during ontogenesis. FEBS Lett. 580:2836–42
  • Suter MA, Ma J, Vuguin PM, et al. (2014). In utero exposure to a maternal high-fat diet alters the epigenetic histone code in a murine model. Am J Obstet Gynecol. 210:463. e1–11
  • Suwazono Y, Nogawa K. (2014). Effect of shift work on blood pressure. Nihon Rinsho. 72:1497–502
  • Taegtmeyer H, Sen S, Vela D. (2010). Return to the fetal gene program: A suggested metabolic link to gene expression in the heart. Ann NY Acad Sci. 1188:191–8
  • Tao W, Chen S, Shi G, et al. (2011). SWItch/sucrose nonfermentable (SWI/SNF) complex subunit BAF60a integrates hepatic circadian clock and energy metabolism. Hepatology 54:1410–20
  • Trottier G, Koski KG, Brun T, et al. (1998). Increased fat intake during lactation modifies hypothalamic-pituitary-adrenal responsiveness in developing rat pups: A possible role for leptin. Endocrinology 139:3704–11
  • van der Veen DR, Minh NL, Gos P, et al. (2006). Impact of behavior on central and peripheral circadian clocks in the common vole Microtus arvalis, a mammal with ultradian rhythms. Proc Natl Acad Sci USA 103:3393–8
  • Whitaker RC. (2004). Predicting preschooler obesity at birth: The role of maternal obesity in early pregnancy. Pediatrics 114:e29–36
  • Wu Z, Zhao J, Xu H, et al. (2014). Maternal quercetin administration during gestation and lactation decrease endoplasmic reticulum stress and related inflammation in the adult offspring of obese female rats. Eur J Nutr. 53:1669–83
  • Yin SA. (2008). The investigation of Chinese women's nutrition and health (fertile women, pregnant women and lactating women)—the 2002 China nutrition and health survey. Beijing, China: People's Medical Publishing House

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.