Skip to main content

EDITORIAL article

Front. Endocrinol., 16 July 2020
Sec. Translational Endocrinology
This article is part of the Research Topic Decoding the fetal circadian system and its role in adult sickness and health: Melatonin, a dark history. View all 6 articles

Editorial: Decoding the Fetal Circadian System and Its Role in Adult Sickness and Health: Melatonin, a Dark History

  • 1Laboratory of Developmental Chronobiology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
  • 2Centro Interdisciplinario de Estudios del Sistema Nervioso (CISNe), Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
  • 3Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
  • 4Department of Biology, Washington University, St. Louis, MO, United States

Throughout gestation, the newborn follows a precise development, preparing for a successful transition to extrauterine life. Maternal circadian signals not only play a key role in the precise daily delivery of oxygen, nutrients, hormones, and other biophysical signals but also synchronize daily rhythms in the fetus to the external photoperiod. As a result of changes in fetal demands during pregnancy, the maternal cardiac, respiratory, immune, renal, hepatic, and gastrointestinal, and endocrine systems differ from those of non-pregnant women (1, 2). The circadian system is not an exception; pregnancy-induced functional reorganization of daily rhythms in physiology and behavior has been reported in several species (35). Indeed, Martin-Fairey et al. (6) recently reported that pregnancy induces an earlier chronotype of activity in mice and women, with decreases in total activity close to delivery. This suggests that the maternal suprachiasmatic nucleus (SCN) and/or other components of the circadian system are modified during gestation. Consequently, this current special issue explores our current understanding about the role played by the circadian system in maternal well-being, fetal development, and diseases in adult life [(6); McCarthy et al.; van Dalum et al.; Beñaldo et al.; Mendez et al.].

As explained by McCarthy et al., melatonin serves as a key hormone signal from mother to fetus in several species, including humans. Expanding on this, articles in this issue by the groups of England and Herzog review how melatonin or other daily maternal signals may contribute to fetal development and the timing of birth and how circadian disruption may lead to adverse birth outcomes including preterm delivery [(6); McCarthy et al.; Bates and Herzog]. van Dalum et al. highlight how maternal photoperiodic programming, through a well-defined action of melatonin on the pars tuberalis, may provide an experimental paradigm to investigate the mechanisms by which early life experience establishes long-term patterns of hypothalamic regulation. For example, a recent report found that melatonin suppression during gestation resulted in decreased hepatic transcript levels of four enzymes involved in DNA methylation; maternal melatonin treatment reversed the effect in three of these enzymes. These results indicate that maternal melatonin modifies the epigenome in the fetal liver and opens the possibility of effects in other fetal organs (7).

Accordingly, Torres-Farfan et al. (7) and Cipolla-Neto and Amaral (8) provide insight into how melatonin treatment may be a therapeutic against circadian misalignment. Importantly, special attention should be taken about the precise timing of melatonin treatment. For example, the recent studies of Beñaldo et al. in sheep found that melatonin treatment when the newborn did not produce melatonin induced negative effects in cardiovascular function and in response to chronic hypoxia.

Finally, Mendez et al. report here that the rat offspring from mothers exposed to chronic photoperiod shifts suffer several alterations in the maturation of renal and immune function, consistent with adult hypertension (9). Taken together, the articles in this special issue emphasize how irregular daily schedules during pregnancy can impact maternal–fetal communication and fetal development and put the offspring at risk for adult disorders including hypertension, type II diabetes, and obesity.

Author Contributions

All authors listed have made a substantial, direct and intellectual contribution to the work, and approved it for publication.

Funding

This work was supported by Grants 1191207 (CT-F) from Fondo Nacional de Desarrollo Científico y Tecnológico, Chile (FONDECYT), and FAPESP 2014/50457-0 (JC) and Grant NIGMS 131403 (EH).

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Serón-Ferré M, Richter HG, Valenzuela GJ, Torres-Farfan C. Circadian rhythms in the fetus and newborn: significance of interactions with maternal physiology and the environment. In: Walker DW, editor. Prenatal and Postnatal Determinants of Development. Vol. 109. New York, NY: Springer (2016). p. 147–65. doi: 10.1007/978-1-4939-3014-2_7

CrossRef Full Text | Google Scholar

2. Cunningham FG, Leveno KJ, Bloom S, Gilstrap L, Cunningham FG. Williams Obstetrics. 23rd ed. New York, NY: McGraw-Hill Professional Publishing (2010).

Google Scholar

3. Schrader JA, Nunez AA, Smale L. Changes in and dorsal to the rat suprachiasmatic nucleus during early pregnancy. Neuroscience. (2010) 171:513–23. doi: 10.1016/j.neuroscience.2010.08.057

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Schrader JA, Smale L, Nunez AA. Pregnancy affects FOS rhythms in brain regions regulating sleep/wake state and body temperature in rats. Brain Res. (2012) 1480:53–60. doi: 10.1016/j.brainres.2012.09.003

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Schrader JA, Nunez AA, Smale L. Site-specific changes in brain extra-SCN oscillators during early pregnancy in the rat. J Biol Rhythms. (2011) 26:363–7. doi: 10.1177/0748730411409714

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Martin-Fairey CA, Zhao P, Wan L, Roenneberg T, Fay J, Ma X, et al. Pregnancy induces an earlier chronotype in both mice and women. J Biol Rhythms. (2019) 34:323–31. doi: 10.1177/0748730419844650

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Torres-Farfan C, Mendez N, Ehrenfeld P, Seron-Ferre M. In utero circadian changes; facing light pollution. Curr Opin Physiol. (2020) 13:128–34. doi: 10.1016/j.cophys.2019.11.005

CrossRef Full Text | Google Scholar

8. Cipolla-Neto J, do Amaral FG. Melatonin as a hormone: new physiological and clinical insights. Endocr Rev. (2018) 39:990–1028. doi: 10.1210/er.2018-00084

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Mendez N, Diego Halabi, Carlos Spichiger, Esteban R., Salazar, Karina Vergara, Pamela Alonso-Vasquez, et al. Gestational chronodisruption impairs circadian physiology in rat male offspring, increasing the risk of chronic disease. Endocrinology. (2016) 157:4654–68. doi: 10.1210/en.2016-1282

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: circadian rhythms, melatonin, DOHaD (developmental origins of health and disease), pregnancy, epigenetic

Citation: Torres-Farfan C, Cipolla Neto J and Herzog ED (2020) Editorial: Decoding the Fetal Circadian System and Its Role in Adult Sickness and Health: Melatonin, a Dark History. Front. Endocrinol. 11:380. doi: 10.3389/fendo.2020.00380

Received: 06 May 2020; Accepted: 13 May 2020;
Published: 16 July 2020.

Edited and reviewed by: James M. Olcese, Florida State University, United States

Copyright © 2020 Torres-Farfan, Cipolla Neto and Herzog. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Claudia Torres-Farfan, claudia.torres@uach.cl

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.