Prague Med. Rep. 2015, 116, 40-48

https://doi.org/10.14712/23362936.2015.44

Daily Profiles of Dehydroepiandrosterone and Its Hydroxylated Metabolites with Respect to Food Intake

Luboslav Stárka, Beata Rácz, Monika Šrámková, Martin Hill, Michaela Dušková

Institute of Endocrinology, Prague, Czech Republic

Received January 23, 2015
Accepted March 12, 2015

References

1. Baulieu, E. E. (1996) Dehydroepiandrosterone (DHEA): a fountain of youth? J. Clin. Endocrinol. Metab. 81(9), 3147–3151. <https://doi.org/10.1210/jcem.81.9.8784058>
2. Ghiciuc, C. M., Cozma-Dima, C. L., Pasquali, V., Renzi, P., Simeoni, S., Lupusoru, C. E., Patacchioli, F. R. (2011) Awakening responses and diurnal fluctuations of salivary cortisol, DHEA-S and α-amylase in healthy male subjects. Neuro Endocrinol. Lett. 32(4), 475–480.
3. Hampl, R., Morfin, R., Stárka, L. (1997) 7-hydroxylated C19-steroids: what are they good for? Endocr. Regul. 31, 211–218.
4. Hampl, R., Stárka, L. (2000) Minireview: 16α-hydroxylated metabolites of dehydroepiandrosterone and their biological significance. Endocr. Regul. 34(3), 161–163.
5. Heaney, J. L., Phillips, A. C., Carroll, D. (2012) Ageing, physical function, and the diurnal rhythms of cortisol and dehydroepiandrosterone. Psychoneuroendocrinology 37(3), 341–349. <https://doi.org/10.1016/j.psyneuen.2011.07.001>
6. Hennebert, O., Chalbot, S., Alran, S., Morfin, R. (2007a) Dehydroepiandrosterone 7alpha-hydroxylation in human tissues: Possible interference with type 1 11beta-hydroxysteroid dehydrogenase-mediated processes. J. Steroid Biochem. Mol. Biol. 104(3–5), 326–333. <https://doi.org/10.1016/j.jsbmb.2007.03.026>
7. Hennebert, O., Pernelle, C., Ferroud, C., Morfin, R. (2007b) 7alpha- and 7beta-hydroxy-epiandrosterone as substrates and inhibitors for the human 11beta-hydroxysteroid dehydrogenase type 1. J. Steroid Biochem. Mol. Biol. 105(1–5), 159–165. <https://doi.org/10.1016/j.jsbmb.2006.11.021>
8. Hill, M., Pařízek, A., Cibula, D., Kancheva, R., Jirásek, J. E., Jirkovská, M., Velíková, M., Kubátová, J., Klímková, M., Pašková, A., Žižka, Z., Kancheva, L., Kazihnitková, H., Zamrazilová, L., Stárka, L. (2010) Steroid metabolome in fetal and maternal body fluids in human late pregnancy. J. Steroid Biochem. Mol. Biol. 122, 114–132. <https://doi.org/10.1016/j.jsbmb.2010.05.007>
9. Hill, M., Dušková, M., Stárka, L. (2015) Dehydroepiandrosterone, its metabolites and ion channels. J. Steroid Biochem. Mol. Biol. 145, 293–314. <https://doi.org/10.1016/j.jsbmb.2014.05.006>
10. Máčová, L., Bičíková, M., Zamrazilová, H., Hill, M., Kazihnitková, H., Sedláčková, B., Stárka, L. (2014) Reduced levels of circulating 7alpha-hydroxy-dehydroepiandrosterone in treated adolescent obese patients. Physiol. Res. 63(1), 95–101.
11. Meloun, M., Hill, M., Militký, J., Kupka, A. K. (2000) Transformation in the PC-aided biochemical data analysis. Clin. Chem. Lab. Med. 38, 553–559. <https://doi.org/10.1515/CCLM.2000.081>
12. Meloun, M., Militký, J., Hill, M., Brereton, R. G. (2002) Crucial problems in regression modelling and their solutions. Analyst 127, 433–450. <https://doi.org/10.1039/b110779h>
13. Meloun, M., Hill, M., Militký, J., Vrbíková, J., Stanická, S., Škrha, J. (2004) New methodology of influential point detection in regression model building for the prediction of metabolic clearance rate of glucose. Clin. Chem. Lab. Med. 42, 311–322. <https://doi.org/10.1515/CCLM.2004.057>
14. Mikulecky, M., Kreze, A., Putz, Z., Moravcik, M. (1995) Daily variation of serum cortisol, 17-hydroxyprogesterone and five androgens in healthy women. Braz. J. Med. Biol. Res. 28(4), 485–490.
15. Morfin, R., Stárka, L. (2001) Neurosteroid 7-hydroxylation products in the brain. Int. Rev. Neurobiol. 46, 79–95. <https://doi.org/10.1016/S0074-7742(01)46059-4>
16. Neaves, W. B., Johnson, L., Porter, J. C., Parker, C. R. Jr., Petty, C. S. (1984) Leydig cell numbers, daily sperm production, and serum gonadotropin levels in aging men. J. Clin. Endocrinol. Metab. 59(4), 756–763. <https://doi.org/10.1210/jcem-59-4-756>
17. Ohlsson, C., Vandenput, L., Tivesten, Å. (2015) DHEA and mortality: What is the nature of the association? J. Steroid Biochem. Mol. Biol. 145, 248–253. <https://doi.org/10.1016/j.jsbmb.2014.03.006>
18. Prom-Wormley, E. C., York, T. P., Jacobson, K. C., Eaves, L. J., Mendoza, S. P., Hellhammer, D., Maninger, N., Levine, S., Lupien, S., Lyons, M. J., Hauger, R., Xian, H., Franz, C. E., Kremen, W. S. (2011) Genetic and environmental effects on diurnal dehydroepiandrosterone sulfate concentrations in middle-aged men. Psychoneuroendocrinology 36(10), 1441–1452. <https://doi.org/10.1016/j.psyneuen.2011.03.018> <PubMed>
19. Raju, U., Bradlow, H. L., Skidmore, F. D., Levitz, M. (1989) The concentration of 16x-hydroxy androgens in serum and cyst fluid of women with gross cystic disease of the breast. Steroids 54(1), 101–112. <https://doi.org/10.1016/0039-128X(89)90077-9>
20. Sedláčková, B., Dušátková, L., Zamrazilová, H., Matucha, P., Bičíková, M., Stárka, L. (2012) 7-oxygenated derivatives of dehydroepiandrosterone and obesity. Prague Med. Rep. 113(2), 147–155. <https://doi.org/10.14712/23362936.2015.29>
21. Stárka, L., Šulcová, J., Šilink, K. (1962) Die Harnausscheidung des 7-Hydroxydehydroepiandrosteronsulfats. Clin. Chim. Acta 7, 309–316. <https://doi.org/10.1016/0009-8981(62)90029-3>
22. Stárka, L., Dušková, M., Hill, M. (2015) Dehydroepiandrosterone as a neurosteroid. J. Steroid Biochem. Mol. Biol. 145, 254–260. <https://doi.org/10.1016/j.jsbmb.2014.03.008>
23. Warren, W. B., Gurewitsch, E. D., Goland, R. S. (1995) Corticotropin-releasing hormone and pituitary-adrenal hormones in pregnancies complicated by chronic hypertension. Am. J. Obstet. Gynecol. 172(2 Pt 1), 661–666. <https://doi.org/10.1016/0002-9378(95)90589-8>
front cover

ISSN 1214-6994 (Print) ISSN 2336-2936 (Online)

Archive