Hostname: page-component-7c8c6479df-94d59 Total loading time: 0 Render date: 2024-03-28T02:24:16.726Z Has data issue: false hasContentIssue false

Adult male mice conceived by in vitro fertilization exhibit increased glucocorticoid receptor expression in fat tissue

Published online by Cambridge University Press:  29 October 2015

R. K. Simbulan
Affiliation:
Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
X. Liu
Affiliation:
Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
S. K. Feuer
Affiliation:
Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
E. Maltepe
Affiliation:
Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
A. Donjacour
Affiliation:
Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
P. Rinaudo*
Affiliation:
Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, San Francisco, CA, USA
*
*Address for correspondence: P. Rinaudo, Division of Reproductive Endocrinology and Infertility, 513 Parnassus Ave, HSW 1463E San Francisco, CA 94143, USA. (Email Paolo.Rinaudo@ucsf.edu)

Abstract

Prenatal development is highly plastic and readily influenced by the environment. Adverse conditions have been shown to alter organ development and predispose offspring to chronic diseases, including diabetes and hypertension. Notably, it appears that the changes in glucocorticoid hormones or glucocorticoid receptor (GR) levels in peripheral tissues could play a role in the development of chronic diseases. We have previously demonstrated that in vitro fertilization (IVF) and preimplantation embryo culture is associated with growth alterations and glucose intolerance in mice. However, it is unknown if GR signaling is affected in adult IVF offspring. Here we show that GR expression is increased in inbred (C57Bl6/J) and outbred (CF-1× B6D2F1/J) blastocysts following in vitro culture and elevated levels are also present in the adipose tissue of adult male mice. Importantly, genes involved in lipolysis and triglyceride synthesis and responsive to GR were also increased in adipose tissue, indicating that increased GR activates downstream gene pathways. The promoter region of GR, previously reported to be epigenetically modified by perinatal manipulation, showed no changes in DNA methylation status. Our findings demonstrate that IVF results in a long-term change in GR gene expression in a sex- and tissue-specific manner. These changes in adipose tissues may well contribute to the metabolic phenotype in mice conceived by IVF.

Type
Original Article
Copyright
© Cambridge University Press and the International Society for Developmental Origins of Health and Disease 2015 

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

1. Barker, D. Mothers, Babies, and Health in Later Life, 2nd edn, 1998. Churchill Livingstone: Edinburgh.Google Scholar
2. Godfrey, KM, Barker, DJ. Fetal programming and adult health. Public Health Nutr. 2001; 4, 611624.CrossRefGoogle ScholarPubMed
3. Langley-Evans, SC, Gardner, DS, Jackson, AA. Maternal protein restriction influences the programming of the rat hypothalamic-pituitary-adrenal axis. J Nutr. 1996; 126, 15781585.CrossRefGoogle ScholarPubMed
4. Ford, SP, Hess, BW, Schwope, MM, et al. Maternal undernutrition during early to mid-gestation in the ewe results in altered growth, adiposity, and glucose tolerance in male offspring. J Anim Sci. 2007; 85, 12851294.CrossRefGoogle ScholarPubMed
5. Barker, DJ, Osmond, C. Infant mortality, childhood nutrition, and ischaemic heart disease in England and Wales. Lancet. 1986; 1, 10771081.CrossRefGoogle ScholarPubMed
6. Barker, DJ. The developmental origins of insulin resistance. Horm Res. 2005; 64(Suppl. 3), 27.Google ScholarPubMed
7. Fowden, AL, Forhead, AJ. Endocrine mechanisms of intrauterine programming. Reproduction. 2004; 127, 515526.CrossRefGoogle ScholarPubMed
8. Benediktsson, R, Lindsay, RS, Noble, J, Seckl, JR, Edwards, CR. Glucocorticoid exposure in utero: new model for adult hypertension. Lancet. 1993; 341, 339341.CrossRefGoogle ScholarPubMed
9. Nyirenda, MJ, Lindsay, RS, Kenyon, CJ, Burchell, A, Seckl, JR. Glucocorticoid exposure in late gestation permanently programs rat hepatic phosphoenolpyruvate carboxykinase and glucocorticoid receptor expression and causes glucose intolerance in adult offspring. J Clin Invest. 1998; 101, 21742181.CrossRefGoogle ScholarPubMed
10. Rinaudo, PF, Lamb, J. Fetal origins of perinatal morbidity and/or adult disease. Semin Reprod Med. 2008; 26(5), 436445.CrossRefGoogle ScholarPubMed
11. Rinaudo, P, Wang, E. Fetal programming and metabolic syndrome. Annu Rev Physiol. 2012; 74, 107130.CrossRefGoogle ScholarPubMed
12. Jaddoe, VW, Witteman, JC. Hypotheses on the fetal origins of adult diseases: contributions of epidemiological studies. Eur J Epidemiol. 2006; 21, 91102.CrossRefGoogle ScholarPubMed
13. Liu, D, Diorio, J, Tannenbaum, B, et al. Maternal care, hippocampal glucocorticoid receptors, and hypothalamic-pituitary-adrenal responses to stress. Science. 1997; 277, 16591662.CrossRefGoogle ScholarPubMed
14. Bertram, C, Trowern, AR, Copin, N, Jackson, AA, Whorwood, CB. The maternal diet during pregnancy programs altered expression of the glucocorticoid receptor and type 2 11beta-hydroxysteroid dehydrogenase: potential molecular mechanisms underlying the programming of hypertension in utero. Endocrinology. 2001; 142, 28412853.CrossRefGoogle ScholarPubMed
15. Turner, JD, Alt, SR, Cao, L, et al. Transcriptional control of the glucocorticoid receptor: CpG islands, epigenetics and more. Biochem Pharmacol. 2010; 80, 18601868.CrossRefGoogle ScholarPubMed
16. Weaver, IC, Cervoni, N, Champagne, FA, et al. Epigenetic programming by maternal behavior. Nat Neurosci. 2004; 7, 847854.CrossRefGoogle ScholarPubMed
17. Lillycrop, KA, Slater-Jefferies, JL, Hanson, MA, et al. Induction of altered epigenetic regulation of the hepatic glucocorticoid receptor in the offspring of rats fed a protein-restricted diet during pregnancy suggests that reduced DNA methyltransferase-1 expression is involved in impaired DNA methylation and changes in histone modifications. Br J Nutr. 2007; 97, 10641073.CrossRefGoogle ScholarPubMed
18. Whorwood, CB, Donovan, SJ, Flanagan, D, Phillips, DI, Byrne, CD. Increased glucocorticoid receptor expression in human skeletal muscle cells may contribute to the pathogenesis of the metabolic syndrome. Diabetes. 2002; 51, 10661075.CrossRefGoogle Scholar
19. Feuer, SK, Camarano, L, Rinaudo, PF. ART and health: clinical outcomes and insights on molecular mechanisms from rodent studies. Mol Hum Reprod. 2013; 19, 189204.CrossRefGoogle ScholarPubMed
20. Schieve, LA, Meikle, SF, Ferre, C, et al. Low and very low birth weight in infants conceived with use of assisted reproductive technology. N Engl J Med. 2002; 346, 731737.CrossRefGoogle ScholarPubMed
21. Ceelen, M, van Weissenbruch, MM, Vermeiden, JP, van Leeuwen, FE, Delemarre-van de Waal, HA. Cardiometabolic differences in children born after in vitro fertilization: follow-up study. J Clin Endocrinol Metab. 2008; 93, 16821688.CrossRefGoogle ScholarPubMed
22. Ceelen, M, van Weissenbruch, MM, Vermeiden, JP, van Leeuwen, FE, Delemarre-van de Waal, HA. Growth and development of children born after in vitro fertilization. Fertil Steril. 2008; 90, 16621673.CrossRefGoogle ScholarPubMed
23. Giritharan, G, Talbi, S, Donjacour, A, et al. Effect of in vitro fertilization on gene expression and development of mouse preimplantation embryos. Reproduction. 2007; 134, 6372.CrossRefGoogle ScholarPubMed
24. Rinaudo, PF, Giritharan, G, Talbi, S, Dobson, AT, Schultz, RM. Effects of oxygen tension on gene expression in preimplantation mouse embryos. Fertil Steril. 2006; 86(4 Suppl.), 12521265.CrossRefGoogle ScholarPubMed
25. Fernandez-Gonzalez, R, de Dios Hourcade, J, Lopez-Vidriero, I, et al. Analysis of gene transcription alterations at the blastocyst stage related to the long-term consequences of in vitro culture in mice. Reproduction. 2009; 137, 271283.CrossRefGoogle Scholar
26. Delle Piane, L, Lin, W, Liu, X, et al. Effect of the method of conception and embryo transfer procedure on mid-gestation placenta and fetal development in an IVF mouse model. Hum Reprod. 2010; 25, 20392046.CrossRefGoogle Scholar
27. Bloise, E, Lin, W, Liu, X, et al. Impaired placental nutrient transport in mice generated by in vitro fertilization. Endocrinology. 2012; 153, 34573467.CrossRefGoogle ScholarPubMed
28. Ecker, DJ, Stein, P, Xu, Z, et al. Long-term effects of culture of preimplantation mouse embryos on behavior. Proc Natl Acad Sci U S A. 2004; 101, 15951600.CrossRefGoogle ScholarPubMed
29. Donjacour, A, Liu, X, Lin, W, Simbulan, R, Rinaudo, PF. In vitro fertilization affects growth and glucose metabolism in a sex-specific manner in an outbred mouse model. Biol Reprod. 2014; 90, 80.CrossRefGoogle Scholar
30. Feuer, SK, Liu, X, Donjacour, A, et al. Use of a mouse in vitro fertilization model to understand the developmental origins of health and disease hypothesis. Endocrinology. 2014; 155, 19561969.CrossRefGoogle ScholarPubMed
31. Rexhaj, E, Paoloni-Giacobino, A, Rimoldi, SF, et al. Mice generated by in vitro fertilization exhibit vascular dysfunction and shortened life span. J Clin Invest. 2013; 123, 50525060.CrossRefGoogle ScholarPubMed
32. Feuer, SK, Donjacour, A, Simbulan, RK, et al. Sexually dimorphic effect of in vitro fertilization (IVF) on adult mouse fat and liver metabolomes. Endocrinology. 2014; 155(11), 45544567.CrossRefGoogle ScholarPubMed
33. Yu, CY, Mayba, O, Lee, JV, et al. Genome-wide analysis of glucocorticoid receptor binding regions in adipocytes reveal gene network involved in triglyceride homeostasis. PLoS One. 2010; 5, e15188.CrossRefGoogle ScholarPubMed
34. Rinaudo, P, Schultz, RM. Effects of embryo culture on global pattern of gene expression in preimplantation mouse embryos. Reproduction. 2004; 128, 301311.CrossRefGoogle ScholarPubMed
35. Baxter, JD, Rousseau, GG. Glucocorticoid hormone action: an overview. Monogr Endocrinol. 1979; 12, 124.CrossRefGoogle ScholarPubMed
36. Burnstein, KL, Cidlowski, JA. The down side of glucocorticoid receptor regulation. Mol Cell Endocrinol. 1992; 83, C1C8.CrossRefGoogle ScholarPubMed
37. Makino, Y, Yoshikawa, N, Okamoto, K, et al. Direct association with thioredoxin allows redox regulation of glucocorticoid receptor function. J Biol Chem. 1999; 274, 31823188.CrossRefGoogle ScholarPubMed
38. Dong, Y, Aronsson, M, Gustafsson, JA, Okret, S. The mechanism of cAMP-induced glucocorticoid receptor expression. Correlation to cellular glucocorticoid response. J Biol Chem. 1989; 264, 1367913683.CrossRefGoogle ScholarPubMed
39. Manejwala, FM, Schultz, RM. Blastocoel expansion in the preimplantation mouse embryo: stimulation of sodium uptake by cAMP and possible involvement of cAMP-dependent protein kinase. Dev Biol. 1989; 136, 560563.CrossRefGoogle ScholarPubMed
40. Goto, Y, Noda, Y, Mori, T, Nakano, M. Increased generation of reactive oxygen species in embryos cultured in vitro . Free Radic Biol Med. 1993; 15, 6975.CrossRefGoogle ScholarPubMed
41. Svec, F. Glucocorticoid receptor regulation. Life Sci. 1985; 36, 23592366.CrossRefGoogle ScholarPubMed
42. Vegiopoulos, A, Herzig, S. Glucocorticoids, metabolism and metabolic diseases. Mol Cell Endocrinol. 2007; 275, 4361.CrossRefGoogle ScholarPubMed
43. Walker, BR. Cortisol--cause and cure for metabolic syndrome? Diabet Med. 2006; 23, 12811288.CrossRefGoogle ScholarPubMed
44. Weinstock, M. The long-term behavioural consequences of prenatal stress. Neurosci Biobehav Rev. 2008; 32, 10731086.CrossRefGoogle ScholarPubMed
45. Negron-Perez, VM, Echevarria, FD, Huffman, SR, Rivera, RM. Determination of allelic expression of h19 in pre- and peri-implantation mouse embryos. Biol Rep. 2013; 88, 97.CrossRefGoogle ScholarPubMed
46. Reik, W, Dean, W, Walter, J. Epigenetic reprogramming in mammalian development. Science. 2001; 293, 10891093.CrossRefGoogle ScholarPubMed
47. Reik, W. Stability and flexibility of epigenetic gene regulation in mammalian development. Nature. 2007; 447, 425432.CrossRefGoogle Scholar
48. Doherty, AS, Mann, MR, Tremblay, KD, Bartolomei, MS, Schultz, RM. Differential effects of culture on imprinted H19 expression in the preimplantation mouse embryo. Biol Reprod. 2000; 62, 15261535.CrossRefGoogle ScholarPubMed
49. Kember, RL, Dempster, EL, Lee, TH, et al. Maternal separation is associated with strain-specific responses to stress and epigenetic alterations to Nr3c1, Avp, and Nr4a1 in mouse. Brain Behav. 2012; 2(4), 455467.CrossRefGoogle Scholar
50. Lesage, J, Dufourny, L, Laborie, C, et al. Perinatal malnutrition programs sympathoadrenal and hypothalamic-pituitary-adrenal axis responsiveness to restraint stress in adult male rats. J Neuroendocrinol. 2002; 14, 135143.CrossRefGoogle ScholarPubMed
51. Leonhardt, M, Lesage, J, Dufourny, L, et al. Perinatal maternal food restriction induces alterations in hypothalamo-pituitary-adrenal axis activity and in plasma corticosterone-binding globulin capacity of weaning rat pups. Neuroendocrinology. 2002; 75, 4554.CrossRefGoogle ScholarPubMed
52. Sheppard, KE, Roberts, JL, Blum, M. Adrenocorticotropin-releasing factor down-regulates glucocorticoid receptor expression in mouse corticotrope tumor cells via an adenylate cyclase-dependent mechanism. Endocrinology. 1991; 129, 663670.CrossRefGoogle ScholarPubMed
53. Strata, F, Giritharan, G, Sebastiano, FD, et al. Behavior and brain gene expression changes in mice exposed to preimplantation and prenatal stress. Reprod Sci. 2015; 22, 2330.CrossRefGoogle ScholarPubMed