Skip to main content
Top
Published in: Reproductive Biology and Endocrinology 1/2020

Open Access 01-12-2020 | Epigenetics | Review

Review of psychological stress on oocyte and early embryonic development in female mice

Authors: Qiu-Yue Zhai, Jun-Jie Wang, Yu Tian, Xiaofang Liu, Zhenhua Song

Published in: Reproductive Biology and Endocrinology | Issue 1/2020

Login to get access

Abstract

Psychological stress can cause adverse health effects in animals and humans. Accumulating evidence suggests that psychological stress in female mice is associated with ovarian developmental abnormalities accompanied by follicle and oocyte defects. Oocyte and early embryonic development are impaired in mice facing psychological stress, likely resulting from hormone signalling disorders, reactive oxygen species (ROS) accumulation and alterations in epigenetic modifications, which are primarily mediated by the hypothalamic-pituitary-adrenal (HPA) and hypothalamic-pituitary-ovarian (HPO) axes. The present evidence suggests that psychological stress is increasingly becoming the most common causative factor for female subfertility. Here, we review recent progress on the impact of psychological stress on female reproduction, particularly for oocyte and early embryonic development in female mice. This review highlights the connection between psychological stress and reproductive health and provides novel insight on human subfertility.
Literature
1.
go back to reference Schneiderman N, Ironson G, Siegel SD. Stress and health: psychological, behavioral, and biological determinants. Annu Rev Clin Psychol. 2005;1:607–28.PubMedPubMedCentralCrossRef Schneiderman N, Ironson G, Siegel SD. Stress and health: psychological, behavioral, and biological determinants. Annu Rev Clin Psychol. 2005;1:607–28.PubMedPubMedCentralCrossRef
2.
go back to reference Cohen S, Kessler RC, Gordon LU, JMsAgfh, scientists s. Strategies for measuring stress in studies of psychiatric and physical disorders; 1995. p. 3–26. Cohen S, Kessler RC, Gordon LU, JMsAgfh, scientists s. Strategies for measuring stress in studies of psychiatric and physical disorders; 1995. p. 3–26.
3.
go back to reference Van Praag HM, de Kloet ER, Van Os J. Stress, the brain and depression: Cambridge University Press; 2004. Van Praag HM, de Kloet ER, Van Os J. Stress, the brain and depression: Cambridge University Press; 2004.
4.
5.
6.
go back to reference Abe H, Hidaka N, Kawagoe C, Odagiri K, Watanabe Y, Ikeda T, Ishizuka Y, Hashiguchi H, Takeda R, Nishimori T, Ishida Y. Prenatal psychological stress causes higher emotionality, depression-like behavior, and elevated activity in the hypothalamo-pituitary-adrenal axis. Neurosci Res. 2007;59:145–51.PubMedCrossRef Abe H, Hidaka N, Kawagoe C, Odagiri K, Watanabe Y, Ikeda T, Ishizuka Y, Hashiguchi H, Takeda R, Nishimori T, Ishida Y. Prenatal psychological stress causes higher emotionality, depression-like behavior, and elevated activity in the hypothalamo-pituitary-adrenal axis. Neurosci Res. 2007;59:145–51.PubMedCrossRef
7.
go back to reference Lin TK, Zhong L, Santiago JL. Association between stress and the HPA Axis in the atopic dermatitis. Int J Mol Sci. 2017;18. Lin TK, Zhong L, Santiago JL. Association between stress and the HPA Axis in the atopic dermatitis. Int J Mol Sci. 2017;18.
8.
go back to reference Hall JM, Podawiltz A, Mummert DI, Jones H, Mummert ME. Psychological stress and the cutaneous immune response: roles of the HPA axis and the sympathetic nervous system in atopic dermatitis and psoriasis, vol. 2012; 2012. Hall JM, Podawiltz A, Mummert DI, Jones H, Mummert ME. Psychological stress and the cutaneous immune response: roles of the HPA axis and the sympathetic nervous system in atopic dermatitis and psoriasis, vol. 2012; 2012.
9.
go back to reference Holsboer F. The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology. 2000;23:477–501.PubMedCrossRef Holsboer F. The corticosteroid receptor hypothesis of depression. Neuropsychopharmacology. 2000;23:477–501.PubMedCrossRef
10.
go back to reference Pariante CM, Lightman SL. The HPA axis in major depression: classical theories and new developments. Trends Neurosci. 2008;31:464–8.PubMedCrossRef Pariante CM, Lightman SL. The HPA axis in major depression: classical theories and new developments. Trends Neurosci. 2008;31:464–8.PubMedCrossRef
11.
go back to reference Elenkov IJ, Chrousos GP. Stress system--organization, physiology and immunoregulation. Neuroimmunomodulation. 2006;13:257–67.PubMedCrossRef Elenkov IJ, Chrousos GP. Stress system--organization, physiology and immunoregulation. Neuroimmunomodulation. 2006;13:257–67.PubMedCrossRef
12.
go back to reference Bernardini R, Chiarenza A, Kamilaris TC, Renaud N, Lempereur L, Demitrack M, Gold PW, Chrousos GP. In vivo and in vitro effects of arginine-vasopressin receptor antagonists on the hypothalamic-pituitary-adrenal axis in the rat. Neuroendocrinology. 1994;60:503–8.PubMedCrossRef Bernardini R, Chiarenza A, Kamilaris TC, Renaud N, Lempereur L, Demitrack M, Gold PW, Chrousos GP. In vivo and in vitro effects of arginine-vasopressin receptor antagonists on the hypothalamic-pituitary-adrenal axis in the rat. Neuroendocrinology. 1994;60:503–8.PubMedCrossRef
13.
go back to reference Garg A, Chren MM, Sands LP, Matsui MS, Marenus KD, Feingold KR, Elias PM. Psychological stress perturbs epidermal permeability barrier homeostasis: implications for the pathogenesis of stress-associated skin disorders. Arch Dermatol. 2001;137:53–9.PubMedCrossRef Garg A, Chren MM, Sands LP, Matsui MS, Marenus KD, Feingold KR, Elias PM. Psychological stress perturbs epidermal permeability barrier homeostasis: implications for the pathogenesis of stress-associated skin disorders. Arch Dermatol. 2001;137:53–9.PubMedCrossRef
14.
go back to reference Copper RL, Goldenberg RL, Das A, Elder N, Swain M, Norman G, Ramsey R, Cotroneo P, Collins BA, Johnson F, et al. The preterm prediction study: maternal stress is associated with spontaneous preterm birth at less than thirty-five weeks' gestation. National Institute of Child Health and Human Development maternal-fetal medicine units network. Am J Obstet Gynecol. 1996;175:1286–92.PubMedCrossRef Copper RL, Goldenberg RL, Das A, Elder N, Swain M, Norman G, Ramsey R, Cotroneo P, Collins BA, Johnson F, et al. The preterm prediction study: maternal stress is associated with spontaneous preterm birth at less than thirty-five weeks' gestation. National Institute of Child Health and Human Development maternal-fetal medicine units network. Am J Obstet Gynecol. 1996;175:1286–92.PubMedCrossRef
15.
go back to reference Klonoff-Cohen H, Chu E, Natarajan L, Sieber W. A prospective study of stress among women undergoing in vitro fertilization or gamete intrafallopian transfer. Fertil Steril. 2001;76:675–87.CrossRefPubMed Klonoff-Cohen H, Chu E, Natarajan L, Sieber W. A prospective study of stress among women undergoing in vitro fertilization or gamete intrafallopian transfer. Fertil Steril. 2001;76:675–87.CrossRefPubMed
16.
go back to reference Neggers Y, Goldenberg R, Cliver S, Hauth J. The relationship between psychosocial profile, health practices, and pregnancy outcomes. Acta Obstet Gynecol Scand. 2006;85:277–85.PubMedCrossRef Neggers Y, Goldenberg R, Cliver S, Hauth J. The relationship between psychosocial profile, health practices, and pregnancy outcomes. Acta Obstet Gynecol Scand. 2006;85:277–85.PubMedCrossRef
17.
go back to reference Stickel S, Wagels L, Wudarczyk O, Jaffee S, Habel U, Schneider F, Chechko N. Neural correlates of depression in women across the reproductive lifespan - an fMRI review. J Affect Disord. 2019;246:556–70.PubMedCrossRef Stickel S, Wagels L, Wudarczyk O, Jaffee S, Habel U, Schneider F, Chechko N. Neural correlates of depression in women across the reproductive lifespan - an fMRI review. J Affect Disord. 2019;246:556–70.PubMedCrossRef
18.
go back to reference Schroder AK, Katalinic A, Diedrich K, Ludwig M. Cumulative pregnancy rates and drop-out rates in a German IVF programme: 4102 cycles in 2130 patients. Reprod BioMed Online. 2004;8:600–6.PubMedCrossRef Schroder AK, Katalinic A, Diedrich K, Ludwig M. Cumulative pregnancy rates and drop-out rates in a German IVF programme: 4102 cycles in 2130 patients. Reprod BioMed Online. 2004;8:600–6.PubMedCrossRef
19.
go back to reference Damti OB, Sarid O, Sheiner E, Zilberstein T. Cwikel J: [stress and distress in infertility among women]. Harefuah. 2008;147:256–60 276.PubMed Damti OB, Sarid O, Sheiner E, Zilberstein T. Cwikel J: [stress and distress in infertility among women]. Harefuah. 2008;147:256–60 276.PubMed
20.
go back to reference Liu YX, Cheng YN, Miao YL, Wei DL, Zhao LH, Luo MJ, Tan JH. Psychological stress on female mice diminishes the developmental potential of oocytes: a study using the predatory stress model. PLoS One. 2012;7:e48083.PubMedPubMedCentralCrossRef Liu YX, Cheng YN, Miao YL, Wei DL, Zhao LH, Luo MJ, Tan JH. Psychological stress on female mice diminishes the developmental potential of oocytes: a study using the predatory stress model. PLoS One. 2012;7:e48083.PubMedPubMedCentralCrossRef
21.
go back to reference Zhang SY, Wang JZ, Li JJ, Wei DL, Sui HS, Zhang ZH, Zhou P, Tan JH. Maternal restraint stress diminishes the developmental potential of oocytes. Biol Reprod. 2011;84:672–81.PubMedCrossRef Zhang SY, Wang JZ, Li JJ, Wei DL, Sui HS, Zhang ZH, Zhou P, Tan JH. Maternal restraint stress diminishes the developmental potential of oocytes. Biol Reprod. 2011;84:672–81.PubMedCrossRef
22.
go back to reference Zhou P, Lian HY, Cui W, Wei DL, Li Q, Liu YX, Liu XY, Tan JH. Maternal-restraint stress increases oocyte aneuploidy by impairing metaphase I spindle assembly and reducing spindle assembly checkpoint proteins in mice. Biol Reprod. 2012;86:83.PubMedCrossRef Zhou P, Lian HY, Cui W, Wei DL, Li Q, Liu YX, Liu XY, Tan JH. Maternal-restraint stress increases oocyte aneuploidy by impairing metaphase I spindle assembly and reducing spindle assembly checkpoint proteins in mice. Biol Reprod. 2012;86:83.PubMedCrossRef
23.
go back to reference Wiebold JL, Stanfield PH, Becker WC, Hillers JK. The effect of restraint stress in early pregnancy in mice. J Reprod Fertil. 1986;78:185–92.PubMedCrossRef Wiebold JL, Stanfield PH, Becker WC, Hillers JK. The effect of restraint stress in early pregnancy in mice. J Reprod Fertil. 1986;78:185–92.PubMedCrossRef
24.
go back to reference Sugino N, Nakamura Y, Okuno N, Shimamura K, Teyama T, Ishimatsu M, Kato H. Effects of restraint stress on luteal function in rats during mid-pregnancy. J Reprod Fertil. 1994;101:23–6.PubMedCrossRef Sugino N, Nakamura Y, Okuno N, Shimamura K, Teyama T, Ishimatsu M, Kato H. Effects of restraint stress on luteal function in rats during mid-pregnancy. J Reprod Fertil. 1994;101:23–6.PubMedCrossRef
25.
go back to reference Mairesse J, Lesage J, Breton C, Bréant B, Hahn T, Darnaudéry M, Dickson SL, Seckl J, Blondeau B, Vieau DJ. Metabolism: Maternal stress alters endocrine function of the feto-placental unit in rats. Am J Physiol Endocrinol Metab. 2007;292:E1526–33.PubMedCrossRef Mairesse J, Lesage J, Breton C, Bréant B, Hahn T, Darnaudéry M, Dickson SL, Seckl J, Blondeau B, Vieau DJ. Metabolism: Maternal stress alters endocrine function of the feto-placental unit in rats. Am J Physiol Endocrinol Metab. 2007;292:E1526–33.PubMedCrossRef
26.
go back to reference Smith J, Ferguson D, Jauregui G, Panarace M, Medina M, Lehnert S, Hill JR. Short-term maternal psychological stress in the post-conception period in ewes affects fetal growth and gestation length. Reproduction. 2008;136:259–66.PubMedCrossRef Smith J, Ferguson D, Jauregui G, Panarace M, Medina M, Lehnert S, Hill JR. Short-term maternal psychological stress in the post-conception period in ewes affects fetal growth and gestation length. Reproduction. 2008;136:259–66.PubMedCrossRef
27.
go back to reference Peltoniemi OA, Love RJ, Heinonen M, Tuovinen V, Saloniemi H. Seasonal and management effects on fertility of the sow: a descriptive study. Anim Reprod Sci. 1999;55:47–61.PubMedCrossRef Peltoniemi OA, Love RJ, Heinonen M, Tuovinen V, Saloniemi H. Seasonal and management effects on fertility of the sow: a descriptive study. Anim Reprod Sci. 1999;55:47–61.PubMedCrossRef
28.
go back to reference Saiti D, Lacham-Kaplan O. Mouse Germ Cell Development in-vivo and in-vitro. Biomark Insights. 2007;2:117727190700200024.CrossRef Saiti D, Lacham-Kaplan O. Mouse Germ Cell Development in-vivo and in-vitro. Biomark Insights. 2007;2:117727190700200024.CrossRef
30.
go back to reference Pepling ME, Spradling AC. Mouse ovarian germ cell cysts undergo programmed breakdown to form primordial follicles. Dev Biol. 2001;234:339–51.PubMedCrossRef Pepling ME, Spradling AC. Mouse ovarian germ cell cysts undergo programmed breakdown to form primordial follicles. Dev Biol. 2001;234:339–51.PubMedCrossRef
31.
go back to reference Edson MA, Nagaraja AK, Matzuk MM. The mammalian ovary from genesis to revelation. Endocrine Rev. 2009;30:624–712.CrossRef Edson MA, Nagaraja AK, Matzuk MM. The mammalian ovary from genesis to revelation. Endocrine Rev. 2009;30:624–712.CrossRef
32.
go back to reference Baerwald AR, Adams GP, Pierson RA. Ovarian antral folliculogenesis during the human menstrual cycle: a review. Hum Reprod Update. 2012;18:73–91.CrossRefPubMed Baerwald AR, Adams GP, Pierson RA. Ovarian antral folliculogenesis during the human menstrual cycle: a review. Hum Reprod Update. 2012;18:73–91.CrossRefPubMed
35.
go back to reference Saitou M, Yamaji M. Primordial germ cells in mice. Cold Spring Harb Perspect Biol. 2012;4. Saitou M, Yamaji M. Primordial germ cells in mice. Cold Spring Harb Perspect Biol. 2012;4.
36.
go back to reference Carson DD, Bagchi I, Dey SK, Enders AC, Fazleabas AT, Lessey BA, Yoshinaga K. Embryo implantation. Dev Biol. 2000;223:217–37.PubMedCrossRef Carson DD, Bagchi I, Dey SK, Enders AC, Fazleabas AT, Lessey BA, Yoshinaga K. Embryo implantation. Dev Biol. 2000;223:217–37.PubMedCrossRef
37.
go back to reference Kala M, Nivsarkar M. Role of cortisol and superoxide dismutase in psychological stress induced anovulation. Gen Comp Endocrinol. 2016;225:117–24.PubMedCrossRef Kala M, Nivsarkar M. Role of cortisol and superoxide dismutase in psychological stress induced anovulation. Gen Comp Endocrinol. 2016;225:117–24.PubMedCrossRef
38.
go back to reference Wu LM, Liu YS, Tong XH, Shen N, Jin RT, Han H, Hu MH, Wang W, Zhou GX. Inhibition of follicular development induced by chronic unpredictable stress is associated with growth and differentiation factor 9 and gonadotropin in mice. Biol Reprod. 2012;86:121.PubMed Wu LM, Liu YS, Tong XH, Shen N, Jin RT, Han H, Hu MH, Wang W, Zhou GX. Inhibition of follicular development induced by chronic unpredictable stress is associated with growth and differentiation factor 9 and gonadotropin in mice. Biol Reprod. 2012;86:121.PubMed
39.
go back to reference Gong S, Miao YL, Jiao GZ, Sun MJ, Li H, Lin J, Luo MJ, Tan JH. Dynamics and correlation of serum cortisol and corticosterone under different physiological or stressful conditions in mice. PLoS One. 2015;10:e0117503.PubMedPubMedCentralCrossRef Gong S, Miao YL, Jiao GZ, Sun MJ, Li H, Lin J, Luo MJ, Tan JH. Dynamics and correlation of serum cortisol and corticosterone under different physiological or stressful conditions in mice. PLoS One. 2015;10:e0117503.PubMedPubMedCentralCrossRef
40.
go back to reference Gao Y, Chen F, Kong QQ, Ning SF, Yuan HJ, Lian HY, Luo MJ, Tan JH. Stresses on female mice impair oocyte developmental potential: effects of stress severity and duration on oocytes at the growing follicle stage. Reprod Sci. 2016;23:1148–57.PubMedCrossRef Gao Y, Chen F, Kong QQ, Ning SF, Yuan HJ, Lian HY, Luo MJ, Tan JH. Stresses on female mice impair oocyte developmental potential: effects of stress severity and duration on oocytes at the growing follicle stage. Reprod Sci. 2016;23:1148–57.PubMedCrossRef
41.
go back to reference Sun J, Guo Y, Zhang Q, Bu S, Li B, Wang Q, Lai DJCC. Chronic restraint stress disturbs meiotic resumption through APC/C-mediated cyclin B1 excessive degradation in mouse oocytes. Cell Cycle. 2018;17:1591–601.PubMedPubMedCentralCrossRef Sun J, Guo Y, Zhang Q, Bu S, Li B, Wang Q, Lai DJCC. Chronic restraint stress disturbs meiotic resumption through APC/C-mediated cyclin B1 excessive degradation in mouse oocytes. Cell Cycle. 2018;17:1591–601.PubMedPubMedCentralCrossRef
42.
go back to reference McArthur R, Borsini F. Animal models of depression in drug discovery: a historical perspective. Pharmacol Biochem Behav. 2006;84:436–52.PubMedCrossRef McArthur R, Borsini F. Animal models of depression in drug discovery: a historical perspective. Pharmacol Biochem Behav. 2006;84:436–52.PubMedCrossRef
43.
go back to reference Sugino N, Takiguchi S, Kashida S, Karube A, Nakamura Y, Kato H. Superoxide dismutase expression in the human corpus luteum during the menstrual cycle and in early pregnancy. Mol Hum Reprod. 2000;6:19–25.PubMedCrossRef Sugino N, Takiguchi S, Kashida S, Karube A, Nakamura Y, Kato H. Superoxide dismutase expression in the human corpus luteum during the menstrual cycle and in early pregnancy. Mol Hum Reprod. 2000;6:19–25.PubMedCrossRef
44.
go back to reference Golub MS, Campbell MA, Kaufman FL, Iyer P, Li LH, Donald JM, Morgan JE. Effects of restraint stress in gestation: implications for rodent developmental toxicology studies. Birth Defects Res B Dev Reprod Toxicol. 2004;71:26–36.PubMedCrossRef Golub MS, Campbell MA, Kaufman FL, Iyer P, Li LH, Donald JM, Morgan JE. Effects of restraint stress in gestation: implications for rodent developmental toxicology studies. Birth Defects Res B Dev Reprod Toxicol. 2004;71:26–36.PubMedCrossRef
45.
go back to reference Ghatebi M, Zavareh S, Lashkarbolouki T, Elahdadi Salmani M. Implications from early life stress on the development of mouse ovarian follicles: focus on oxidative stress. J Obstet Gynaecol Res. 2019;45:1506–14.PubMedCrossRef Ghatebi M, Zavareh S, Lashkarbolouki T, Elahdadi Salmani M. Implications from early life stress on the development of mouse ovarian follicles: focus on oxidative stress. J Obstet Gynaecol Res. 2019;45:1506–14.PubMedCrossRef
46.
go back to reference Wu LM, Hu MH, Tong XH, Han H, Shen N, Jin RT, Wang W, Zhou GX, He GP, Liu YS. Chronic unpredictable stress decreases expression of brain-derived neurotrophic factor (BDNF) in mouse ovaries: relationship to oocytes developmental potential. PLoS One. 2012;7:e52331.PubMedPubMedCentralCrossRef Wu LM, Hu MH, Tong XH, Han H, Shen N, Jin RT, Wang W, Zhou GX, He GP, Liu YS. Chronic unpredictable stress decreases expression of brain-derived neurotrophic factor (BDNF) in mouse ovaries: relationship to oocytes developmental potential. PLoS One. 2012;7:e52331.PubMedPubMedCentralCrossRef
47.
go back to reference Higaki S, Kishi M, Koyama K, Nagano M, Katagiri S, Takada T, Takahashi Y. Early germinal vesicle breakdown is a predictor of high preimplantation developmental competent oocytes in mice. Zygote. 2017;25:41–8.PubMedCrossRef Higaki S, Kishi M, Koyama K, Nagano M, Katagiri S, Takada T, Takahashi Y. Early germinal vesicle breakdown is a predictor of high preimplantation developmental competent oocytes in mice. Zygote. 2017;25:41–8.PubMedCrossRef
48.
go back to reference Wu XF, Yuan HJ, Li H, Gong S, Lin J, Miao YL, Wang TY, Tan JH. Restraint stress on female mice diminishes the developmental potential of oocytes: roles of chromatin configuration and histone modification in germinal vesicle stage oocytes. Biol Reprod. 2015;92:13.PubMed Wu XF, Yuan HJ, Li H, Gong S, Lin J, Miao YL, Wang TY, Tan JH. Restraint stress on female mice diminishes the developmental potential of oocytes: roles of chromatin configuration and histone modification in germinal vesicle stage oocytes. Biol Reprod. 2015;92:13.PubMed
49.
go back to reference Dorfman M, Arancibia S, Fiedler JL, Lara HE. Chronic intermittent cold stress activates ovarian sympathetic nerves and modifies ovarian follicular development in the rat. Biol Reprod. 2003;68:2038–43.PubMedCrossRef Dorfman M, Arancibia S, Fiedler JL, Lara HE. Chronic intermittent cold stress activates ovarian sympathetic nerves and modifies ovarian follicular development in the rat. Biol Reprod. 2003;68:2038–43.PubMedCrossRef
50.
go back to reference Xu HX, Lin SX, Gong Y, Huo ZX, Zhao CY, Zhu HM, Xi SY. Chaiyu-Dixian formula exerts protective effects on ovarian follicular abnormal development in chronic unpredictable mild stress (CUMS) rat model. Front Pharmacol. 2020;11:245.PubMedPubMedCentralCrossRef Xu HX, Lin SX, Gong Y, Huo ZX, Zhao CY, Zhu HM, Xi SY. Chaiyu-Dixian formula exerts protective effects on ovarian follicular abnormal development in chronic unpredictable mild stress (CUMS) rat model. Front Pharmacol. 2020;11:245.PubMedPubMedCentralCrossRef
51.
go back to reference Divyashree S, Yajurvedi HN. Long-term chronic stress exposure induces PCO phenotype in rat. Reproduction. 2016;152:765–74.PubMedCrossRef Divyashree S, Yajurvedi HN. Long-term chronic stress exposure induces PCO phenotype in rat. Reproduction. 2016;152:765–74.PubMedCrossRef
52.
go back to reference Fu XY, Chen HH, Zhang N, Ding MX, Qiu YE, Pan XM, Fang YS, Lin YP, Zheng Q, Wang WQ. Effects of chronic unpredictable mild stress on ovarian reserve in female rats: Feasibility analysis of a rat model of premature ovarian failure. Mol Med Rep. 2018;18:532–40.PubMed Fu XY, Chen HH, Zhang N, Ding MX, Qiu YE, Pan XM, Fang YS, Lin YP, Zheng Q, Wang WQ. Effects of chronic unpredictable mild stress on ovarian reserve in female rats: Feasibility analysis of a rat model of premature ovarian failure. Mol Med Rep. 2018;18:532–40.PubMed
53.
go back to reference Hobel C, Culhane J. Role of psychosocial and nutritional stress on poor pregnancy outcome. J Nutr. 2003;133:1709S–17S.PubMedCrossRef Hobel C, Culhane J. Role of psychosocial and nutritional stress on poor pregnancy outcome. J Nutr. 2003;133:1709S–17S.PubMedCrossRef
54.
go back to reference Zhao LH, Cui XZ, Yuan HJ, Liang B, Zheng LL, Liu YX, Luo MJ, Tan JH. Restraint stress inhibits mouse implantation: temporal window and the involvement of HB-EGF, estrogen and progesterone. PLoS One. 2013;8:e80472.PubMedPubMedCentralCrossRef Zhao LH, Cui XZ, Yuan HJ, Liang B, Zheng LL, Liu YX, Luo MJ, Tan JH. Restraint stress inhibits mouse implantation: temporal window and the involvement of HB-EGF, estrogen and progesterone. PLoS One. 2013;8:e80472.PubMedPubMedCentralCrossRef
55.
go back to reference deCatanzaro D, Macniven E. Psychogenic pregnancy disruptions in mammals. Neurosci Biobehav Rev. 1992;16:43–53.PubMedCrossRef deCatanzaro D, Macniven E. Psychogenic pregnancy disruptions in mammals. Neurosci Biobehav Rev. 1992;16:43–53.PubMedCrossRef
56.
go back to reference Burkus J, Kacmarova M, Kubandova J, Kokosova N, Fabianova K, Fabian D, Koppel J, Cikos S. Stress exposure during the preimplantation period affects blastocyst lineages and offspring development. J Reprod Dev. 2015;61:325–31.PubMedPubMedCentralCrossRef Burkus J, Kacmarova M, Kubandova J, Kokosova N, Fabianova K, Fabian D, Koppel J, Cikos S. Stress exposure during the preimplantation period affects blastocyst lineages and offspring development. J Reprod Dev. 2015;61:325–31.PubMedPubMedCentralCrossRef
57.
go back to reference Burkus J, Cikos S, Fabian D, Kubandova J, Czikkova S, Koppel J. Maternal restraint stress negatively influences growth capacity of preimplantation mouse embryos. Gen Physiol Biophys. 2013;32:129–37.PubMedCrossRef Burkus J, Cikos S, Fabian D, Kubandova J, Czikkova S, Koppel J. Maternal restraint stress negatively influences growth capacity of preimplantation mouse embryos. Gen Physiol Biophys. 2013;32:129–37.PubMedCrossRef
58.
go back to reference Li XH, Pang HQ, Qin L, Jin S, Zeng X, Bai Y, Li SW. HSP70 overexpression may play a protective role in the mouse embryos stimulated by CUMS. Reprod Biol Endocrinol. 2015;13:125.PubMedPubMedCentralCrossRef Li XH, Pang HQ, Qin L, Jin S, Zeng X, Bai Y, Li SW. HSP70 overexpression may play a protective role in the mouse embryos stimulated by CUMS. Reprod Biol Endocrinol. 2015;13:125.PubMedPubMedCentralCrossRef
59.
go back to reference Zheng LL, Tan XW, Cui XZ, Yuan HJ, Li H, Jiao GZ, Ji CL, Tan JH. Preimplantation maternal stress impairs embryo development by inducing oviductal apoptosis with activation of the Fas system. Mol Hum Reprod. 2016;22:778–90.PubMedCrossRef Zheng LL, Tan XW, Cui XZ, Yuan HJ, Li H, Jiao GZ, Ji CL, Tan JH. Preimplantation maternal stress impairs embryo development by inducing oviductal apoptosis with activation of the Fas system. Mol Hum Reprod. 2016;22:778–90.PubMedCrossRef
60.
go back to reference Camille Melon L, Maguire J. GABAergic regulation of the HPA and HPG axes and the impact of stress on reproductive function. J Steroid Biochem Mol Biol. 2016;160:196–203.PubMedCrossRef Camille Melon L, Maguire J. GABAergic regulation of the HPA and HPG axes and the impact of stress on reproductive function. J Steroid Biochem Mol Biol. 2016;160:196–203.PubMedCrossRef
61.
go back to reference Brunton PJ. Neuroactive steroids and stress axis regulation: pregnancy and beyond. J Steroid Biochem Mol Biol. 2016;160:160–8.PubMedCrossRef Brunton PJ. Neuroactive steroids and stress axis regulation: pregnancy and beyond. J Steroid Biochem Mol Biol. 2016;160:160–8.PubMedCrossRef
62.
go back to reference Maguire J. Neuroactive steroids and GABAergic involvement in the neuroendocrine dysfunction associated with major depressive disorder and postpartum depression. Front Cell Neurosci. 2019;13:83.PubMedPubMedCentralCrossRef Maguire J. Neuroactive steroids and GABAergic involvement in the neuroendocrine dysfunction associated with major depressive disorder and postpartum depression. Front Cell Neurosci. 2019;13:83.PubMedPubMedCentralCrossRef
63.
go back to reference Stephens MA, Wand G. Stress and the HPA axis: role of glucocorticoids in alcohol dependence. Alcohol Res. 2012;34:468–83.PubMedPubMedCentral Stephens MA, Wand G. Stress and the HPA axis: role of glucocorticoids in alcohol dependence. Alcohol Res. 2012;34:468–83.PubMedPubMedCentral
64.
65.
go back to reference Grad I, Picard D. The glucocorticoid responses are shaped by molecular chaperones. Mol Cell Endocrinol. 2007;275:2–12.PubMedCrossRef Grad I, Picard D. The glucocorticoid responses are shaped by molecular chaperones. Mol Cell Endocrinol. 2007;275:2–12.PubMedCrossRef
66.
go back to reference Michael AE, Pester LA, Curtis P, Shaw RW, Edwards CR, Cooke BA. Direct inhibition of ovarian steroidogenesis by cortisol and the modulatory role of 11 beta-hydroxysteroid dehydrogenase. Clin Endocrinol. 1993;38:641–4.CrossRef Michael AE, Pester LA, Curtis P, Shaw RW, Edwards CR, Cooke BA. Direct inhibition of ovarian steroidogenesis by cortisol and the modulatory role of 11 beta-hydroxysteroid dehydrogenase. Clin Endocrinol. 1993;38:641–4.CrossRef
67.
go back to reference Yuan HJ, Han X, He N, Wang GL, Gong S, Lin J, Gao M, Tan JH. Glucocorticoids impair oocyte developmental potential by triggering apoptosis of ovarian cells via activating the Fas system. Sci Rep. 2016;6:24036.PubMedPubMedCentralCrossRef Yuan HJ, Han X, He N, Wang GL, Gong S, Lin J, Gao M, Tan JH. Glucocorticoids impair oocyte developmental potential by triggering apoptosis of ovarian cells via activating the Fas system. Sci Rep. 2016;6:24036.PubMedPubMedCentralCrossRef
68.
go back to reference Namwanje M, Brown CW. Activins and Inhibins: roles in development, physiology, and disease. Cold Spring Harb Perspect Biol. 2016;8. Namwanje M, Brown CW. Activins and Inhibins: roles in development, physiology, and disease. Cold Spring Harb Perspect Biol. 2016;8.
69.
go back to reference Palermo R. Differential actions of FSH and LH during folliculogenesis. Reprod BioMed Online. 2007;15:326–37.PubMedCrossRef Palermo R. Differential actions of FSH and LH during folliculogenesis. Reprod BioMed Online. 2007;15:326–37.PubMedCrossRef
70.
go back to reference Filicori M, Cognigni GE, Samara A, Melappioni S, Perri T, Cantelli B, Parmegiani L, Pelusi G, DeAloysio D. The use of LH activity to drive folliculogenesis: exploring uncharted territories in ovulation induction. Hum Reprod Update. 2002;8:543–57.PubMedCrossRef Filicori M, Cognigni GE, Samara A, Melappioni S, Perri T, Cantelli B, Parmegiani L, Pelusi G, DeAloysio D. The use of LH activity to drive folliculogenesis: exploring uncharted territories in ovulation induction. Hum Reprod Update. 2002;8:543–57.PubMedCrossRef
71.
go back to reference Vegetti W, Alagna F. FSH and folliculogenesis: from physiology to ovarian stimulation. Reprod BioMed Online. 2006;12:684–94.CrossRefPubMed Vegetti W, Alagna F. FSH and folliculogenesis: from physiology to ovarian stimulation. Reprod BioMed Online. 2006;12:684–94.CrossRefPubMed
72.
go back to reference Kalantaridou SN, Makrigiannakis A, Zoumakis E, Chrousos GP. Stress and the female reproductive system. J Reprod Immunol. 2004;62:61–8.PubMedCrossRef Kalantaridou SN, Makrigiannakis A, Zoumakis E, Chrousos GP. Stress and the female reproductive system. J Reprod Immunol. 2004;62:61–8.PubMedCrossRef
73.
go back to reference Chrousos GP. The hypothalamic-pituitary-adrenal axis and immune-mediated inflammation. N Engl J Med. 1995;332:1351–62.PubMedCrossRef Chrousos GP. The hypothalamic-pituitary-adrenal axis and immune-mediated inflammation. N Engl J Med. 1995;332:1351–62.PubMedCrossRef
74.
go back to reference Mastorakos G, Scopa CD, Vryonidou A, Friedman TC, Kattis D, Phenekos C, Merino MJ, Chrousos GP. Presence of immunoreactive corticotropin-releasing hormone in normal and polycystic human ovaries. J Clin Endocrinol Metab. 1994;79:1191–7.PubMed Mastorakos G, Scopa CD, Vryonidou A, Friedman TC, Kattis D, Phenekos C, Merino MJ, Chrousos GP. Presence of immunoreactive corticotropin-releasing hormone in normal and polycystic human ovaries. J Clin Endocrinol Metab. 1994;79:1191–7.PubMed
75.
go back to reference Kiapekou E, Zapanti E, Mastorakos G, Loutradis D. Update on the role of ovarian corticotropin-releasing hormone. Ann N Y Acad Sci. 2010;1205:225–9.PubMedCrossRef Kiapekou E, Zapanti E, Mastorakos G, Loutradis D. Update on the role of ovarian corticotropin-releasing hormone. Ann N Y Acad Sci. 2010;1205:225–9.PubMedCrossRef
76.
go back to reference Liang B, Wei DL, Cheng YN, Yuan HJ, Lin J, Cui XZ, Luo MJ, Tan JH. Restraint stress impairs oocyte developmental potential in mice: role of CRH-induced apoptosis of ovarian cells. Biol Reprod. 2013;89:64.PubMed Liang B, Wei DL, Cheng YN, Yuan HJ, Lin J, Cui XZ, Luo MJ, Tan JH. Restraint stress impairs oocyte developmental potential in mice: role of CRH-induced apoptosis of ovarian cells. Biol Reprod. 2013;89:64.PubMed
77.
79.
go back to reference Shkolnik K, Tadmor A, Ben-Dor S, Nevo N, Galiani D, Dekel N. Reactive oxygen species are indispensable in ovulation. Proc Natl Acad Sci U S A. 2011;108:1462–7.PubMedPubMedCentralCrossRef Shkolnik K, Tadmor A, Ben-Dor S, Nevo N, Galiani D, Dekel N. Reactive oxygen species are indispensable in ovulation. Proc Natl Acad Sci U S A. 2011;108:1462–7.PubMedPubMedCentralCrossRef
80.
go back to reference Agarwal A, Aponte-Mellado A, Premkumar BJ, Shaman A, Gupta S. The effects of oxidative stress on female reproduction: a review. Reprod Biol Endocrinol. 2012;10:49.PubMedPubMedCentralCrossRef Agarwal A, Aponte-Mellado A, Premkumar BJ, Shaman A, Gupta S. The effects of oxidative stress on female reproduction: a review. Reprod Biol Endocrinol. 2012;10:49.PubMedPubMedCentralCrossRef
81.
go back to reference Guerin P, El Mouatassim S, Menezo Y. Oxidative stress and protection against reactive oxygen species in the pre-implantation embryo and its surroundings. Hum Reprod Update. 2001;7:175–89.PubMedCrossRef Guerin P, El Mouatassim S, Menezo Y. Oxidative stress and protection against reactive oxygen species in the pre-implantation embryo and its surroundings. Hum Reprod Update. 2001;7:175–89.PubMedCrossRef
82.
go back to reference Ruder EH, Hartman TJ, Blumberg J, Goldman MB. Oxidative stress and antioxidants: exposure and impact on female fertility. Hum Reprod Update. 2008;14:345–57.PubMedCrossRef Ruder EH, Hartman TJ, Blumberg J, Goldman MB. Oxidative stress and antioxidants: exposure and impact on female fertility. Hum Reprod Update. 2008;14:345–57.PubMedCrossRef
83.
go back to reference Chaudhary GR, Yadav PK, Yadav AK, Tiwari M, Gupta A, Sharma A, Pandey AN, Pandey AK, Chaube SK. Necroptosis in stressed ovary. J Biomed Sci. 2019;26:11.PubMedPubMedCentralCrossRef Chaudhary GR, Yadav PK, Yadav AK, Tiwari M, Gupta A, Sharma A, Pandey AN, Pandey AK, Chaube SK. Necroptosis in stressed ovary. J Biomed Sci. 2019;26:11.PubMedPubMedCentralCrossRef
84.
go back to reference Nakayama M, Manabe N, Inoue N, Matsui T, Miyamoto H. Changes in the expression of tumor necrosis factor (TNF) alpha, TNFalpha receptor (TNFR) 2, and TNFR-associated factor 2 in granulosa cells during atresia in pig ovaries. Biol Reprod. 2003;68:530–5.PubMedCrossRef Nakayama M, Manabe N, Inoue N, Matsui T, Miyamoto H. Changes in the expression of tumor necrosis factor (TNF) alpha, TNFalpha receptor (TNFR) 2, and TNFR-associated factor 2 in granulosa cells during atresia in pig ovaries. Biol Reprod. 2003;68:530–5.PubMedCrossRef
86.
go back to reference Wu W, Liu P, Li J. Necroptosis: an emerging form of programmed cell death. Crit Rev Oncol Hematol. 2012;82:249–58.PubMedCrossRef Wu W, Liu P, Li J. Necroptosis: an emerging form of programmed cell death. Crit Rev Oncol Hematol. 2012;82:249–58.PubMedCrossRef
87.
go back to reference Aitken RJ. Impact of oxidative stress on male and female germ cells: implications for fertility. Reproduction. 2020;159:R189–201.PubMedCrossRef Aitken RJ. Impact of oxidative stress on male and female germ cells: implications for fertility. Reproduction. 2020;159:R189–201.PubMedCrossRef
88.
go back to reference Tatone C, Di Emidio G, Vitti M, Di Carlo M, Santini S Jr, D'Alessandro AM, Falone S, Amicarelli F. Sirtuin functions in female fertility: possible role in oxidative stress and aging. Oxidative Med Cell Longev. 2015;2015:659687.CrossRef Tatone C, Di Emidio G, Vitti M, Di Carlo M, Santini S Jr, D'Alessandro AM, Falone S, Amicarelli F. Sirtuin functions in female fertility: possible role in oxidative stress and aging. Oxidative Med Cell Longev. 2015;2015:659687.CrossRef
89.
go back to reference Ishii T, Miyazawa M, Takanashi Y, Tanigawa M, Yasuda K, Onouchi H, Kawabe N, Mitsushita J, Hartman PS, Ishii N. Genetically induced oxidative stress in mice causes thrombocytosis, splenomegaly and placental angiodysplasia that leads to recurrent abortion. Redox Biol. 2014;2:679–85.PubMedPubMedCentralCrossRef Ishii T, Miyazawa M, Takanashi Y, Tanigawa M, Yasuda K, Onouchi H, Kawabe N, Mitsushita J, Hartman PS, Ishii N. Genetically induced oxidative stress in mice causes thrombocytosis, splenomegaly and placental angiodysplasia that leads to recurrent abortion. Redox Biol. 2014;2:679–85.PubMedPubMedCentralCrossRef
90.
go back to reference Wyck S, Herrera C, Requena CE, Bittner L, Hajkova P, Bollwein H, Santoro R. Oxidative stress in sperm affects the epigenetic reprogramming in early embryonic development. Epigenetics Chromatin. 2018;11:60.PubMedPubMedCentralCrossRef Wyck S, Herrera C, Requena CE, Bittner L, Hajkova P, Bollwein H, Santoro R. Oxidative stress in sperm affects the epigenetic reprogramming in early embryonic development. Epigenetics Chromatin. 2018;11:60.PubMedPubMedCentralCrossRef
91.
go back to reference Sun YC, Wang YY, Ge W, Cheng SF, Dyce PW, Shen W. Epigenetic regulation during the differentiation of stem cells to germ cells. Oncotarget. 2017;8:57836–44.PubMedPubMedCentralCrossRef Sun YC, Wang YY, Ge W, Cheng SF, Dyce PW, Shen W. Epigenetic regulation during the differentiation of stem cells to germ cells. Oncotarget. 2017;8:57836–44.PubMedPubMedCentralCrossRef
92.
go back to reference Saitou M, Kagiwada S, Kurimoto K. Epigenetic reprogramming in mouse pre-implantation development and primordial germ cells. Development. 2012;139:15–31.PubMedCrossRef Saitou M, Kagiwada S, Kurimoto K. Epigenetic reprogramming in mouse pre-implantation development and primordial germ cells. Development. 2012;139:15–31.PubMedCrossRef
93.
go back to reference Darnaudery M, Maccari S. Epigenetic programming of the stress response in male and female rats by prenatal restraint stress. Brain Res Rev. 2008;57:571–85.PubMedCrossRef Darnaudery M, Maccari S. Epigenetic programming of the stress response in male and female rats by prenatal restraint stress. Brain Res Rev. 2008;57:571–85.PubMedCrossRef
94.
95.
go back to reference Dreher ML. Dietary fiber in health and disease: Springer; 2018. Dreher ML. Dietary fiber in health and disease: Springer; 2018.
96.
go back to reference Bohacek J, Mansuy IM. Molecular insights into transgenerational non-genetic inheritance of acquired behaviours. Nat Rev Genet. 2015;16:641–52.PubMedCrossRef Bohacek J, Mansuy IM. Molecular insights into transgenerational non-genetic inheritance of acquired behaviours. Nat Rev Genet. 2015;16:641–52.PubMedCrossRef
97.
go back to reference Klengel T, Dias BG, Ressler KJ. Models of intergenerational and Transgenerational transmission of risk for psychopathology in mice. Neuropsychopharmacology. 2016;41:219–31.PubMedCrossRef Klengel T, Dias BG, Ressler KJ. Models of intergenerational and Transgenerational transmission of risk for psychopathology in mice. Neuropsychopharmacology. 2016;41:219–31.PubMedCrossRef
98.
go back to reference Evans NP, Bellingham M, Robinson JE. Prenatal programming of neuroendocrine reproductive function. Theriogenology. 2016;86:340–8.PubMedCrossRef Evans NP, Bellingham M, Robinson JE. Prenatal programming of neuroendocrine reproductive function. Theriogenology. 2016;86:340–8.PubMedCrossRef
99.
go back to reference Argyraki M, Damdimopoulou P, Chatzimeletiou K, Grimbizis GF, Tarlatzis BC, Syrrou M, Lambropoulos A. In-utero stress and mode of conception: impact on regulation of imprinted genes, fetal development and future health. Hum Reprod Update. 2019;25:777–801.PubMedCrossRef Argyraki M, Damdimopoulou P, Chatzimeletiou K, Grimbizis GF, Tarlatzis BC, Syrrou M, Lambropoulos A. In-utero stress and mode of conception: impact on regulation of imprinted genes, fetal development and future health. Hum Reprod Update. 2019;25:777–801.PubMedCrossRef
Metadata
Title
Review of psychological stress on oocyte and early embryonic development in female mice
Authors
Qiu-Yue Zhai
Jun-Jie Wang
Yu Tian
Xiaofang Liu
Zhenhua Song
Publication date
01-12-2020
Publisher
BioMed Central
Keyword
Epigenetics
Published in
Reproductive Biology and Endocrinology / Issue 1/2020
Electronic ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-020-00657-1

Other articles of this Issue 1/2020

Reproductive Biology and Endocrinology 1/2020 Go to the issue