Impact of psychological stress on ovarian function: Insights, mechanisms and intervention strategies (Review)
- Authors:
- Yu Hu
- Wuyang Wang
- Wenqing Ma
- Wenwen Wang
- Wu Ren
- Shixuan Wang
- Fangfang Fu
- Yan Li
-
Affiliations: Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China - Published online on: December 18, 2024 https://doi.org/10.3892/ijmm.2024.5475
- Article Number: 34
-
Copyright: © Hu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
This article is mentioned in:
Abstract
Wang S, Quan L, Chavarro JE, Slopen N, Kubzansky LD, Koenen KC, Kang JH, Weisskopf MG, Branch-Elliman W and Roberts AL: Associations of depression, anxiety, worry, perceived stress, and loneliness prior to infection with risk of post-COVID-19 conditions. JAMA Psychiatry. 79:1081–1091. 2022. View Article : Google Scholar : PubMed/NCBI | |
Valsamakis G, Chrousos G and Mastorakos G: Stress, female reproduction and pregnancy. Psychoneuroendocrinology. 100:48–57. 2019. View Article : Google Scholar | |
Kivimäki M, Bartolomucci A and Kawachi I: The multiple roles of life stress in metabolic disorders. Nat Rev Endocrinol. 19:10–27. 2023. View Article : Google Scholar | |
Bala R, Singh V, Rajender S and Singh K: Environment, lifestyle, and female infertility. Reprod Sci. 28:617–638. 2021. View Article : Google Scholar | |
Rooney KL and Domar AD: The relationship between stress and infertility. Dialogues Clin Neurosci. 20:41–47. 2018. View Article : Google Scholar : PubMed/NCBI | |
Maeda E, Nomura K, Hiraike O, Sugimori H, Kinoshita A and Osuga Y: Domestic work stress and self-rated psychological health among women: A cross-sectional study in Japan. Environ Health Prev Med. 24:752019. View Article : Google Scholar : PubMed/NCBI | |
Liu Y and Aungsuroch Y: Work stress, perceived social support, self-efficacy and burnout among Chinese registered nurses. J Nurs Manag. 27:1445–1453. 2019. View Article : Google Scholar : PubMed/NCBI | |
Bapayeva G, Aimagambetova G, Issanov A, Terzic S, Ukybassova T, Aldiyarova A, Utepova G, Daribay Z, Bekbossinova G, Balykov A, et al: The effect of stress, anxiety and depression on in vitro fertilization outcome in kazakhstani public clinical setting: A cross-sectional study. J Clin Med. 10:9372021. View Article : Google Scholar : PubMed/NCBI | |
Xu H, Ouyang N, Li R, Tuo P, Mai M and Wang W: The effects of anxiety and depression on in vitro fertilisation outcomes of infertile Chinese women. Psychol Health Med. 22:37–43. 2017. View Article : Google Scholar | |
Sallem A, Essoussi H, Mustapha HB, Zaouali M and Ajina M: Impact of psychological stress on the outcomes of assisted reproduction in Tunisian infertile women. Pan Afr Med J. 40:2502021. View Article : Google Scholar | |
Klonoff-Cohen H, Chu E, Natarajan L and Sieber W: A prospective study of stress among women undergoing in vitro fertilization or gamete intrafallopian transfer. Fertil Steril. 76:675–687. 2001. View Article : Google Scholar : PubMed/NCBI | |
Buck Louis GM, Lum KJ, Sundaram R, Chen Z, Kim S, Lynch CD, Schisterman EF and Pyper C: Stress reduces conception probabilities across the fertile window: Evidence in support of relaxation. Fertil Steril. 95:2184–2189. 2011. View Article : Google Scholar | |
Mínguez-Alarcón L, Williams PL, Souter I, Ford JB, Hauser R and Chavarro JE; Earth Study Team: Perceived stress and markers of ovarian reserve among subfertile women. Reprod Biomed Online. 46:956–964. 2023. View Article : Google Scholar : PubMed/NCBI | |
Golenbock SW, Wise LA, Lambert-Messerlian GM, Eklund EE and Harlow BL: Association between a history of depression and anti-müllerian hormone among late-reproductive aged women: The Harvard study of moods and cycles. Womens Midlife Health. 6:92020. View Article : Google Scholar | |
Allshouse AA, Semple AL and Santoro NF: Evidence for prolonged and unique amenorrhea-related symptoms in women with premature ovarian failure/primary ovarian insufficiency. Menopause. 22:166–174. 2015. View Article : Google Scholar | |
Lawson EA, Donoho D, Miller KK, Misra M, Meenaghan E, Lydecker J, Wexler T, Herzog DB and Klibanski A: Hypercortisolemia is associated with severity of bone loss and depression in hypothalamic amenorrhea and anorexia nervosa. J Clin Endocrinol Metab. 94:4710–4716. 2009. View Article : Google Scholar : PubMed/NCBI | |
Goodarzi MO, Dumesic DA, Chazenbalk G and Azziz R: Polycystic ovary syndrome: Etiology, pathogenesis and diagnosis. Nat Rev Endocrinol. 7:219–231. 2011. View Article : Google Scholar : PubMed/NCBI | |
Freeman EW, Sammel MD, Lin H and Nelson DB: Associations of hormones and menopausal status with depressed mood in women with no history of depression. Arch Gen Psychiatry. 63:375–382. 2006. View Article : Google Scholar : PubMed/NCBI | |
Gao L, Zhao F, Zhang Y, Wang W and Cao Q: Diminished ovarian reserve induced by chronic unpredictable stress in C57BL/6 mice. Gynecol Endocrinol. 36:49–54. 2020. View Article : Google Scholar | |
Jiang Y, Xu J, Tao C, Lin Y, Lin X, Li K, Liu Q, Saiyin H, Hu S, Yao G, et al: Chronic stress induces meiotic arrest failure and ovarian reserve decline via the cAMP signaling pathway. Front Endocrinol (Lausanne). 14:11770612023. View Article : Google Scholar : PubMed/NCBI | |
Xu M, Sun J, Wang Q, Zhang Q, Wei C and Lai D: Chronic restraint stress induces excessive activation of primordial follicles in mice ovaries. PLoS One. 13:e01948942018. View Article : Google Scholar : PubMed/NCBI | |
Poole L and Hackett RA: Diabetes distress: The psychological burden of living with diabetes. Lancet Diabetes Endocrinol. 12:439–441. 2024. View Article : Google Scholar : PubMed/NCBI | |
Bartoskova Polcrova A, Dalecka A, Szabo D, Gonzalez Rivas JP, Bobak M and Pikhart H: Social and environmental stressors of cardiometabolic health. Sci Rep. 14:141792024. View Article : Google Scholar : PubMed/NCBI | |
Schliep KC, Mumford SL, Vladutiu CJ, Ahrens KA, Perkins NJ, Sjaarda LA, Kissell KA, Prasad A, Wactawski-Wende J and Schisterman EF: Perceived stress, reproductive hormones, and ovulatory function: A prospective cohort study. Epidemiology. 26:177–184. 2015. View Article : Google Scholar : PubMed/NCBI | |
Harlow BL, Wise LA, Otto MW, Soares CN and Cohen LS: Depression and its influence on reproductive endocrine and menstrual cycle markers associated with perimenopause: The Harvard study of moods and cycles. Arch Gen Psychiatry. 60:29–36. 2003. View Article : Google Scholar : PubMed/NCBI | |
Albert KM and Newhouse PA: Estrogen, stress, and depression: cognitive and biological interactions. Annu Rev Clin Psychol. 15:399–423. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yatsenko SA and Rajkovic A: Genetics of human female infertility. Biol Reprod. 101:549–566. 2019. View Article : Google Scholar : PubMed/NCBI | |
Matthiesen SMS, Frederiksen Y, Ingerslev HJ and Zachariae R: Stress, distress and outcome of assisted reproductive technology (ART): A meta-analysis. Hum Reprod. 26:2763–2776. 2011. View Article : Google Scholar : PubMed/NCBI | |
Massey AJ, Campbell B, Raine-Fenning N, Aujla N and Vedhara K: The association of physiological cortisol and IVF treatment outcomes: A systematic review. Reprod Med Biol. 13:161–176. 2014. View Article : Google Scholar : PubMed/NCBI | |
Chen TH, Chang SP, Tsai CF and Juang KD: Prevalence of depressive and anxiety disorders in an assisted reproductive technique clinic. Hum Reprod. 19:2313–2318. 2004. View Article : Google Scholar : PubMed/NCBI | |
Volgsten H, Skoog Svanberg A, Ekselius L, Lundkvist O and Sundström Poromaa I: Prevalence of psychiatric disorders in infertile women and men undergoing in vitro fertilization treatment. Hum Reprod. 23:2056–2063. 2008. View Article : Google Scholar : PubMed/NCBI | |
Pasch LA, Holley SR, Bleil ME, Shehab D, Katz PP and Adler NE: Addressing the needs of fertility treatment patients and their partners: Are they informed of and do they receive mental health services? Fertil Steril. 106:209–215.e2. 2016. View Article : Google Scholar : PubMed/NCBI | |
Lakatos E, Szigeti JF, Ujma PP, Sexty R and Balog P: Anxiety and depression among infertile women: A cross-sectional survey from Hungary. BMC Womens Health. 17:482017. View Article : Google Scholar : PubMed/NCBI | |
An Y, Sun Z, Li L, Zhang Y and Ji H: Relationship between psychological stress and reproductive outcome in women undergoing in vitro fertilization treatment: Psychological and neurohormonal assessment. J Assist Reprod Genet. 30:35–41. 2013. View Article : Google Scholar : | |
Terzioglu F, Turk R, Yucel C, Dilbaz S, Cinar O and Karahalil B: The effect of anxiety and depression scores of couples who underwent assisted reproductive techniques on the pregnancy outcomes. Afr Health Sci. 16:441–450. 2016. View Article : Google Scholar : PubMed/NCBI | |
Aimagambetova G, Issanov A, Terzic S, Bapayeva G, Ukybassova T, Baikoshkarova S, Aldiyarova A, Shauyen F and Terzic M: The effect of psychological distress on IVF outcomes: Reality or speculations? PLoS One. 15:e02420242020. View Article : Google Scholar : PubMed/NCBI | |
Turner K, Reynolds-May MF, Zitek EM, Tisdale RL, Carlisle AB and Westphal LM: Stress and anxiety scores in first and repeat IVF cycles: A pilot study. PLoS One. 8:e637432013. View Article : Google Scholar : PubMed/NCBI | |
Quant HS, Zapantis A, Nihsen M, Bevilacqua K, Jindal S and Pal L: Reproductive implications of psychological distress for couples undergoing IVF. J Assist Reprod Genet. 30:1451–1458. 2013. View Article : Google Scholar : PubMed/NCBI | |
Csemiczky G, Landgren BM and Collins A: The influence of stress and state anxiety on the outcome of IVF-treatment: Psychological and endocrinological assessment of Swedish women entering IVF-treatment: Stress and state anxiety on outcome of IVF-treatment. Acta Obstet Gynecol Scand. 79:113–118. 2000. View Article : Google Scholar : PubMed/NCBI | |
Boivin J, Griffiths E and Venetis CA: Emotional distress in infertile women and failure of assisted reproductive technologies: Meta-analysis of prospective psychosocial studies. BMJ. 342:d2232011. View Article : Google Scholar : PubMed/NCBI | |
Pasch LA, Gregorich SE, Katz PK, Millstein SG, Nachtigall RD, Bleil ME and Adler NE: Psychological distress and in vitro fertilization outcome. Fertil Steril. 98:459–464. 2012. View Article : Google Scholar : PubMed/NCBI | |
Chai Y, Li Q, Wang Y, Niu B, Chen H, Fan T, Ke X and Zou H: Cortisol dysregulation in anxiety infertile women and the influence on IVF treatment outcome. Front Endocrinol (Lausanne). 14:11077652023. View Article : Google Scholar : PubMed/NCBI | |
Cesta CE, Johansson ALV, Hreinsson J, Rodriguez-Wallberg KA, Olofsson JI, Holte J, Wramsby H, Wramsby M, Cnattingius S, Skalkidou A and Nyman Iliadou A: A prospective investigation of perceived stress infertility-related stress and cortisol levels in women undergoing in vitro fertilization: Influence on embryo quality and clinical pregnancy rate. Acta Obstet Gynecol Scand. 97:258–268. 2018. View Article : Google Scholar | |
Mirzaasgari H, Momeni F, Pourshahbaz A, Keshavarzi F and Hatami M: The relationship between coping strategies and infertility self-efficacy with pregnancy outcomes of women undergoing in vitro fertilization: A prospective cohort study. Int J Reprod Biomed. 20:539–548. 2022.PubMed/NCBI | |
Peng M, Wen M, Jiang T, Jiang Y, Lv H, Chen T, Ling X, Li H, Meng Q, Huang B, et al: Stress, anxiety, and depression in infertile couples are not associated with a first IVF or ICSI treatment outcome. BMC Pregnancy Childbirth. 21:7252021. View Article : Google Scholar : PubMed/NCBI | |
Park J, Stanford JB, Porucznik CA, Christensen K and Schliep KC: Daily perceived stress and time to pregnancy: A prospective cohort study of women trying to conceive. Psychoneuroendocrinology. 110:1044462019. View Article : Google Scholar : PubMed/NCBI | |
Lynch CD, Sundaram R, Buck Louis GM, Lum KJ and Pyper C: Are increased levels of self-reported psychosocial stress, anxiety, and depression associated with fecundity? Fertil Steril. 98:453–458. 2012. View Article : Google Scholar : PubMed/NCBI | |
Anderheim L, Holter H, Bergh C and Möller A: Does psychological stress affect the outcome of in vitro fertilization? Hum Reprod. 20:2969–2975. 2005. View Article : Google Scholar : PubMed/NCBI | |
Milad MP, Klock SC, Moses S and Chatterton R: Stress and anxiety do not result in pregnancy wastage. Hum Reprod. 13:2296–2300. 1998. View Article : Google Scholar : PubMed/NCBI | |
Butts CD, Bloom MS, Frye CA, Walf AA, Parsons PJ, Steuerwald AJ, Ilonze C and Fujimoto VY: Urine cortisol concentration as a biomarker of stress is unrelated to IVF outcomes in women and men. J Assist Reprod Genet. 31:1647–1653. 2014. View Article : Google Scholar : PubMed/NCBI | |
Stuenkel CA and Gompel A: Primary ovarian insufficiency. N Engl J Med. 388:154–163. 2023. View Article : Google Scholar : PubMed/NCBI | |
Sun J, Fan Y, Guo Y, Pan H, Zhang C, Mao G, Huang Y, Li B, Gu T, Wang L, et al: Chronic and cumulative adverse life events in women with primary ovarian insufficiency: An exploratory qualitative study. Front Endocrinol (Lausanne). 13:8560442022. View Article : Google Scholar : PubMed/NCBI | |
Xu YP, Fu JC, Hong ZL, Zeng DF, Guo CQ, Li P and Wu JX: Psychological stressors involved in the pathogenesis of premature ovarian insufficiency and potential intervention measures. Gynecol Endocrinol. 40:23600852024. View Article : Google Scholar : PubMed/NCBI | |
Schmidt PJ, Luff JA, Haq NA, Vanderhoof VH, Koziol DE, Calis KA, Rubinow DR and Nelson LM: Depression in women with spontaneous 46, XX primary ovarian insufficiency. J Clin Endocrinol Metab. 96:E278–E287. 2011. View Article : Google Scholar : | |
Freeman EW, Sammel MD, Liu L, Gracia CR, Nelson DB and Hollander L: Hormones and menopausal status as predictors of depression in womenin transition to menopause. Arch Gen Psychiatry. 61:62–70. 2004. View Article : Google Scholar : PubMed/NCBI | |
Bleil ME, Adler NE, Pasch LA, Sternfeld B, Gregorich SE, Rosen MP and Cedars MI: Psychological stress and reproductive aging among pre-menopausal women. Hum Reprod. 27:2720–2728. 2012. View Article : Google Scholar : PubMed/NCBI | |
Bleil ME, Adler NE, Pasch LA, Sternfeld B, Gregorich SE, Rosen MP and Cedars MI: Depressive symptomatology, psychological stress, and ovarian reserve: A role for psychological factors in ovarian aging? Menopause. 19:1176–1185. 2012. View Article : Google Scholar : PubMed/NCBI | |
Dong YZ, Zhou FJ and Sun YP: Psychological stress is related to a decrease of serum anti-müllerian hormone level in infertile women. Reprod Biol Endocrinol. 15:512017. View Article : Google Scholar | |
Lania A, Gianotti L, Gagliardi I, Bondanelli M, Vena W and Ambrosio MR: Functional hypothalamic and drug-induced amenorrhea: An overview. J Endocrinol Invest. 42:1001–1010. 2019. View Article : Google Scholar : PubMed/NCBI | |
Gordon CM, Ackerman KE, Berga SL, Kaplan JR, Mastorakos G, Misra M, Murad MH, Santoro NF and Warren MP: Functional hypothalamic amenorrhea: An endocrine society clinical practice guideline. J Clin Endocrinol Metab. 102:1413–1439. 2017. View Article : Google Scholar : PubMed/NCBI | |
Bomba M, Gambera A, Bonini L, Peroni M, Neri F, Scagliola P and Nacinovich R: Endocrine profiles and neuropsychologic correlates of functional hypothalamic amenorrhea in adolescents. Fertil Steril. 87:876–885. 2007. View Article : Google Scholar : PubMed/NCBI | |
Marcus MD, Loucks TL and Berga SL: Psychological correlates of functional hypothalamic amenorrhea. Fertil Steril. 76:310–316. 2001. View Article : Google Scholar : PubMed/NCBI | |
Dundon CM, Rellini AH, Tonani S, Santamaria V and Nappi R: Mood disorders and sexual functioning in women with functional hypothalamic amenorrhea. Fertil Steril. 94:2239–2243. 2010. View Article : Google Scholar : PubMed/NCBI | |
Cai J, Zhang Y, Wang Y, Li S, Wang L, Zheng J, Jiang Y, Dong Y, Zhou H, Hu Y, et al: High thyroid stimulating hormone level is associated with hyperandrogenism in euthyroid polycystic ovary syndrome (PCOS) women, independent of age, BMI, and thyroid autoimmunity: A cross-sectional analysis. Front Endocrinol (Lausanne). 10:2222019. View Article : Google Scholar : PubMed/NCBI | |
Chang AY, Lalia AZ, Jenkins GD, Dutta T, Carter RE, Singh RJ and Nair KS: Combining a nontargeted and targeted metabolomics approach to identify metabolic pathways significantly altered in polycystic ovary syndrome. Metabolism. 71:52–63. 2017. View Article : Google Scholar : PubMed/NCBI | |
Farrell K and Antoni MH: Insulin resistance, obesity, inflammation, and depression in polycystic ovary syndrome: Biobehavioral mechanisms and interventions. Fertil Steril. 94:1565–1574. 2010. View Article : Google Scholar : PubMed/NCBI | |
Hillman SC, Bryce C, Caleyachetty R and Dale J: Women's experiences of diagnosis and management of polycystic ovary syndrome: A mixed-methods study in general practice. Br J Gen Pract. 70:e322–e329. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chrousos GP: Stress and disorders of the stress system. Nat Rev Endocrinol. 5:374–381. 2009. View Article : Google Scholar : PubMed/NCBI | |
Kleinert M, Clemmensen C, Hofmann SM, Moore MC, Renner S, Woods SC, Huypens P, Beckers J, de Angelis MH, Schürmann A, et al: Animal models of obesity and diabetes mellitus. Nat Rev Endocrinol. 14:140–162. 2018. View Article : Google Scholar : PubMed/NCBI | |
Seemann S, Zohles F and Lupp A: Comprehensive comparison of three different animal models for systemic inflammation. J Biomed Sci. 24:602017. View Article : Google Scholar : PubMed/NCBI | |
Cheon DJ and Orsulic S: Mouse models of cancer. Annu Rev Pathol. 6:95–119. 2011. View Article : Google Scholar | |
Antoniuk S, Bijata M, Ponimaskin E and Wlodarczyk J: Chronic unpredictable mild stress for modeling depression in rodents: Meta-analysis of model reliability. Neurosci Biobehav Rev. 99:101–116. 2019. View Article : Google Scholar | |
Du Preez A, Onorato D, Eiben I, Musaelyan K, Egeland M, Zunszain PA, Fernandes C, Thuret S and Pariante CM: Chronic stress followed by social isolation promotes depressive-like behaviour, alters microglial and astrocyte biology and reduces hippocampal neurogenesis in male mice. Brain Behav Immun. 91:24–47. 2021. View Article : Google Scholar | |
Khan AR, Geiger L, Wiborg O and Czéh B: Stress-induced morphological, cellular and molecular changes in the brain-lessons learned from the chronic mild stress model of depression. Cells. 9:10262020. View Article : Google Scholar : PubMed/NCBI | |
Xiang Y, Jiang L, Gou J, Sun Y, Zhang D, Xin X, Song Z and Huang J: Chronic unpredictable mild stress-induced mouse ovarian insufficiency by interrupting lipid homeostasis in the ovary. Front Cell Dev Biol. 10:9336742022. View Article : Google Scholar : PubMed/NCBI | |
Fu XY, Chen HH, Zhang N, Ding MX, Qiu YE, Pan XM, Fang YS, Lin YP, Zheng Q and 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. 18:532–540. 2018.PubMed/NCBI | |
Ma J, Wang L, Yang D, Luo J, Gao J, Wang J, Guo H, Li J, Wang F, Wu J and Hu R: Chronic stress causes ovarian fibrosis to impair female fertility in mice. Cell Signal. 122:1113342024. View Article : Google Scholar : PubMed/NCBI | |
Ding SM, Shi LG, Xing F, Cui SS, Cheng HR, Liu Y, Ji DM, Liang D, Cao YX and Liu YJ: Melatonin protects against mitochondrial dyshomeostasis and ovarian damage caused by chronic unpredictable mild stress through the eIF2α-AFT4 signaling pathway in mice. Reprod Sci. 31:3191–3201. 2024. View Article : Google Scholar : PubMed/NCBI | |
Fu X, Zheng Q, Zhang N, Ding M, Pan X, Wang W and Chen H: CUMS promotes the development of premature ovarian insufficiency mediated by nerve growth factor and its receptor in rats. BioMed Res Int. 2020:19468532020. View Article : Google Scholar : PubMed/NCBI | |
Xu HX, Lin SX, Gong Y, Huo ZX, Zhao CY, Zhu HM and Xi SY: Chaiyu-dixian formula exerts protective effects on ovarian follicular abnormal development in chronic unpredictable mild stress (CUMS) rat model. Front Pharmacol. 11:2452020. View Article : Google Scholar : PubMed/NCBI | |
Zhao X, Ma R, Zhang X, Cheng R, Jiang N, Guo M, Rong B, Liu Y, Chen M, Feng W and Xia T: Reduced growth capacity of preimplantation mouse embryos in chronic unpredictable stress model. Mol Reprod Dev. 88:80–95. 2021. View Article : Google Scholar | |
Wu LM, Hu MH, Tong XH, Han H, Shen N, Jin RT, Wang W, Zhou GX, He GP and Liu YS: Chronic unpredictable stress decreases expression of brain-derived neurotrophic factor (BDNF) in mouse ovaries: relationship to oocytes developmental potential. PLoS One. 7:e523312012. View Article : Google Scholar | |
Li XH, Pang HQ, Qin L, Jin S, Zeng X, Bai Y and Li SW: HSP70 overexpression may play a protective role in the mouse embryos stimulated by CUMS. Reprod Biol Endocrinol. 13:1252015. View Article : Google Scholar : PubMed/NCBI | |
Wu LM, Liu YS, Tong XH, Shen N, Jin RT, Han H, Hu MH, Wang W and 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. 86:1212012. View Article : Google Scholar | |
Sun J, Guo Y, Fan Y, Wang Q, Zhang Q and Lai D: Decreased expression of IDH1 by chronic unpredictable stress suppresses proliferation and accelerates senescence of granulosa cells through ROS activated MAPK signaling pathways. Free Radic Biol Med. 169:122–136. 2021. View Article : Google Scholar : PubMed/NCBI | |
Du Y, Ma J, Wu T, Li F, Pan J, Du L, Zhang M, Diao X and Wu R: Downgrading breast imaging reporting and data system categories in ultrasound using strain elastography and computer-aided diagnosis system: A multicenter, prospective study. Br J Radiol. 97:1653–1660. 2024. View Article : Google Scholar : PubMed/NCBI | |
Kala M and Nivsarkar M: Role of cortisol and superoxide dismutase in psychological stress induced anovulation. Gen Comp Endocrinol. 225:117–124. 2016. View Article : Google Scholar | |
Gao Y, Chen F, Kong QQ, Ning SF, Yuan HJ, Lian HY, Luo MJ and 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. 23:1148–1157. 2016. View Article : Google Scholar : PubMed/NCBI | |
Scott K, Phan TT, Boukelmoune N, Heijnen CJ and Dantzer R: Chronic restraint stress impairs voluntary wheel running but has no effect on food-motivated behavior in mice. Brain Behav Immun. 107:319–329. 2023. View Article : Google Scholar | |
Chen H, Fu S, Li X, Shi M, Qian J, Zhao S, Yuan P, Ding L, Xia X and Zheng JC: Microglial glutaminase 1 mediates chronic restraint stress-induced depression-like behaviors and synaptic damages. Signal Transduct Target Ther. 8:4522023. View Article : Google Scholar : PubMed/NCBI | |
Chiba S, Numakawa T, Ninomiya M, Richards MC, Wakabayashi C and Kunugi H: Chronic restraint stress causes anxiety- and depression-like behaviors, downregulates glucocorticoid receptor expression, and attenuates glutamate release induced by brain-derived neurotrophic factor in the prefrontal cortex. Prog Neuropsychopharmacol Biol Psychiatry. 39:112–119. 2012. View Article : Google Scholar : PubMed/NCBI | |
Buynitsky T and Mostofsky DI: Restraint stress in biobehavioral research: Recent developments. Neurosci Biobehav Rev. 33:1089–1098. 2009. View Article : Google Scholar : PubMed/NCBI | |
Sun J, Guo Y, Zhang Q, Bu S, Li B, Wang Q and Lai D: Chronic restraint stress disturbs meiotic resumption through APC/C-mediated cyclin B1 excessive degradation in mouse oocytes. Cell Cycle. 17:1591–1601. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhang SY, Wang JZ, Li JJ, Wei DL, Sui HS, Zhang ZH, Zhou P and Tan JH: Maternal restraint stress diminishes the developmental potential of oocytes. Biol Reprod. 84:672–681. 2011. View Article : Google Scholar | |
Geraghty AC, Muroy SE, Zhao S, Bentley GE, Kriegsfeld LJ and Kaufer D: Knockdown of hypothalamic RFRP3 prevents chronic stress-induced infertility and embryo resorption. Life. 4:e043162015. | |
Liang B, Wei DL, Cheng YN, Yuan HJ, Lin J, Cui XZ, Luo MJ and Tan JH: Restraint stress impairs oocyte developmental potential in mice: Role of CRH-induced apoptosis of ovarian cells. Biol Reprod. 89:642013. View Article : Google Scholar : PubMed/NCBI | |
Li CY, Li ZB, Kong QQ, Han X, Xiao B, Li X, Chang ZL and Tan JH: Restraint-induced corticotrophin-releasing hormone elevation triggers apoptosis of ovarian cells and impairs oocyte competence via activation of the Fas/FasL system. Biol Reprod. 99:828–837. 2018. View Article : Google Scholar : PubMed/NCBI | |
Huang Y, Liu Q, Huang G, Wen J and Chen G: Hypothalamic kisspeptin neurons regulates energy metabolism and reproduction under chronic stress. Front Endocrinol (Lausanne). 13:8443972022. View Article : Google Scholar : PubMed/NCBI | |
Sugino N, Nakamura Y, Okuno N, Shimamura K, Teyama T, Ishimatsu M and Kato H: Effects of restraint stress on luteal function in rats during mid-pregnancy. J Reprod Fertil. 101:23–26. 1994. View Article : Google Scholar : PubMed/NCBI | |
Zhou P, Lian HY, Cui W, Wei DL, Li Q, Liu YX, Liu XY and Tan JH: Maternal-restraint stress increases oocyte aneuploidy by impairing metaphase I spindle assembly and reducing spindle assembly checkpoint proteins in mice. Biol Reprod. 86:832012. View Article : Google Scholar | |
Lian HY, Gao Y, Jiao GZ, Sun MJ, Wu XF, Wang TY, Li H and Tan JH: Antioxidant supplementation overcomes the deleterious effects of maternal restraint stress-induced oxidative stress on mouse oocytes. Reproduction. 146:559–568. 2013. View Article : Google Scholar : PubMed/NCBI | |
Higaki S, Kishi M, Koyama K, Nagano M, Katagiri S, Takada T and Takahashi Y: Early germinal vesicle breakdown is a predictor of high preimplantation developmental competent oocytes in mice. Zygote. 25:41–48. 2017. View Article : Google Scholar | |
Zhao XY, Li ZB, Yuan HJ, Han X, Wu JS, Feng XY, Zhang M and Tan JH: Restraint stress and elevation of corticotrophin-releasing hormone in female mice impair oocyte competence through activation of the tumour necrosis factor α (TNF-α) system. Reprod Fertil Dev. 32:862–872. 2020. View Article : Google Scholar : PubMed/NCBI | |
Guo Y, Sun J, Bu S, Li B, Zhang Q, Wang Q and Lai D: Melatonin protects against chronic stress-induced oxidative meiotic defects in mice MII oocytes by regulating SIRT1. Cell Cycle. 19:1677–1695. 2020. View Article : Google Scholar : PubMed/NCBI | |
Chen RR, Wang J, Zhang M, Kong QQ, Sun GY, Jin CH and Tan JH: Restraint stress of female mice during oocyte development facilitates oocyte postovulatory aging. Aging (Albany NY). 14:9186–9199. 2022. View Article : Google Scholar : PubMed/NCBI | |
Dorfman M, Arancibia S, Fiedler JL and Lara HE: Chronic intermittent cold stress activates ovarian sympathetic nerves and modifies ovarian follicular development in the rat. Biol Reprod. 68:2038–2043. 2003. View Article : Google Scholar : PubMed/NCBI | |
Bernuci MP, Szawka RE, Helena CVV, Leite CM, Lara HE and Anselmo-Franci JA: Locus coeruleus mediates cold stress-induced polycystic ovary in rats. Endocrinology. 149:2907–2916. 2008. View Article : Google Scholar : PubMed/NCBI | |
Barra R, Cruz G, Mayerhofer A, Paredes A and Lara HE: Maternal sympathetic stress impairs follicular development and puberty of the offspring. Reproduction. 148:137–145. 2014. View Article : Google Scholar : PubMed/NCBI | |
Bernuci MP, Leite CM, Barros P, Kalil B, Leoni GB, Del Bianco-Borges B, Franci CR, Szawka RE, Lara HE and Anselmo-Franci JA: Transitory activation of the central and ovarian norepinephrine systems during cold stress-induced polycystic ovary in rats. J Neuroendocrinol. 25:23–33. 2013. View Article : Google Scholar | |
Pacak K, Palkovits M, Yadid G, Kvetnansky R, Kopin IJ and Goldstein DS: Heterogeneous neurochemical responses to different stressors: A test of Selye's doctrine of nonspecificity. Am J Physiol. 275:R1247–R1255. 1998.PubMed/NCBI | |
Riquelme R, Ruz F, Mayerhofer A and Lara HE: Role of ovarian sympathetic nerves and cholinergic local system during cold stress. J Endocrinol. 242:115–124. 2019. View Article : Google Scholar : PubMed/NCBI | |
Casillas F, Betancourt M, Juárez-Rojas L, Ducolomb Y, López A, Ávila-Quintero A, Zamora J, Ommati MM and Retana-Márquez S: Chronic stress detrimentally affects in vivo maturation in rat oocytes and oocyte viability at all phases of the estrous cycle. Animals (Basel). 11:24782021. View Article : Google Scholar : PubMed/NCBI | |
Wang D, Cheng X, Fang H, Ren Y, Li X, Ren W, Xue B and Yang C: Effect of cold stress on ovarian & uterine microcirculation in rats and the role of endothelin system. Reprod Biol Endocrinol. 18:292020. View Article : Google Scholar | |
Retana-Márquez S, Juárez-Rojas L, Ávila-Quintero A, Rojas-Maya S, Perera G, Casillas F, Betancourt M and Gómez-Quiroz L: Neuroendocrine disruption is associated to infertility in chronically stressed female rats. Reprod Biol. 20:474–483. 2020. View Article : Google Scholar : PubMed/NCBI | |
Keremu A, Yaoliwasi A, Tuerhong M, Kadeer N, Heyi, Yiming A and Yilike X: Research on the establishment of chronic stress-induced premature ovarian failure the rat model and effects of Chinese medicine Muniziqi treatment. Mol Reprod Dev. 86:175–186. 2019. View Article : Google Scholar | |
Ernsberger U, Deller T and Rohrer H: The sympathies of the body: Functional organization and neuronal differentiation in the peripheral sympathetic nervous system. Cell Tissue Res. 386:455–475. 2021. View Article : Google Scholar : PubMed/NCBI | |
Espinoza JA, Navarrete MI, Linares R, Chaparro-Ortega A, Ramírez DA, Rosas G, Vieyra E, Domínguez R and Morales-Ledesma L: Effects of chronic exposure to cold stress on ovarian functions in prepubertal rats. Reprod Biol. 23:1007562023. View Article : Google Scholar : PubMed/NCBI | |
Divyashree S and Yajurvedi HN: Long-term chronic stress exposure induces PCO phenotype in rat. Reproduction. 152:765–774. 2016. View Article : Google Scholar : PubMed/NCBI | |
Bunsueb S, Lapyuneyong N, Tongpan S, Arun S and Iamsaard S: Chronic stress increases the tyrosine phosphorylation in female reproductive organs: An experimental study. Int J Reprod Biomed. 19:87–96. 2021.PubMed/NCBI | |
Kim J and You S: High housing density-induced chronic stress diminishes ovarian reserve via granulosa cell apoptosis by angiotensin II overexpression in mice. Int J Mol Sci. 23:86142022. View Article : Google Scholar : PubMed/NCBI | |
Xi W, Mao H, Cui Z, Yao H, Shi R and Gao Y: Scream sound-induced chronic psychological stress results in diminished ovarian reserve in adult female rat. Endocrinology. 163:bqac0422022. View Article : Google Scholar : PubMed/NCBI | |
Ghatebi M, Zavareh S, Lashkarbolouki T and Elahdadi Salmani M: Implications from early life stress on the development of mouse ovarian follicles: Focus on oxidative stress. J Obstet Gynaecol Res. 45:1506–1514. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yuan J, Yu Y, Liu D and Sun Y: Associations between distinct dimensions of early life adversity and accelerated reproductive strategy among middle-aged women in China. Am J Obstet Gynecol. 226:104.e1–104.e14. 2022. View Article : Google Scholar | |
Mishra GD, Cooper R, Tom SE and Kuh D: Early life circumstances and their impact on menarche and menopause. Womens Health (Lond). 5:175–190. 2009. View Article : Google Scholar : PubMed/NCBI | |
Wang N, Huang Y, Wen J, Su Q, Huang Y, Cai L, Lin W, Zong L, Huang H, Qian X, et al: Early life exposure to famine and reproductive aging among Chinese women. Menopause. 26:463–468. 2019. View Article : Google Scholar | |
Liu YX, Cheng YN, Miao YL, Wei DL, Zhao LH, Luo MJ and Tan JH: Psychological stress on female mice diminishes the developmental potential of oocytes: A study using the predatory stress model. PLoS One. 7:e480832012. View Article : Google Scholar : PubMed/NCBI | |
Onalan E, Erbay B, Buran İK, Erol D, Tektemur A, Kuloglu T and Ozercan IH: Effects and mechanism of AP39 on ovarian functions in rats exposed to cisplatin and chronic immobilization stress. J Menopausal Med. 30:104–119. 2024. View Article : Google Scholar : PubMed/NCBI | |
Yuan HJ, Li ZB, Zhao XY, Sun GY, Wang GL, Zhao YQ, Zhang M and Tan JH: Glucocorticoids impair oocyte competence and trigger apoptosis of ovarian cells via activating the TNF-α system. Reproduction. 160:129–140. 2020. View Article : Google Scholar : PubMed/NCBI | |
Becker M, Pinhasov A and Ornoy A: Animal models of depression: What can they teach Us about the human disease? Diagnostics (Basel). 11:1232021. View Article : Google Scholar : PubMed/NCBI | |
Kendler KS: The genealogy of major depression: Symptoms and signs of melancholia from 1880 to 1900. Mol Psychiatry. 22:1539–1553. 2017. View Article : Google Scholar : PubMed/NCBI | |
Liu MY, Yin CY, Zhu LJ, Zhu XH, Xu C, Luo CX, Chen H, Zhu DY and Zhou QG: Sucrose preference test for measurement of stress-induced anhedonia in mice. Nat Protoc. 13:1686–1698. 2018. View Article : Google Scholar : PubMed/NCBI | |
Primo MJ, Fonseca-Rodrigues D, Almeida A, Teixeira PM and Pinto-Ribeiro F: Sucrose preference test: A systematic review of protocols for the assessment of anhedonia in rodents. Eur Neuropsychopharmacol. 77:80–92. 2023. View Article : Google Scholar : PubMed/NCBI | |
Verharen JPH, de Jong JW, Zhu Y and Lammel S: A computational analysis of mouse behavior in the sucrose preference test. Nat Commun. 14:24192023. View Article : Google Scholar : PubMed/NCBI | |
Porsolt RD, Le Pichon M and Jalfre M: Depression: A new animal model sensitive to antidepressant treatments. Nature. 266:730–732. 1977. View Article : Google Scholar : PubMed/NCBI | |
Cryan JF, Mombereau C and Vassout A: The tail suspension test as a model for assessing antidepressant activity: Review of pharmacological and genetic studies in mice. Neurosci Biobehav Rev. 29:571–625. 2005. View Article : Google Scholar : PubMed/NCBI | |
Prut L and Belzung C: The open field as a paradigm to measure the effects of drugs on anxiety-like behaviors: A review. Eur J Pharmacol. 463:3–33. 2003. View Article : Google Scholar : PubMed/NCBI | |
Kraeuter AK, Guest PC and Sarnyai Z: The open field test for measuring locomotor activity and anxiety-like behavior. Methods Mol Biol. 1916:99–103. 2019. View Article : Google Scholar | |
Seibenhener ML and Wooten MC: Use of the open field maze to measure locomotor and anxiety-like behavior in mice. J Vis Exp. e524342015.PubMed/NCBI | |
Bourin M and Hascoët M: The mouse light/dark box test. Eur J Pharmacol. 463:55–65. 2003. View Article : Google Scholar : PubMed/NCBI | |
Walf AA and Frye CA: The use of the elevated plus maze as an assay of anxiety-related behavior in rodents. Nat Protoc. 2:322–328. 2007. View Article : Google Scholar : PubMed/NCBI | |
Kraeuter AK, Guest PC and Sarnyai Z: The elevated plus maze test for measuring anxiety-like behavior in rodents. Methods Mol Biol. 1916:69–74. 2019. View Article : Google Scholar | |
Schrader AJ, Taylor RM, Lowery-Gionta EG and Moore NLT: Repeated elevated plus maze trials as a measure for tracking within-subjects behavioral performance in rats (Rattus norvegicus). PLoS One. 13:e02078042018. View Article : Google Scholar : PubMed/NCBI | |
Pietropaolo S: Mood and anxiety-related phenotypes in mice: Characterization using behavioral tests-Edited by T. D. Gould. Genes Brain Behav. 9:544. 2010. View Article : Google Scholar | |
Bae J, Lynch CD, Kim S, Sundaram R, Sapra KJ and Buck Louis GM: Preconception stress and the secondary sex ratio in a population-based preconception cohort. Fertil Steril. 107:714–722. 2017. View Article : Google Scholar : PubMed/NCBI | |
Ulrich-Lai YM and Herman JP: Neural regulation of endocrine and autonomic stress responses. Nat Rev Neurosci. 10:397–409. 2009. View Article : Google Scholar : PubMed/NCBI | |
Jacobson L: Hypothalamic-pituitary-adrenocortical axis regulation. Endocrinol Metab Clin North Am. 34:271–292. vii2005. View Article : Google Scholar : PubMed/NCBI | |
McCall JG, Al-Hasani R, Siuda ER, Hong DY, Norris AJ, Ford CP and Bruchas MR: CRH engagement of the locus coeruleus noradrenergic system mediates stress-induced anxiety. Neuron. 87:605–620. 2015. View Article : Google Scholar : PubMed/NCBI | |
Agorastos A and Chrousos GP: The neuroendocrinology of stress: The stress-related continuum of chronic disease development. Mol Psychiatry. 27:502–513. 2022. View Article : Google Scholar | |
Yoo ES, Yu J and Sohn JW: Neuroendocrine control of appetite and metabolism. Exp Mol Med. 53:505–516. 2021. View Article : Google Scholar : PubMed/NCBI | |
Di Comite G, Grazia Sabbadini M, Corti A, Rovere-Querini P and Manfredi AA: Conversation galante: How the immune and the neuroendocrine systems talk to each other. Autoimmun Rev. 7:23–29. 2007. View Article : Google Scholar : PubMed/NCBI | |
Smeenk JMJ, Verhaak CM, Vingerhoets AJJM, Sweep CGJ, Merkus JMWM, Willemsen SJ, van Minnen A, Straatman H and Braat DD: Stress and outcome success in IVF: The role of self-reports and endocrine variables. Hum Reprod. 20:991–996. 2005. View Article : Google Scholar : PubMed/NCBI | |
Hjollund NHI, Jensen TK, Bonde JP, Henriksen TB, Andersson AM, Kolstad HA, Ernst E, Giwercman A, Skakkebaek NE and Olsen J: Distress and reduced fertility: A follow-up study of first-pregnancy planners. Fertil Steril. 72:47–53. 1999. View Article : Google Scholar : PubMed/NCBI | |
Ates S, Yesil G, Sevket O, Molla T and Yildiz S: Comparison of metabolic profile and abdominal fat distribution between karyotypically normal women with premature ovarian insufficiency and age matched controls. Maturitas. 79:306–310. 2014. View Article : Google Scholar : PubMed/NCBI | |
Huang Y, Lv Y, Qi T, Luo Z, Meng X, Ying Q, Li D, Li C, Lan Y, Chu K, et al: Metabolic profile of women with premature ovarian insufficiency compared with that of age-matched healthy controls. Maturitas. 148:33–39. 2021. View Article : Google Scholar : PubMed/NCBI | |
Ferin M: Clinical review 105: Stress and the reproductive cycle. J Clin Endocrinol Metab. 84:1768–1774. 1999. View Article : Google Scholar : PubMed/NCBI | |
Chrousos GP, Torpy DJ and Gold PW: Interactions between the hypothalamic-pituitary-adrenal axis and the female reproductive system: Clinical implications. Ann Intern Med. 129:229–240. 1998. View Article : Google Scholar : PubMed/NCBI | |
Harlow CR, Fahy UM, Talbot WM, Wardle PG and Hull MG: Stress and stress-related hormones during in-vitro fertilization treatment. Hum Reprod. 11:274–279. 1996. View Article : Google Scholar : PubMed/NCBI | |
Demyttenaere K, Nijs P, Evers-Kiebooms G and Koninckx PR: Coping and the ineffectiveness of coping influence the outcome of in vitro fertilization through stress responses. Psychoneuroendocrinology. 17:655–665. 1992. View Article : Google Scholar : PubMed/NCBI | |
Pal L, Bevilacqua K and Santoro NF: Chronic psychosocial stressors are detrimental to ovarian reserve: A study of infertile women. J Psychosom Obstet Gynecol. 31:130–139. 2010. View Article : Google Scholar | |
Andersen CY: Effect of glucocorticoids on spontaneous and follicle-stimulating hormone induced oocyte maturation in mouse oocytes during culture. J Steroid Biochem Mol Biol. 85:423–427. 2003. View Article : Google Scholar : PubMed/NCBI | |
Yuan HJ, Han X, He N, Wang GL, Gong S, Lin J, Gao M and Tan JH: Glucocorticoids impair oocyte developmental potential by triggering apoptosis of ovarian cells via activating the Fas system. Sci Rep. 6:240362016. View Article : Google Scholar : PubMed/NCBI | |
Kalantaridou SN, Zoumakis E, Makrigiannakis A, Lavasidis LG, Vrekoussis T and Chrousos GP: Corticotropin-releasing hormone, stress and human reproduction: An update. J Reprod Immunol. 85:33–39. 2010. View Article : Google Scholar : PubMed/NCBI | |
Rivier C and Vale W: Influence of corticotropin-releasing factor on reproductive functions in the rat. Endocrinology. 114:914–921. 1984. View Article : Google Scholar : PubMed/NCBI | |
Breen KM, Thackray VG, Hsu T, Mak-McCully RA, Coss D and Mellon PL: Stress levels of glucocorticoids inhibit LHβ-subunit gene expression in gonadotrope cells. Mol Endocrinol. 26:1716–1731. 2012. View Article : Google Scholar : PubMed/NCBI | |
Luo E, Stephens SBZ, Chaing S, Munaganuru N, Kauffman AS and Breen KM: Corticosterone blocks ovarian cyclicity and the LH surge via decreased kisspeptin neuron activation in female mice. Endocrinology. 157:1187–1199. 2016. View Article : Google Scholar : | |
Roozendaal MM, Swarts JJM, Van Maanen JC, Wiegant VM and Mattheij JA: Inhibition of the LH surge in cyclic rats by stress is not mediated by opioids. Life Sci. 60:735–742. 1997. View Article : Google Scholar : PubMed/NCBI | |
Horowitz MA and Zunszain PA: Neuroimmune and neuroendocrine abnormalities in depression: Two sides of the same coin. Ann N Y Acad Sci. 1351:68–79. 2015. View Article : Google Scholar : PubMed/NCBI | |
Berga SL, Daniels TL and Giles DE: Women with functional hypothalamic amenorrhea but not other forms of anovulation display amplified cortisol concentrations. Fertil Steril. 67:1024–1030. 1997. View Article : Google Scholar : PubMed/NCBI | |
Deady LD and Sun J: A follicle rupture assay reveals an essential role for follicular adrenergic signaling in drosophila ovulation. PLoS Genet. 11:e10056042015. View Article : Google Scholar : PubMed/NCBI | |
Aguado LI: Role of the central and peripheral nervous system in the ovarian function. Microsc Res Tech. 59:462–473. 2002. View Article : Google Scholar : PubMed/NCBI | |
Lara HE, Dorfman M, Venegas M, Luza SM, Luna SL, Mayerhofer A, Guimaraes MA, Rosa E Silva AA and Ramírez VD: Changes in sympathetic nerve activity of the mammalian ovary during a normal estrous cycle and in polycystic ovary syndrome: Studies on norepinephrine release. Microsc Res Tech. 59:495–502. 2002. View Article : Google Scholar : PubMed/NCBI | |
Morales-Ledesma L, Linares R, Rosas G, Morán C, Chavira R, Cárdenas M and Domínguez R: Unilateral sectioning of the superior ovarian nerve of rats with polycystic ovarian syndrome restores ovulation in the innervated ovary. Reprod Biol Endocrinol. 8:992010. View Article : Google Scholar : PubMed/NCBI | |
Dissen GA, Romero C, Paredes A and Ojeda SR: Neurotrophic control of ovarian development. Microsc Res Tech. 59:509–515. 2002. View Article : Google Scholar : PubMed/NCBI | |
Uchida S and Kagitani F: Autonomic nervous regulation of ovarian function by noxious somatic afferent stimulation. J Physiol Sci. 65:1–9. 2015. View Article : Google Scholar | |
Acuña E, Fornes R, Fernandois D, Garrido MP, Greiner M, Lara HE and Paredes AH: Increases in norepinephrine release and ovarian cyst formation during ageing in the rat. Reprod Biol Endocrinol. 7:642009. View Article : Google Scholar : PubMed/NCBI | |
Casillas F, Flores-González A, Juárez-Rojas L, López A, Betancourt M, Casas E, Bahena I, Bonilla E and Retana-Márquez S: Chronic stress decreases fertility parameters in female rats. Syst Biol Reprod Med. 69:234–244. 2023. View Article : Google Scholar : PubMed/NCBI | |
Fernandois D, Cruz G, Na EK, Lara HE and Paredes AH: Kisspeptin level in the aging ovary is regulated by the sympathetic nervous system. J Endocrinol. 232:97–105. 2017. View Article : Google Scholar | |
Saller S, Merz-Lange J, Raffael S, Hecht S, Pavlik R, Thaler C, Berg D, Berg U, Kunz L and Mayerhofer A: Norepinephrine, active norepinephrine transporter, and norepinephrine-metabolism are involved in the generation of reactive oxygen species in human ovarian granulosa cells. Endocrinology. 153:1472–1483. 2012. View Article : Google Scholar : PubMed/NCBI | |
Zhang L, Gao J and Cui S: miR-21 is involved in norepinephrine-mediated rat granulosa cell apoptosis by targeting SMAD7. J Mol Endocrinol. 58:199–210. 2017. View Article : Google Scholar : PubMed/NCBI | |
Li XH, Ma YG, Geng LH, Qin L, Hu H and Li SW: Baseline psychological stress and ovarian norepinephrine levels negatively affect the outcome of in vitro fertilisation. Gynecol Endocrinol. 27:139–143. 2011. View Article : Google Scholar | |
Lynch CD, Sundaram R, Maisog JM, Sweeney AM and Louis GMB: Preconception stress increases the risk of infertility: Results from a couple-based prospective cohort study-the LIFE study. Hum Reprod. 29:1067–1075. 2014. View Article : Google Scholar : PubMed/NCBI | |
Hackett RA and Steptoe A: Type 2 diabetes mellitus and psychological stress-a modifiable risk factor. Nat Rev Endocrinol. 13:547–560. 2017. View Article : Google Scholar : PubMed/NCBI | |
Afrisham R, Paknejad M, Soliemanifar O, Sadegh-Nejadi S, Meshkani R and Ashtary-Larky D: The influence of psychological stress on the initiation and progression of diabetes and cancer. Int J Endocrinol Metab. 17:e674002019. | |
Joseph JJ and Golden SH: Cortisol dysregulation: The bidirectional link between stress, depression, and type 2 diabetes mellitus. Ann N Y Acad Sci. 1391:20–34. 2017. View Article : Google Scholar : | |
Balkan F, Cetin N, Usluogullari CA, Unal OK and Usluogullari B: Evaluation of the ovarian reserve function in patients with metabolic syndrome in relation to healthy controls and different age groups. J Ovarian Res. 7:632014. View Article : Google Scholar : PubMed/NCBI | |
Isik S, Ozcan HN, Ozuguz U, Tutuncu YA, Berker D, Alimli AG, Akbaba G, Karademir MA and Guler S: Evaluation of ovarian reserve based on hormonal parameters, ovarian volume, and antral follicle count in women with type 2 diabetes mellitus. J Clin Endocrinol Metab. 97:261–269. 2012. View Article : Google Scholar | |
Lustman PJ, Anderson RJ, Freedland KE, De Groot M, Carney RM and Clouse RE: Depression and poor glycemic control: A meta-analytic review of the literature. Diabetes Care. 23:934–942. 2000. View Article : Google Scholar : PubMed/NCBI | |
Yaribeygi H, Sathyapalan T, Atkin SL and Sahebkar A: Molecular mechanisms linking oxidative stress and diabetes mellitus. Oxid Med Cell Longev. 2020:86092132020. View Article : Google Scholar : PubMed/NCBI | |
Grigsby AB, Anderson RJ, Freedland KE, Clouse RE and Lustman PJ: Prevalence of anxiety in adults with diabetes: A systematic review. J Psychosom Res. 53:1053–1060. 2002. View Article : Google Scholar : PubMed/NCBI | |
Razzoli M, Pearson C, Crow S and Bartolomucci A: Stress, overeating, and obesity: Insights from human studies and preclinical models. Neurosci Biobehav Rev. 76:154–162. 2017. View Article : Google Scholar : PubMed/NCBI | |
Codner E, Merino PM and Tena-Sempere M: Female reproduction and type 1 diabetes: From mechanisms to clinical findings. Hum Reprod Update. 18:568–585. 2012. View Article : Google Scholar : PubMed/NCBI | |
Nandi A and Poretsky L: Diabetes and the female reproductive system. Endocrinol Metab Clin North Am. 42:915–946. 2013. View Article : Google Scholar : PubMed/NCBI | |
Dri M, Klinger FG and De Felici M: The ovarian reserve as target of insulin/IGF and ROS in metabolic disorder-dependent ovarian dysfunctions. Reprod Fertil. 2:R103–R112. 2021. View Article : Google Scholar | |
Barbieri RL, Makris A and Ryan KJ: Effects of insulin on steroidogenesis in cultured porcine ovarian theca. Fertil Steril. 40:237–241. 1983. View Article : Google Scholar : PubMed/NCBI | |
Barbieri RL, Makris A, Randall RW, Daniels G, Kistner RW and Ryan KJ: Insulin stimulates androgen accumulation in incubations of ovarian stroma obtained from women with hyperandrogenism. J Clin Endocrinol Metab. 62:904–910. 1986. View Article : Google Scholar : PubMed/NCBI | |
Cara JF and Rosenfield RL: Insulin-like growth factor I and insulin potentiate luteinizing hormone-induced androgen synthesis by rat ovarian thecal-interstitial cells. Endocrinology. 123:733–739. 1988. View Article : Google Scholar : PubMed/NCBI | |
Hudish LI, Reusch JE and Sussel L: β Cell dysfunction during progression of metabolic syndrome to type 2 diabetes. J Clin Invest. 129:4001–4008. 2019. View Article : Google Scholar : PubMed/NCBI | |
Escobar-Morreale HF, Roldán B, Barrio R, Alonso M, Sancho J, De La Calle H and García-Robles R: High prevalence of the polycystic ovary syndrome and hirsutism in women with type 1 diabetes mellitus. J Clin Endocrinol Metab. 85:4182–4187. 2000.PubMed/NCBI | |
Diamanti-Kandarakis E, Piperi C, Patsouris E, Korkolopoulou P, Panidis D, Pawelczyk L, Papavassiliou AG and Duleba AJ: Immunohistochemical localization of advanced glycation end-products (AGEs) and their receptor (RAGE) in polycystic and normal ovaries. Histochem Cell Biol. 127:581–589. 2007. View Article : Google Scholar : PubMed/NCBI | |
Secomandi L, Borghesan M, Velarde M and Demaria M: The role of cellular senescence in female reproductive aging and the potential for senotherapeutic interventions. Hum Reprod Update. 28:172–189. 2022. View Article : Google Scholar : | |
Willis DS, Watson H, Mason HD, Galea R, Brincat M and Franks S: Premature response to luteinizing hormone of granulosa cells from anovulatory women with polycystic ovary syndrome: Relevance to mechanism of anovulation. J Clin Endocrinol Metab. 83:3984–3991. 1998.PubMed/NCBI | |
Lee J, Lee HC, Kim SY, Cho GJ and Woodruff TK: Poorly-controlled type 1 diabetes mellitus impairs LH-LHCGR signaling in the ovaries and decreases female fertility in mice. Yonsei Med J. 60:667–678. 2019. View Article : Google Scholar : PubMed/NCBI | |
Zargar AH, Gupta VK, Wani AI, Masoodi SR, Bashir MI, Laway BA, Ganie MA and Salahuddin M: Prevalence of ultrasonography proved polycystic ovaries in North Indian women with type 2 diabetes mellitus. Reprod Biol Endocrinol. 3:352005. View Article : Google Scholar : PubMed/NCBI | |
Peppard HR, Marfori J, Iuorno MJ and Nestler JE: Prevalence of polycystic ovary syndrome among premenopausal women with type 2 diabetes. Diabetes Care. 24:1050–1052. 2001. View Article : Google Scholar : PubMed/NCBI | |
Khalil N, Sutton-Tyrrell K, Strotmeyer ES, Greendale GA, Vuga M, Selzer F, Crandall CJ and Cauley JA: Menopausal bone changes and incident fractures in diabetic women: A cohort study. Osteoporos Int. 22:1367–1376. 2011. View Article : Google Scholar | |
Sekhar TVDS, Medarametla S, Rahman A and Adapa SS: Early menopause in type 2 diabetes-A study from a South Indian tertiary care centre. J Clin Diagn Res. 9:OC08–OC10. 2015.PubMed/NCBI | |
Qin X, Du J, He R, Li Y, Zhu Q, Li Y, Li H and Liang X: Adverse effects of type 2 diabetes mellitus on ovarian reserve and pregnancy outcomes during the assisted reproductive technology process. Front Endocrinol (Lausanne). 14:12743272023. View Article : Google Scholar : PubMed/NCBI | |
Carr MC: The emergence of the metabolic syndrome with menopause. J Clin Endocrinol Metab. 88:2404–2411. 2003. View Article : Google Scholar : PubMed/NCBI | |
Kanaya AM, Herrington D, Vittinghoff E, Lin F, Grady D, Bittner V, Cauley JA and Barrett-Connor E; Heart and Estrogen/progestin Replacement Study: Glycemic effects of postmenopausal hormone therapy: The heart and estrogen/progestin replacement study. A randomized, double-blind, placebo-controlled trial. Ann Intern Med. 138:1–9. 2003. View Article : Google Scholar : PubMed/NCBI | |
Margolis KL, Bonds DE, Rodabough RJ, Tinker L, Phillips LS, Allen C, Bassford T, Burke G, Torrens J and Howard BV; Women's Health Initiative Investigators: Effect of oestrogen plus progestin on the incidence of diabetes in postmenopausal women: Results from the women's health initiative hormone trial. Diabetologia. 47:1175–1187. 2004. View Article : Google Scholar : PubMed/NCBI | |
Wagner JD, Thomas MJ, Williams JK, Zhang L, Greaves KA and Cefalu WT: Insulin sensitivity and cardiovascular risk factors in ovariectomized monkeys with estradiol alone or combined with nomegestrol acetate. J Clin Endocrinol Metab. 83:896–901. 1998.PubMed/NCBI | |
Kumagai S, Holmäng A and Björntorp P: The effects of oestrogen and progesterone on insulin sensitivity in female rats. Acta Physiol Scand. 149:91–97. 1993. View Article : Google Scholar : PubMed/NCBI | |
Cantave CY, Ruttle PL, Coté SM, Lupien SJ, Geoffroy MC, Vitaro F, Brendgen M, Tremblay R, Boivin M and Ouellet-Morin I: Body mass index across development and adolescent hair cortisol: The role of persistence, variability, and timing of exposure. Int J Obes (Lond). Oct 4–2024.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI | |
Christakoudi S, Asimakopoulos AG, Riboli E and Tsilidis KK: Links between the genetic determinants of morning plasma cortisol and body shape: A two-sample Mendelian randomisation study. Sci Rep. 14:32302024. View Article : Google Scholar : PubMed/NCBI | |
Kühnel A, Hagenberg J, Knauer-Arloth J, Ködel M, Czisch M, Sämann PG; BeCOME working group; Binder EB and Kroemer NB: Stress-induced brain responses are associated with BMI in women. Commun Biol. 6:10312023. View Article : Google Scholar : PubMed/NCBI | |
Lin YC, Lin CY, Saffari M, Tsai MC, Chang YH, Strong C, Chen JK, Hsieh YP, Yang YN and Latner JD: Weight stigma is associated with body mass index among college students in Taiwan: The mediated role of internalized weight stigma. BMC Psychol. 11:3652023. View Article : Google Scholar : PubMed/NCBI | |
Fardet L and Fève B: Systemic glucocorticoid therapy: A review of its metabolic and cardiovascular adverse events. Drugs. 74:1731–1745. 2014. View Article : Google Scholar : PubMed/NCBI | |
Santosa A, Rosengren A, Ramasundarahettige C, Rangarajan S, Gulec S, Chifamba J, Lear SA, Poirier P, Yeates KE, Yusuf R, et al: Psychosocial risk factors and cardiovascular disease and death in a population-based cohort from 21 low-, middle-, and high-income countries. JAMA Netw Open. 4:e21389202021. View Article : Google Scholar : PubMed/NCBI | |
Ponti F, Santoro A, Mercatelli D, Gasperini C, Conte M, Martucci M, Sangiorgi L, Franceschi C and Bazzocchi A: Aging and imaging assessment of body composition: From fat to facts. Front Endocrinol (Lausanne). 10:8612020. View Article : Google Scholar : PubMed/NCBI | |
Van Der Valk ES, Savas M and Van Rossum EFC: Stress and obesity: Are there more susceptible individuals? Curr Obes Rep. 7:193–203. 2018. View Article : Google Scholar : PubMed/NCBI | |
Chen X, Xiao Z, Cai Y, Huang L and Chen C: Hypothalamic mechanisms of obesity-associated disturbance of hypothalamic-pituitary-ovarian axis. Trends Endocrinol Metab. 33:206–217. 2022. View Article : Google Scholar : PubMed/NCBI | |
Morimoto A, Rose RD, Smith KM, Dinh DT, Umehara T, Winstanley YE, Shibahara H, Russell DL and Robker RL: Granulosa cell metabolism at ovulation correlates with oocyte competence and is disrupted by obesity and aging. Hum Reprod. 39:2053–2066. 2024. View Article : Google Scholar : PubMed/NCBI | |
Schon SB, Cabre HE and Redman LM: The impact of obesity on reproductive health and metabolism in reproductive-age females. Fertil Steril. 122:194–203. 2024. View Article : Google Scholar : PubMed/NCBI | |
Martinuzzi K, Ryan S, Luna M and Copperman AB: Elevated body mass index (BMI) does not adversely affect in vitro fertilization outcome in young women. J Assist Reprod Genet. 25:169–175. 2008. View Article : Google Scholar : PubMed/NCBI | |
Bernardi LA, Carnethon MR, de Chavez PJ, Ikhena DE, Neff LM, Baird DD and Marsh EE: Relationship between obesity and anti-Müllerian hormone in reproductive-aged African American women. Obesity (Silver Spring). 25:229–235. 2017. View Article : Google Scholar | |
Moy V, Jindal S, Lieman H and Buyuk E: Obesity adversely affects serum anti-müllerian hormone (AMH) levels in Caucasian women. J Assist Reprod Genet. 32:1305–1311. 2015. View Article : Google Scholar : PubMed/NCBI | |
Freeman EW, Gracia CR, Sammel MD, Lin H, Lim LCL and Strauss JF III: Association of anti-mullerian hormone levels with obesity in late reproductive-age women. Fertil Steril. 87:101–106. 2007. View Article : Google Scholar | |
Wang Y, Wu L, Yang Z, Xu R, Duan Y, Lin J, Cui X, Fan C, Zhou Y, Bao W, et al: Association of body mass index with serum anti-Müllerian hormone and inhibin B levels among 8323 women attending a reproductive medical center: A cross-sectional study. Endocrine. 75:284–292. 2022. View Article : Google Scholar | |
Douchi T, Kuwahata R, Yamamoto S, Oki T, Yamasaki H and Nagata Y: Relationship of upper body obesity to menstrual disorders. Acta Obstet Gynecol Scand. 81:147–150. 2002. View Article : Google Scholar : PubMed/NCBI | |
Rittmaster RS, Deshwal N and Lehman L: The role of adrenal hyperandrogenism, insulin resistance, and obesity in the pathogenesis of polycystic ovarian syndrome. J Clin Endocrinol Metab. 76:1295–1300. 1993.PubMed/NCBI | |
Di Berardino C, Peserico A, Capacchietti G, Zappacosta A, Bernabò N, Russo V, Mauro A, El Khatib M, Gonnella F, Konstantinidou F, et al: High-fat diet and female fertility across lifespan: A comparative lesson from mammal models. Nutrients. 14:43412022. View Article : Google Scholar : PubMed/NCBI | |
Wang N, Luo LL, Xu JJ, Xu MY, Zhang XM, Zhou XL, Liu WJ and Fu YC: Obesity accelerates ovarian follicle development and follicle loss in rats. Metabolism. 63:94–103. 2014. View Article : Google Scholar | |
Jungheim ES, Schoeller EL, Marquard KL, Louden ED, Schaffer JE and Moley KH: Diet-induced obesity model: Abnormal oocytes and persistent growth abnormalities in the offspring. Endocrinology. 151:4039–4046. 2010. View Article : Google Scholar : PubMed/NCBI | |
Luzzo KM, Wang Q, Purcell SH, Chi M, Jimenez PT, Grindler N, Schedl T and Moley KH: High fat diet induced developmental defects in the mouse: Oocyte meiotic aneuploidy and fetal growth retardation/brain defects. PLoS One. 7:e492172012. View Article : Google Scholar : PubMed/NCBI | |
Reynolds KA, Boudoures AL, Chi MMY, Wang Q and Moley KH: Adverse effects of obesity and/or high-fat diet on oocyte quality and metabolism are not reversible with resumption of regular diet in mice. Reprod Fertil Dev. 27:716–724. 2015. View Article : Google Scholar : PubMed/NCBI | |
Machtinger R, Combelles CMH, Missmer SA, Correia KF, Fox JH and Racowsky C: The association between severe obesity and characteristics of failed fertilized oocytes. Hum Reprod. 27:3198–3207. 2012. View Article : Google Scholar : PubMed/NCBI | |
Kloos J, Perez J and Weinerman R: Increased body mass index is negatively associated with ovarian reserve as measured by anti-Müllerian hormone in patients with polycystic ovarian syndrome. Clin Obes. 29:e126382024. View Article : Google Scholar | |
Beroukhim G, Esencan E and Seifer DB: Impact of sleep patterns upon female neuroendocrinology and reproductive outcomes: A comprehensive review. Reprod Biol Endocrinol. 20:162022. View Article : Google Scholar | |
Yan J, Zhang X, Zhu K, Yu M, Liu Q, De Felici M, Zhang T, Wang J and Shen W: Sleep deprivation causes gut dysbiosis impacting on systemic metabolomics leading to premature ovarian insufficiency in adolescent mice. Theranostics. 14:3760–3776. 2024. View Article : Google Scholar : PubMed/NCBI | |
Weng L, Hong H, Zhang Q, Xiao C, Zhang Q, Wang Q, Huang J and Lai D: Sleep deprivation triggers the excessive activation of ovarian primordial follicles via β2 adrenergic receptor signaling. Adv Sci (Weinh). 11:e24023932024. View Article : Google Scholar | |
Andersen ML, Alvarenga TAF, Guindalini C, Perry JC, Silva A, Zager A and Tufik S: Paradoxical sleep deprivation influences sexual behavior in female rats. J Sex Med. 6:2162–2172. 2009. View Article : Google Scholar : PubMed/NCBI | |
Koehl M, Battle S and Meerlo P: Sex differences in sleep: The response to sleep deprivation and restraint stress in mice. Sleep. 29:1224–1231. 2006. View Article : Google Scholar : PubMed/NCBI | |
Yang CK, Tsai HD, Wu CH and Cho CL: The role of glucocorticoids in ovarian development of sleep deprived rats. Taiwan J Obstet Gynecol. 58:122–127. 2019. View Article : Google Scholar : PubMed/NCBI | |
Yang CK, Wu RSC, Wu CH, Lin TRY and Tsai HD: Sleep deprivation enhances peripheral serotonin secretion to regulate the large follicle steroidogenesis of rats. Taiwan J Obstet Gynecol. 54:260–265. 2015. View Article : Google Scholar : PubMed/NCBI | |
Stock D, Knight JA, Raboud J, Cotterchio M, Strohmaier S, Willett W, Eliassen AH, Rosner B, Hankinson SE and Schernhammer E: Rotating night shift work and menopausal age. Hum Reprod. 34:539–548. 2019. View Article : Google Scholar : PubMed/NCBI | |
Reiter RJ, Mayo JC, Tan DX, Sainz RM, Alatorre-Jimenez M and Qin L: Melatonin as an antioxidant: Under promises but over delivers. J Pineal Res. 61:253–278. 2016. View Article : Google Scholar : PubMed/NCBI | |
Tamura H, Takasaki A, Taketani T, Tanabe M, Kizuka F, Lee L, Tamura I, Maekawa R, Asada H, Yamagata Y and Sugino N: Melatonin as a free radical scavenger in the ovarian follicle. Endocr J. 60:1–13. 2013. View Article : Google Scholar | |
Lahiri DK, Ge Y, Sharman EH and Bondy SC: Age-related changes in serum melatonin in mice: Higher levels of combined melatonin and 6-hydroxymelatonin sulfate in the cerebral cortex than serum, heart, liver and kidney tissues. J Pineal Res. 36:217–223. 2004. View Article : Google Scholar : PubMed/NCBI | |
Tanabe M, Tamura H, Taketani T, Okada M, Lee L, Tamura I, Maekawa R, Asada H, Yamagata Y and Sugino N: Melatonin protects the integrity of granulosa cells by reducing oxidative stress in nuclei, mitochondria, and plasma membranes in mice. J Reprod Dev. 61:35–41. 2015. View Article : Google Scholar : | |
Shen M, Cao Y, Jiang Y, Wei Y and Liu H: Melatonin protects mouse granulosa cells against oxidative damage by inhibiting FOXO1-mediated autophagy: Implication of an antioxidation-independent mechanism. Redox Biol. 18:138–157. 2018. View Article : Google Scholar : PubMed/NCBI | |
Zhai B, Li X, Zhao Z, Cao Y, Liu X, Liu Z, Ma H and Lu W: Melatonin protects the apoptosis of sheep granulosa cells by suppressing oxidative stress via MAP3K8 and FOS pathway. Genes (Basel). 14:10672023. View Article : Google Scholar : PubMed/NCBI | |
Xu G, Dong Y, Wang Z, Ding H, Wang J, Zhao J, Liu H and Lv W: Melatonin attenuates oxidative stress-induced apoptosis of bovine ovarian granulosa cells by promoting mitophagy via SIRT1/FoxO1 signaling pathway. Int J Mol Sci. 24:128542023. View Article : Google Scholar : PubMed/NCBI | |
Tamura H, Takasaki A, Miwa I, Taniguchi K, Maekawa R, Asada H, Taketani T, Matsuoka A, Yamagata Y, Shimamura K, et al: Oxidative stress impairs oocyte quality and melatonin protects oocytes from free radical damage and improves fertilization rate. J Pineal Res. 44:280–287. 2008. View Article : Google Scholar : PubMed/NCBI | |
Zhang H, Li C, Wen D, Li R, Lu S, Xu R, Tang Y, Sun Y, Zhao X, Pan M and Ma B: Melatonin improves the quality of maternally aged oocytes by maintaining intercellular communication and antioxidant metabolite supply. Redox Biol. 49:1022152022. View Article : Google Scholar | |
Lord T, Nixon B, Jones KT and Aitken RJ: Melatonin prevents postovulatory oocyte aging in the mouse and extends the window for optimal fertilization in vitro. Biol Reprod. 88:672013. View Article : Google Scholar : PubMed/NCBI | |
Zhang M, Lu Y, Chen Y, Zhang Y and Xiong B: Insufficiency of melatonin in follicular fluid is a reversible cause for advanced maternal age-related aneuploidy in oocytes. Redox Biol. 28:1013272020. View Article : Google Scholar | |
Yang D, Mu Y, Wang J, Zou W, Zou H, Yang H, Zhang C, Fan Y, Zhang H, Zhang H, et al: Melatonin enhances the developmental potential of immature oocytes from older reproductive-aged women by improving mitochondrial function. Heliyon. 9:e193662023. View Article : Google Scholar : PubMed/NCBI | |
Li C, He X, Huang Z, Han L, Wu X, Li L, Xin Y, Ge J, Sha J, Yin Z and Wang Q: Melatonin ameliorates the advanced maternal age-associated meiotic defects in oocytes through the SIRT2-dependent H4K16 deacetylation pathway. Aging (Albany NY). 12:1610–1623. 2020. View Article : Google Scholar : PubMed/NCBI | |
Takasaki A, Nakamura Y, Tamura H, Shimamura K and Morioka H: Melatonin as a new drug for improving oocyte quality. Reprod Med Biol. 2:139–144. 2004. View Article : Google Scholar : PubMed/NCBI | |
Baden MY, Hu FB, Vetter C, Schernhammer E, Redline S and Huang T: Sleep duration patterns in early to middle adulthood and subsequent risk of type 2 diabetes in women. Diabetes Care. 43:1219–1226. 2020. View Article : Google Scholar : PubMed/NCBI | |
Rooney KL and Domar AD: The impact of stress on fertility treatment. Curr Opin Obstet Gynecol. 28:198–201. 2016. View Article : Google Scholar : PubMed/NCBI | |
Ramezanzadeh F, Noorbala AA, Abedinia N, Rahimi Forooshani A and Naghizadeh MM: Psychiatric intervention improved pregnancy rates in infertile couples. Malays J Med Sci. 18:16–24. 2011.PubMed/NCBI | |
Rock J, Tietze C and McLaughlin HB: Effect of adoption on infertility. Fertil Steril. 16:305–312. 1965. View Article : Google Scholar : PubMed/NCBI | |
Weir WC and Weir DR: Adoption and subsequent conceptions. Fertil Steril. 17:283–288. 1966. View Article : Google Scholar : PubMed/NCBI | |
Nakao M, Shirotsuki K and Sugaya N: Cognitive-behavioral therapy for management of mental health and stress-related disorders: Recent advances in techniques and technologies. Biopsychosoc Med. 15:162021. View Article : Google Scholar : PubMed/NCBI | |
Hoffman SG and Smits JAJ: Cognitive-behavioral therapy for adult anxiety disorders: A meta-analysis of randomized placebo-controlled trials. J Clin Psychiatry. 69:621–632. 2008. View Article : Google Scholar | |
Powers MB, Halpern JM, Ferenschak MP, Gillihan SJ and Foa EB: A meta-analytic review of prolonged exposure for posttraumatic stress disorder. Clin Psychol Rev. 30:635–641. 2010. View Article : Google Scholar : PubMed/NCBI | |
Olatunji BO, Davis ML, Powers MB and Smits JAJ: Cognitive-behavioral therapy for obsessive-compulsive disorder: A meta-analysis of treatment outcome and moderators. J Psychiatr Res. 47:33–41. 2013. View Article : Google Scholar | |
Cuijpers P, Berking M, Andersson G, Quigley L, Kleiboer A and Dobson KS: A meta-analysis of cognitive-behavioural therapy for adult depression, alone and in comparison with other treatments. Can J Psychiatry. 58:376–385. 2013. View Article : Google Scholar : PubMed/NCBI | |
Hofmann SG, Asnaani A, Vonk IJJ, Sawyer AT and Fang A: The efficacy of cognitive behavioral therapy: A review of meta-analyses. Cogn Ther Res. 36:427–440. 2012. View Article : Google Scholar | |
Golshani F, Hasanpour S, Mirghafourvand M and Esmaeilpour K: Effect of cognitive behavioral therapy-based counseling on perceived stress in pregnant women with history of primary infertility: A controlled randomized clinical trial. BMC Psychiatry. 21:2782021. View Article : Google Scholar : PubMed/NCBI | |
Domar AD, Rooney KL, Wiegand B, Orav EJ, Alper MM, Berger BM and Nikolovski J: Impact of a group mind/body intervention on pregnancy rates in IVF patients. Fertil Steril. 95:2269–2273. 2011. View Article : Google Scholar : PubMed/NCBI | |
Faramarzi M, Pasha H, Esmailzadeh S, Kheirkhah F, Heidary S and Afshar Z: The effect of the cognitive behavioral therapy and pharmacotherapy on infertility stress: A randomized controlled trial. Int J Fertil Steril. 7:199–206. 2013. | |
Sexton MB, Byrd MR, O'Donohue WT and Jacobs NN: Web-based treatment for infertility-related psychological distress. Arch Womens Ment Health. 13:347–358. 2010. View Article : Google Scholar : PubMed/NCBI | |
John JB, Chellaiyan DVG, Gupta S and Nithyanandham R: How effective the mindfulness-based cognitive behavioral therapy on quality of life in women with menopause. J Midlife Health. 13:169–174. 2022.PubMed/NCBI | |
Green SM and Furtado M: Cognitive behavioral therapy for sexual concerns during perimenopause: A four session study protocol. Front Glob Womens Health. 2:7447482021. View Article : Google Scholar : PubMed/NCBI | |
Green SM, Donegan E, Frey BN, Fedorkow DM, Key BL, Streiner DL and McCabe RE: Cognitive behavior therapy for menopausal symptoms (CBT-Meno): A randomized controlled trial. Menopause. 26:972–980. 2019. View Article : Google Scholar : PubMed/NCBI | |
Green SM, Haber E, McCabe RE and Soares CN: Cognitive-behavioral group treatment for menopausal symptoms: A pilot study. Arch Womens Ment Health. 16:325–332. 2013. View Article : Google Scholar : PubMed/NCBI | |
Reddy NV and Omkarappa DB: Cognitive-behavioral therapy for depression among menopausal woman: A randomized controlled trial. J Fam Med Prim Care. 8:1002–1006. 2019. View Article : Google Scholar | |
Donegan E, Frey BN, McCabe RE, Streiner DL, Fedorkow DM, Furtado M and Green SM: Impact of the CBT-Meno protocol on menopause-specific beliefs, dysfunctional attitudes, and coping behaviors. Menopause. 29:963–972. 2022. View Article : Google Scholar : PubMed/NCBI | |
Shabani F, Farvareshi M, Hamdi K, Sadeghzadeh Oskouei B, Montazeri M and Mirghafourvand M: The effect of cognitive-behavioral therapy on stress and anxiety of women with premature ovarian insufficiency: A randomized controlled trial. Post Reprod Health. 28:211–221. 2022. View Article : Google Scholar : PubMed/NCBI | |
Berga SL, Marcus MD, Loucks TL, Hlastala S, Ringham R and Krohn MA: Recovery of ovarian activity in women with functional hypothalamic amenorrhea who were treated with cognitive behavior therapy. Fertil Steril. 80:976–981. 2003. View Article : Google Scholar : PubMed/NCBI | |
Michopoulos V, Mancini F, Loucks TL and Berga SL: Neuroendocrine recovery initiated by cognitive behavioral therapy in women with functional hypothalamic amenorrhea: A randomized, controlled trial. Fertil Steril. 99:2084–2091.e1. 2013. View Article : Google Scholar : PubMed/NCBI | |
Sockalingam S, Leung SE, Ma C, Tomlinson G, Hawa R, Wnuk S, Jackson T, Urbach D, Okrainec A, Brown J, et al: Efficacy of telephone-based cognitive behavioral therapy for weight loss, disordered eating, and psychological distress after bariatric surgery: A randomized clinical trial. JAMA Netw Open. 6:e23270992023. View Article : Google Scholar : PubMed/NCBI | |
Rofey DL, Szigethy EM, Noll RB, Dahl RE, Lobst E and Arslanian SA: Cognitive-behavioral therapy for physical and emotional disturbances in adolescents with polycystic ovary syndrome: A pilot study. J Pediatr Psychol. 34:156–163. 2009. View Article : Google Scholar | |
Cooney LG, Milman LW, Hantsoo L, Kornfield S, Sammel MD, Allison KC, Epperson CN and Dokras A: Cognitive-behavioral therapy improves weight loss and quality of life in women with polycystic ovary syndrome: A pilot randomized clinical trial. Fertil Steril. 110:161–171.e1. 2018. View Article : Google Scholar : PubMed/NCBI | |
Ludwig DS and Kabat-Zinn J: Mindfulness in medicine. JAMA. 300:1350–1352. 2008. View Article : Google Scholar : PubMed/NCBI | |
Grossman P, Niemann L, Schmidt S and Walach H: Mindfulness-based stress reduction and health benefits. A meta-analysis. J Psychosom Res. 57:35–43. 2004. View Article : Google Scholar : PubMed/NCBI | |
Rosenzweig S, Greeson JM, Reibel DK, Green JS, Jasser SA and Beasley D: Mindfulness-based stress reduction for chronic pain conditions: Variation in treatment outcomes and role of home meditation practice. J Psychosom Res. 68:29–36. 2010. View Article : Google Scholar | |
Wong C, Yip BHK, Gao T, Lam KYY, Woo DMS, Yip ALK, Chin CY, Tang WPY, Choy MMT, Tsang KWK, et al: Mindfulness-based stress reduction (MBSR) or psychoeducation for the reduction of menopausal symptoms: A randomized, controlled clinical trial. Sci Rep. 8:66092018. View Article : Google Scholar : PubMed/NCBI | |
Pyri F, Abedi P, Maraghi E and Jefreh M: The effect of mindfulness on quality of life among women with premature ovarian insufficiency: A randomized clinical trial. J Midlife Health. 12:116–121. 2021.PubMed/NCBI | |
Majeed MH, Ali AA and Sudak DM: Mindfulness-based interventions for chronic pain: Evidence and applications. Asian J Psychiatry. 32:79–83. 2018. View Article : Google Scholar | |
Kalhori F, Masoumi SZ, Shamsaei F, Mohammadi Y and Yavangi M: Effect of mindfulness-based group counseling on depression in infertile women: Randomized clinical trial study. Int J Fertil Steril. 14:10–6. 2020.PubMed/NCBI | |
Galhardo A, Cunha M and Pinto-Gouveia J: A 7-year follow-up study of the mindfulness-based program for infertility: Are there long-term effects? Clin Psychol Psychother. 26:409–417. 2019. View Article : Google Scholar : PubMed/NCBI | |
Aalbers S, Fusar-Poli L, Freeman RE, Spreen M, Ket JC, Vink AC, Maratos A, Crawford M, Chen XJ and Gold C: Music therapy for depression. Cochrane Database Syst Rev. 11:CD0045172017.PubMed/NCBI | |
Koçak DY and Varişoğlu Y: The effect of music therapy on menopausal symptoms and depression: A randomized-controlled study. Menopause. 29:545–552. 2022. View Article : Google Scholar : PubMed/NCBI | |
Kim S, Kim SM, Hwang H, Kim MK, Kim HJ, Park S and Han DH: The effects of music therapy on the psychological status of women with perimenopause syndrome. Menopause. 30:1045–1052. 2023. View Article : Google Scholar : PubMed/NCBI | |
Mahmoud MY, Labib K, Sileem SA, Mustafa FA, Hamed WM, Abd Elhamid A, Saleh DM, Alanwar A, Riad AAM, Abdelhakim AM, et al: The impact of music therapy on anxiety and pregnancy rate among infertile women undergoing assisted reproductive technologies: A systematic review and meta-analysis. J Psychosom Obstet Gynecol. 43:205–213. 2022. View Article : Google Scholar | |
Aba YA, Avci D, Guzel Y, Ozcelik SK and Gurtekin B: Effect of music therapy on the anxiety levels and pregnancy rate of women undergoing in vitro fertilization-embryo transfer: A randomized controlled trial. Appl Nurs Res. 36:19–24. 2017. View Article : Google Scholar : PubMed/NCBI | |
Domar AD, Gross J, Rooney K and Boivin J: Exploratory randomized trial on the effect of a brief psychological intervention on emotions, quality of life, discontinuation, and pregnancy rates in in vitro fertilization patients. Fertil Steril. 104:440–451.e7. 2015. View Article : Google Scholar : PubMed/NCBI | |
Clifton J, Parent J, Seehuus M, Worrall G, Forehand R and Domar A: An internet-based mind/body intervention to mitigate distress in women experiencing infertility: A randomized pilot trial. PLoS One. 15:e02293792020. View Article : Google Scholar : PubMed/NCBI | |
Brinsley J, Schuch F, Lederman O, Girard D, Smout M, Immink MA, Stubbs B, Firth J, Davison K and Rosenbaum S: Effects of yoga on depressive symptoms in people with mental disorders: A systematic review and meta-analysis. Br J Sports Med. 55:992–1000. 2021. View Article : Google Scholar | |
Darbandi S, Darbandi M, Khorram Khorshid HR and Sadeghi MR: Yoga can improve assisted reproduction technology outcomes in couples with infertility. Altern Ther Health Med. 24:50–55. 2018. | |
Oron G, Allnutt E, Lackman T, Sokal-Arnon T, Holzer H and Takefman J: A prospective study using hatha yoga for stress reduction among women waiting for IVF treatment. Reprod Biomed Online. 30:542–548. 2015. View Article : Google Scholar : PubMed/NCBI | |
Valoriani V, Lotti F, Vanni C, Noci MC, Fontanarosa N, Ferrari G, Cozzi C and Noci I: Hatha-yoga as a psychological adjuvant for women undergoing IVF: A pilot study. Eur J Obstet Gynecol Reprod Biol. 176:158–162. 2014. View Article : Google Scholar : PubMed/NCBI | |
Lu X, Liu L and Yuan R: Effect of the information support method combined with yoga exercise on the depression, anxiety, and sleep quality of menopausal women. Psychiatr Danub. 32:380–388. 2020. View Article : Google Scholar : PubMed/NCBI | |
Jorge MP, Santaella DF, Pontes IMO, Shiramizu VKM, Nascimento EB, Cabral A, Lemos TM, Silva RH and Ribeiro AM: Hatha yoga practice decreases menopause symptoms and improves quality of life: A randomized controlled trial. Complement Ther Med. 26:128–135. 2016. View Article : Google Scholar : PubMed/NCBI | |
Frederiksen Y, O'Toole MS, Mehlsen MY, Hauge B, Elbaek HO, Zachariae R and Ingerslev HJ: The effect of expressive writing intervention for infertile couples: a randomized controlled trial. Hum Reprod. 32:391–402. 2017. View Article : Google Scholar | |
Hart RJ: Physiological aspects of female fertility: Role of the environment, modern lifestyle, and genetics. Physiol Rev. 96:873–909. 2016. View Article : Google Scholar : PubMed/NCBI | |
Waldman M, Cohen K, Yadin D, Nudelman V, Gorfil D, Laniado-Schwartzman M, Kornwoski R, Aravot D, Abraham NG, Arad M and Hochhauser E: Regulation of diabetic cardiomyopathy by caloric restriction is mediated by intracellular signaling pathways involving 'SIRT1 and PGC-1α'. Cardiovasc Diabetol. 17:1112018. View Article : Google Scholar | |
Bordone L and Guarente L: Calorie restriction, SIRT1 and metabolism: Understanding longevity. Nat Rev Mol Cell Biol. 6:298–305. 2005. View Article : Google Scholar | |
Levine B and Kroemer G: Autophagy in the pathogenesis of disease. Cell. 132:27–42. 2008. View Article : Google Scholar : PubMed/NCBI | |
Selesniemi K, Lee HJ, Muhlhauser A and Tilly JL: Prevention of maternal aging-associated oocyte aneuploidy and meiotic spindle defects in mice by dietary and genetic strategies. Proc Natl Acad Sci USA. 108:12319–12324. 2011. View Article : Google Scholar : PubMed/NCBI | |
Smits A, Marei WFA, Moorkens K, Bols PEJ, De Neubourg D and Leroy JLMR: Obese outbred mice only partially benefit from diet normalization or calorie restriction as preconception care interventions to improve metabolic health and oocyte quality. Hum Reprod. 37:2867–2884. 2022. View Article : Google Scholar : PubMed/NCBI | |
Mishina T, Tabata N, Hayashi T, Yoshimura M, Umeda M, Mori M, Ikawa Y, Hamada H, Nikaido I and Kitajima TS: Single-oocyte transcriptome analysis reveals aging-associated effects influenced by life stage and calorie restriction. Aging Cell. 20:e134282021. View Article : Google Scholar : PubMed/NCBI | |
Słuczanowska-Głąbowska S, Laszczyńska M, Piotrowska K, Grabowska M, Grymuła K and Ratajczak MZ: Caloric restriction increases ratio of estrogen to androgen receptors expression in murine ovaries-potential therapeutic implications. J Ovarian Res. 8:572015. View Article : Google Scholar | |
Sharov AA, Falco G, Piao Y, Poosala S, Becker KG, Zonderman AB, Longo DL, Schlessinger D and Ko MSh: Effects of aging and calorie restriction on the global gene expression profiles of mouse testis and ovary. BMC Biol. 6:242008. View Article : Google Scholar : PubMed/NCBI | |
Garcia DN, Saccon TD, Pradiee J, Rincón JAA, Andrade KRS, Rovani MT, Mondadori RG, Cruz LAX, Barros CC, Masternak MM, et al: Effect of caloric restriction and rapamycin on ovarian aging in mice. Geroscience. 41:395–408. 2019. View Article : Google Scholar : PubMed/NCBI | |
Isola JVV, Zanini BM, Hense JD, Alvarado-Rincón JA, Garcia DN, Pereira GC, Vieira AD, Oliveira TL, Collares T, Gasperin BG, et al: Mild calorie restriction, but not 17α-estradiol, extends ovarian reserve and fertility in female mice. Exp Gerontol. 159:1116692022. View Article : Google Scholar | |
Liu WJ, Zhang XM, Wang N, Zhou XL, Fu YC and Luo LL: Calorie restriction inhibits ovarian follicle development and follicle loss through activating SIRT1 signaling in mice. Eur J Med Res. 20:222015. View Article : Google Scholar : PubMed/NCBI | |
Zhou XL, Xu JJ, Ni YH, Chen XC, Zhang HX, Zhang XM, Liu WJ, Luo LL and Fu YC: SIRT1 activator (SRT1720) improves the follicle reserve and prolongs the ovarian lifespan of diet-induced obesity in female mice via activating SIRT1 and suppressing mTOR signaling. J Ovarian Res. 7:972014. View Article : Google Scholar : PubMed/NCBI | |
Li L, Fu YC, Xu JJ, Lin XH, Chen XC, Zhang XM and Luo LL: Caloric restriction promotes the reserve of follicle pool in adult female rats by inhibiting the activation of mammalian target of rapamycin signaling. Reprod Sci. 22:60–67. 2015. View Article : Google Scholar : | |
Smits A, Marei WFA, De Neubourg D and Leroy JLMR: Diet normalization or caloric restriction as a preconception care strategy to improve metabolic health and oocyte quality in obese outbred mice. Reprod Biol Endocrinol. 19:1662021. View Article : Google Scholar : PubMed/NCBI | |
Chen G, Xie W, Nah J, Sauvat A, Liu P, Pietrocola F, Sica V, Carmona-Gutierrez D, Zimmermann A, Pendl T, et al: 3,4-Dimethoxychalcone induces autophagy through activation of the transcription factors TFE3 and TFEB. EMBO Mol Med. 11:e104692019. View Article : Google Scholar : PubMed/NCBI | |
Kepp O, Chen G, Carmona-Gutierrez D, Madeo F and Kroemer G: A discovery platform for the identification of caloric restriction mimetics with broad health-improving effects. Autophagy. 16:188–189. 2020. View Article : Google Scholar : | |
Zhang H, Ni W, Yu G, Geng Y, Lou J, Qi J, Chen Y, Li F, Ye H, Ma H, et al: 3,4-Dimethoxychalcone, a caloric restriction mimetic, enhances TFEB-mediated autophagy and alleviates pyroptosis and necroptosis after spinal cord injury. Theranostics. 13:810–832. 2023. View Article : Google Scholar : PubMed/NCBI | |
Jafari M, Macho-González A, Diaz A, Lindenau K, Santiago-Fernández O, Zeng M, Massey AC, de Cabo R, Kaushik S and Cuervo AM: Calorie restriction and calorie-restriction mimetics activate chaperone-mediated autophagy. Proc Natl Acad Sci USA. 121:e23179451212024. View Article : Google Scholar : PubMed/NCBI | |
Sciarretta S, Forte M, Castoldi F, Frati G, Versaci F, Sadoshima J, Kroemer G and Maiuri MC: Caloric restriction mimetics for the treatment of cardiovascular diseases. Cardiovasc Res. 117:1434–1449. 2021. View Article : Google Scholar | |
Pallauf K, Günther I, Kühn G, Chin D, de Pascual-Teresa S and Rimbach G: The potential of resveratrol to act as a caloric restriction mimetic appears to be limited: Insights from studies in mice. Adv Nutr. 12:995–1005. 2021. View Article : Google Scholar : | |
Pernicova I and Korbonits M: Metformin-mode of action and clinical implications for diabetes and cancer. Nat Rev Endocrinol. 10:143–156. 2014. View Article : Google Scholar : PubMed/NCBI | |
Viollet B, Guigas B, Garcia NS, Leclerc J, Foretz M and Andreelli F: Cellular and molecular mechanisms of metformin: An overview. Clin Sci (Lond). 122:253–270. 2012. View Article : Google Scholar | |
Masoudi FA, Wang Y, Inzucchi SE, Setaro JF, Havranek EP, Foody JM and Krumholz HM: Metformin and thiazolidinedione use in medicare patients with heart failure. JAMA. 290:81–85. 2003. View Article : Google Scholar : PubMed/NCBI | |
Valencia WM, Palacio A, Tamariz L and Florez H: Metformin and ageing: Improving ageing outcomes beyond glycaemic control. Diabetologia. 60:1630–1638. 2017. View Article : Google Scholar : PubMed/NCBI | |
Morales DR and Morris AD: Metformin in cancer treatment and prevention. Annu Rev Med. 66:17–29. 2015. View Article : Google Scholar | |
Qin X, Du D, Chen Q, Wu M, Wu T, Wen J, Jin Y, Zhang J and Wang S: Metformin prevents murine ovarian aging. Aging (Albany NY). 11:3785–3794. 2019. View Article : Google Scholar : PubMed/NCBI | |
Huang CC, Chou CH, Yang YS, Ho HN, Shun CT, Wen WF, Chen SU and Chen MJ: Metformin: A novel promising option for fertility preservation during cyclophosphamide-based chemotherapy. Mol Hum Reprod. 27:gaaa0842021. View Article : Google Scholar : PubMed/NCBI | |
Cao Y, Wang Z, Zhang C, Bian Y, Zhang X, Liu X, Chen W and Zhao Y: Metformin promotes in vitro maturation of oocytes from aged mice by attenuating mitochondrial oxidative stress via SIRT3-dependent SOD2ac. Front Cell Dev Biol. 10:10285102022. View Article : Google Scholar : PubMed/NCBI | |
Ayhan S, Hancerliogullari N, Guney G, Gozukucuk M, Caydere M, Guney SS, Tokmak A and Ustun Y: Does the addition of metformin to carboplatin treatment decreases ovarian reserve damage associated with carboplatin usage? J Ovarian Res. 16:1842023. View Article : Google Scholar : PubMed/NCBI | |
Landry DA, Yakubovich E, Cook DP, Fasih S, Upham J and Vanderhyden BC: Metformin prevents age-associated ovarian fibrosis by modulating the immune landscape in female mice. Sci Adv. 8:eabq14752022. View Article : Google Scholar : PubMed/NCBI | |
Gambini J, Inglés M, Olaso G, Lopez-Grueso R, Bonet-Costa V, Gimeno-Mallench L, Mas-Bargues C, Abdelaziz KM, Gomez-Cabrera MC, Vina J and Borras C: Properties of resveratrol: In vitro and in vivo studies about metabolism, bioavailability, and biological effects in animal models and humans. Oxid Med Cell Longev. 2015:8370422015. View Article : Google Scholar : PubMed/NCBI | |
Rajman L, Chwalek K and Sinclair DA: Therapeutic potential of NAD-boosting molecules: The in vivo evidence. Cell Metabv. 27:529–547. 2018. View Article : Google Scholar | |
Côté CD, Rasmussen BA, Duca FA, Zadeh-Tahmasebi M, Baur JA, Daljeet M, Breen DM, Filippi BM and Lam TK: Resveratrol activates duodenal Sirt1 to reverse insulin resistance in rats through a neuronal network. Nat Med. 21:498–505. 2015. View Article : Google Scholar : PubMed/NCBI | |
Timmers S, Konings E, Bilet L, Houtkooper RH, van de Weijer T, Goossens GH, Hoeks J, van der Krieken S, Ryu D, Kersten S, et al: Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab. 14:612–622. 2011. View Article : Google Scholar : PubMed/NCBI | |
Battaglia R, Caponnetto A, Caringella AM, Cortone A, Ferrara C, Smirni S, Iannitti R, Purrello M, D'Amato G, Fioretti B and Di Pietro C: Resveratrol treatment induces mito-miRNome modification in follicular fluid from aged women with a poor prognosis for in vitro fertilization cycles. Antioxidants (Basel). 11:10192022. View Article : Google Scholar : PubMed/NCBI | |
Li R, Li E, Kamili G, Ou S and Yang D: Effect of resveratrol on superovulation in mice. Biomed Pharmacother. 146:1125652022. View Article : Google Scholar | |
Gou M, Li J, Yi L, Li H, Ye X, Wang H, Liu L, Sun B, Zhang S, Zhu Z, et al: Reprogramming of ovarian aging epigenome by resveratrol. PNAS Nexus. 2:pgac3102022. View Article : Google Scholar | |
Wu H, Xue J, Di H, Lv C, Hao Y and Nie Z: Resveratrol improves ovarian function in aged rat by inhibiting oxidative stress and activating the Sirt1. Gen Physiol Biophys. 41:53–61. 2022. View Article : Google Scholar : PubMed/NCBI | |
Zhou J, Xue Z, He HN, Liu X, Yin SY, Wu DY, Zhang X, Schatten H and Miao YL: Resveratrol delays postovulatory aging of mouse oocytes through activating mitophagy. Aging (Albany NY). 11:11504–11519. 2019. View Article : Google Scholar : PubMed/NCBI | |
Liang QX, Lin YH, Zhang CH, Sun HM, Zhou L, Schatten H, Sun QY and Qian WP: Resveratrol increases resistance of mouse oocytes to postovulatory aging in vivo. Aging (Albany NY). 10:1586–1596. 2018. View Article : Google Scholar : PubMed/NCBI | |
Liu MJ, Sun AG, Zhao SG, Liu H, Ma SY, Li M, Huai YX, Zhao H and Liu HB: Resveratrol improves in vitro maturation of oocytes in aged mice and humans. Fertil Steril. 109:900–907. 2018. View Article : Google Scholar : PubMed/NCBI | |
Okamoto N, Sato Y, Kawagoe Y, Shimizu T and Kawamura K: Short-term resveratrol treatment restored the quality of oocytes in aging mice. Aging (Albany NY). 14:5628–5640. 2022. View Article : Google Scholar : PubMed/NCBI | |
Sun YL, Tang SB, Shen W, Yin S and Sun QY: Roles of resveratrol in improving the quality of postovulatory aging oocytes in vitro. Cells. 8:11322019. View Article : Google Scholar : PubMed/NCBI | |
Cai M, Sun H, Huang Y, Yao H, Zhao C, Wang J and Zhu H: Resveratrol protects rat ovarian luteinized granulosa cells from H2O2 -induced dysfunction by activating autophagy. Int J Mol Sci. 24:109142023. View Article : Google Scholar | |
Moreira-Pinto B, Costa L, Felgueira E, Fonseca BM and Rebelo I: Low doses of resveratrol protect human granulosa cells from induced-oxidative stress. Antioxidants (Basel). 10:5612021. View Article : Google Scholar : PubMed/NCBI | |
Pruessner M, Hellhammer DH, Pruessner JC and Lupien SJ: Self-reported depressive symptoms and stress levels in healthy young men: Associations with the cortisol response to awakening. Psychosom Med. 65:92–99. 2003. View Article : Google Scholar : PubMed/NCBI | |
Campagne DM: Should fertilization treatment start with reducing stress? Hum Reprod. 21:1651–1658. 2006. View Article : Google Scholar : PubMed/NCBI | |
Obayashi K: Salivary mental stress proteins. Clin Chim Acta. 425:196–201. 2013. View Article : Google Scholar : PubMed/NCBI | |
Shah K, Kumari R and Jain M: Unveiling stress markers: A systematic review investigating psychological stress biomarkers. Dev Psychobiol. 66:e224902024. View Article : Google Scholar : PubMed/NCBI | |
Lavalle S, Masiello E, Iannella G, Magliulo G, Pace A, Lechien JR, Calvo-Henriquez C, Cocuzza S, Parisi FM, Favier V, et al: Unraveling the complexities of oxidative stress and inflammation biomarkers in obstructive sleep apnea syndrome: A comprehensive review. Life (Basel). 14:4252024.PubMed/NCBI | |
Ok J, Park S, Jung YH and Kim T: Wearable and implantable cortisol-sensing electronics for stress monitoring. Adv Mater. 36:22115952024. View Article : Google Scholar | |
Damone AL, Joham AE, Loxton D, Earnest A, Teede HJ and Moran LJ: Depression, anxiety and perceived stress in women with and without PCOS: A community-based study. Psychol Med. 49:1510–1520. 2019. View Article : Google Scholar | |
Tay CT, Teede HJ, Hill B, Loxton D and Joham AE: Increased prevalence of eating disorders, low self-esteem, and psychological distress in women with polycystic ovary syndrome: A community-based cohort study. Fertil Steril. 112:353–361. 2019. View Article : Google Scholar : PubMed/NCBI | |
Rowlands IJ, Teede H, Lucke J, Dobson AJ and Mishra GD: Young women's psychological distress after a diagnosis of polycystic ovary syndrome or endometriosis. Hum Reprod. 31:2072–2081. 2016. View Article : Google Scholar : PubMed/NCBI |