Skip to main content
Erschienen in: Journal of Assisted Reproduction and Genetics 6/2011

01.06.2011 | REVIEW

The impact of obesity on egg quality

verfasst von: Scott H. Purcell, Kelle H. Moley

Erschienen in: Journal of Assisted Reproduction and Genetics | Ausgabe 6/2011

Einloggen, um Zugang zu erhalten

Abstract

Obesity in women is a concern in many countries. This causes numerous health issues; however, this review focuses on the impact of obesity on women’s reproduction, and in particular the oocyte. Data from infertility clinics and experimental animal models that address the effects of obesity are presented. Bidirectional communication and metabolic support from the surrounding cumulus cells are critical for oocyte development, and the impact of obesity on these cells is also addressed. Both oocyte maturation and metabolism are impaired due to obesity, negatively impacting further development. In addition to reproductive hormones, obesity induced elevations in insulin, glucose, or free fatty acids, and changes in adipokines appear to impact the developmental competence of the oocyte. The data indicate that any one of these hormones or metabolites can impair oocyte developmental competence in vivo, and the combination of all of these factors and their interactions are the subject of ongoing investigations.
Literatur
1.
Zurück zum Zitat Vahratian A. Prevalence of overweight and obesity among women of childbearing age: results from the 2002 National Survey of Family Growth. Matern Child Health J. 2009;13:268–73.PubMedCrossRef Vahratian A. Prevalence of overweight and obesity among women of childbearing age: results from the 2002 National Survey of Family Growth. Matern Child Health J. 2009;13:268–73.PubMedCrossRef
2.
Zurück zum Zitat Flegal KM, Carroll MD, Ogden CL, Curtin LR. Prevalence and trends in obesity among US adults, 1999–2008. JAMA. 2010;303:235–41.PubMedCrossRef Flegal KM, Carroll MD, Ogden CL, Curtin LR. Prevalence and trends in obesity among US adults, 1999–2008. JAMA. 2010;303:235–41.PubMedCrossRef
3.
Zurück zum Zitat Gesink Law DC, Maclehose RF, Longnecker MP. Obesity and time to pregnancy. Hum Reprod. 2007;22:414–20.PubMedCrossRef Gesink Law DC, Maclehose RF, Longnecker MP. Obesity and time to pregnancy. Hum Reprod. 2007;22:414–20.PubMedCrossRef
4.
Zurück zum Zitat Rich-Edwards JW, Goldman MB, Willett WC, Hunter DJ, Stampfer MJ, Colditz GA, et al. Adolescent body mass index and infertility caused by ovulatory disorder. Am J Obstet Gynecol. 1994;171:171–7.PubMed Rich-Edwards JW, Goldman MB, Willett WC, Hunter DJ, Stampfer MJ, Colditz GA, et al. Adolescent body mass index and infertility caused by ovulatory disorder. Am J Obstet Gynecol. 1994;171:171–7.PubMed
5.
Zurück zum Zitat Jungheim ES, Moley KH. Current knowledge of obesity’s effects in the pre- and periconceptional periods and avenues for future research. Am J Obstet Gynecol. 2010;203:525–30.PubMedCrossRef Jungheim ES, Moley KH. Current knowledge of obesity’s effects in the pre- and periconceptional periods and avenues for future research. Am J Obstet Gynecol. 2010;203:525–30.PubMedCrossRef
6.
Zurück zum Zitat Wood JR, Dumesic DA, Abbott DH, Strauss 3rd JF. Molecular abnormalities in oocytes from women with polycystic ovary syndrome revealed by microarray analysis. J Clin Endocrinol Metab. 2007;92:705–13.PubMedCrossRef Wood JR, Dumesic DA, Abbott DH, Strauss 3rd JF. Molecular abnormalities in oocytes from women with polycystic ovary syndrome revealed by microarray analysis. J Clin Endocrinol Metab. 2007;92:705–13.PubMedCrossRef
7.
Zurück zum Zitat Dumesic DA, Abbott DH. Implications of polycystic ovary syndrome on oocyte development. Semin Reprod Med. 2008;26:53–61.PubMedCrossRef Dumesic DA, Abbott DH. Implications of polycystic ovary syndrome on oocyte development. Semin Reprod Med. 2008;26:53–61.PubMedCrossRef
8.
Zurück zum Zitat Norman RJ, Dewailly D, Legro RS, Hickey TE. Polycystic ovary syndrome. Lancet. 2007;370:685–97.PubMedCrossRef Norman RJ, Dewailly D, Legro RS, Hickey TE. Polycystic ovary syndrome. Lancet. 2007;370:685–97.PubMedCrossRef
9.
Zurück zum Zitat Cano F, Garcia-Velasco JA, Millet A, Remohi J, Simon C, Pellicer A. Oocyte quality in polycystic ovaries revisited: identification of a particular subgroup of women. J Assist Reprod Genet. 1997;14:254–61.PubMedCrossRef Cano F, Garcia-Velasco JA, Millet A, Remohi J, Simon C, Pellicer A. Oocyte quality in polycystic ovaries revisited: identification of a particular subgroup of women. J Assist Reprod Genet. 1997;14:254–61.PubMedCrossRef
10.
Zurück zum Zitat Jungheim ES, Lanzendorf SE, Odem RR, Moley KH, Chang AS, Ratts VS. Morbid obesity is associated with lower clinical pregnancy rates after in vitro fertilization in women with polycystic ovary syndrome. Fertil Steril. 2009;92:256–61.PubMedCrossRef Jungheim ES, Lanzendorf SE, Odem RR, Moley KH, Chang AS, Ratts VS. Morbid obesity is associated with lower clinical pregnancy rates after in vitro fertilization in women with polycystic ovary syndrome. Fertil Steril. 2009;92:256–61.PubMedCrossRef
11.
Zurück zum Zitat Barker DJ. Developmental origins of adult health and disease. J Epidemiol Community Health. 2004;58:114–5.PubMedCrossRef Barker DJ. Developmental origins of adult health and disease. J Epidemiol Community Health. 2004;58:114–5.PubMedCrossRef
12.
Zurück zum Zitat Bellver J, Melo MA, Bosch E, Serra V, Remohi J, Pellicer A. Obesity and poor reproductive outcome: the potential role of the endometrium. Fertil Steril. 2007;88:446–51.PubMedCrossRef Bellver J, Melo MA, Bosch E, Serra V, Remohi J, Pellicer A. Obesity and poor reproductive outcome: the potential role of the endometrium. Fertil Steril. 2007;88:446–51.PubMedCrossRef
13.
Zurück zum Zitat Bellver J, Rossal LP, Bosch E, Zuniga A, Corona JT, Melendez F, et al. Obesity and the risk of spontaneous abortion after oocyte donation. Fertil Steril. 2003;79:1136–40.PubMedCrossRef Bellver J, Rossal LP, Bosch E, Zuniga A, Corona JT, Melendez F, et al. Obesity and the risk of spontaneous abortion after oocyte donation. Fertil Steril. 2003;79:1136–40.PubMedCrossRef
14.
Zurück zum Zitat DeUgarte DA, DeUgarte CM, Sahakian V. Surrogate obesity negatively impacts pregnancy rates in third-party reproduction. Fertil Steril. 2010;93:1008–10.PubMedCrossRef DeUgarte DA, DeUgarte CM, Sahakian V. Surrogate obesity negatively impacts pregnancy rates in third-party reproduction. Fertil Steril. 2010;93:1008–10.PubMedCrossRef
15.
Zurück zum Zitat Dessolle L, Darai E, Cornet D, Rouzier R, Coutant C, Mandelbaum J, et al. Determinants of pregnancy rate in the donor oocyte model: a multivariate analysis of 450 frozen-thawed embryo transfers. Hum Reprod. 2009;24:3082–9.PubMedCrossRef Dessolle L, Darai E, Cornet D, Rouzier R, Coutant C, Mandelbaum J, et al. Determinants of pregnancy rate in the donor oocyte model: a multivariate analysis of 450 frozen-thawed embryo transfers. Hum Reprod. 2009;24:3082–9.PubMedCrossRef
16.
Zurück zum Zitat Chavarro JE, Toth TL, Wright DL, Meeker JD, Hauser R. Body mass index in relation to semen quality, sperm DNA integrity, and serum reproductive hormone levels among men attending an infertility clinic. Fertil Steril. 2010;93:2222–31.PubMedCrossRef Chavarro JE, Toth TL, Wright DL, Meeker JD, Hauser R. Body mass index in relation to semen quality, sperm DNA integrity, and serum reproductive hormone levels among men attending an infertility clinic. Fertil Steril. 2010;93:2222–31.PubMedCrossRef
17.
Zurück zum Zitat Bakos HW, Henshaw RC, Mitchell M, Lane M. Paternal body mass index is associated with decreased blastocyst development and reduced live birth rates following assisted reproductive technology. Fertil Steril. 2010;95:1700–4. Bakos HW, Henshaw RC, Mitchell M, Lane M. Paternal body mass index is associated with decreased blastocyst development and reduced live birth rates following assisted reproductive technology. Fertil Steril. 2010;95:1700–4.
19.
Zurück zum Zitat Mitchell M, Bakos HW, Lane M. Paternal diet-induced obesity impairs embryo development and implantation in the mouse. Fertil Steril. 2011;95:1349–53. Mitchell M, Bakos HW, Lane M. Paternal diet-induced obesity impairs embryo development and implantation in the mouse. Fertil Steril. 2011;95:1349–53.
20.
Zurück zum Zitat Tamer Erel C, Senturk LM. The impact of body mass index on assisted reproduction. Curr Opin Obstet Gynecol. 2009;21:228–35.PubMedCrossRef Tamer Erel C, Senturk LM. The impact of body mass index on assisted reproduction. Curr Opin Obstet Gynecol. 2009;21:228–35.PubMedCrossRef
21.
Zurück zum Zitat Brewer CJ, Balen AH. The adverse effects of obesity on conception and implantation. Reproduction. 2010;140:347–64.PubMedCrossRef Brewer CJ, Balen AH. The adverse effects of obesity on conception and implantation. Reproduction. 2010;140:347–64.PubMedCrossRef
22.
Zurück zum Zitat Robker RL. Evidence that obesity alters the quality of oocytes and embryos. Pathophysiology. 2008;15:115–21.PubMedCrossRef Robker RL. Evidence that obesity alters the quality of oocytes and embryos. Pathophysiology. 2008;15:115–21.PubMedCrossRef
23.
Zurück zum Zitat Luke B, Brown MB, Stern JE, Missmer SA, Fujimoto VY, Leach R. Female obesity adversely affects assisted reproductive technology (ART) pregnancy and live birth rates. Hum Reprod. 2011;26:245–52.PubMedCrossRef Luke B, Brown MB, Stern JE, Missmer SA, Fujimoto VY, Leach R. Female obesity adversely affects assisted reproductive technology (ART) pregnancy and live birth rates. Hum Reprod. 2011;26:245–52.PubMedCrossRef
24.
Zurück zum Zitat Ramlau-Hansen CH, Thulstrup AM, Nohr EA, Bonde JP, Sorensen TI, Olsen J. Subfecundity in overweight and obese couples. Hum Reprod. 2007;22:1634–7.PubMedCrossRef Ramlau-Hansen CH, Thulstrup AM, Nohr EA, Bonde JP, Sorensen TI, Olsen J. Subfecundity in overweight and obese couples. Hum Reprod. 2007;22:1634–7.PubMedCrossRef
25.
Zurück zum Zitat Wise LA, Rothman KJ, Mikkelsen EM, Sorensen HT, Riis A, Hatch EE. An internet-based prospective study of body size and time-to-pregnancy. Hum Reprod. 2010;25:253–64.PubMedCrossRef Wise LA, Rothman KJ, Mikkelsen EM, Sorensen HT, Riis A, Hatch EE. An internet-based prospective study of body size and time-to-pregnancy. Hum Reprod. 2010;25:253–64.PubMedCrossRef
26.
Zurück zum Zitat Gilchrist RB, Ritter LJ, Armstrong DT. Oocyte-somatic cell interactions during follicle development in mammals. Anim Reprod Sci. 2004;82–83:431–46.PubMedCrossRef Gilchrist RB, Ritter LJ, Armstrong DT. Oocyte-somatic cell interactions during follicle development in mammals. Anim Reprod Sci. 2004;82–83:431–46.PubMedCrossRef
27.
Zurück zum Zitat Albertini DF, Combelles CM, Benecchi E, Carabatsos MJ. Cellular basis for paracrine regulation of ovarian follicle development. Reproduction. 2001;121:647–53.PubMedCrossRef Albertini DF, Combelles CM, Benecchi E, Carabatsos MJ. Cellular basis for paracrine regulation of ovarian follicle development. Reproduction. 2001;121:647–53.PubMedCrossRef
28.
Zurück zum Zitat Matzuk MM, Burns KH, Viveiros MM, Eppig JJ. Intercellular communication in the mammalian ovary: oocytes carry the conversation. Science. 2002;296:2178–80.PubMedCrossRef Matzuk MM, Burns KH, Viveiros MM, Eppig JJ. Intercellular communication in the mammalian ovary: oocytes carry the conversation. Science. 2002;296:2178–80.PubMedCrossRef
29.
Zurück zum Zitat Biggers JD, Whittingham DG, Donahue RP. The pattern of energy metabolism in the mouse oocyte and zygote. Proc Natl Acad Sci USA. 1967;58:560–7.PubMedCrossRef Biggers JD, Whittingham DG, Donahue RP. The pattern of energy metabolism in the mouse oocyte and zygote. Proc Natl Acad Sci USA. 1967;58:560–7.PubMedCrossRef
30.
Zurück zum Zitat Leese HJ, Barton AM. Pyruvate and glucose uptake by mouse ova and preimplantation embryos. J Reprod Fertil. 1984;72:9–13.PubMedCrossRef Leese HJ, Barton AM. Pyruvate and glucose uptake by mouse ova and preimplantation embryos. J Reprod Fertil. 1984;72:9–13.PubMedCrossRef
31.
Zurück zum Zitat Sutton-McDowall ML, Gilchrist RB, Thompson JG. The pivotal role of glucose metabolism in determining oocyte developmental competence. Reproduction. 2010;139:685–95.PubMedCrossRef Sutton-McDowall ML, Gilchrist RB, Thompson JG. The pivotal role of glucose metabolism in determining oocyte developmental competence. Reproduction. 2010;139:685–95.PubMedCrossRef
32.
Zurück zum Zitat Farhi J, Ben-Haroush A, Sapir O, Fisch B, Ashkenazi J. High-quality embryos retain their implantation capability in overweight women. Reprod Biomed Online. 2010;21:706–11.PubMedCrossRef Farhi J, Ben-Haroush A, Sapir O, Fisch B, Ashkenazi J. High-quality embryos retain their implantation capability in overweight women. Reprod Biomed Online. 2010;21:706–11.PubMedCrossRef
33.
Zurück zum Zitat Robker RL, Akison LK, Bennett BD, Thrupp PN, Chura LR, Russell DL, et al. Obese women exhibit differences in ovarian metabolites, hormones, and gene expression compared with moderate-weight women. J Clin Endocrinol Metab. 2009;94:1533–40.PubMedCrossRef Robker RL, Akison LK, Bennett BD, Thrupp PN, Chura LR, Russell DL, et al. Obese women exhibit differences in ovarian metabolites, hormones, and gene expression compared with moderate-weight women. J Clin Endocrinol Metab. 2009;94:1533–40.PubMedCrossRef
34.
Zurück zum Zitat Poretsky L, Cataldo NA, Rosenwaks Z, Giudice LC. The insulin-related ovarian regulatory system in health and disease. Endocr Rev. 1999;20:535–82.PubMedCrossRef Poretsky L, Cataldo NA, Rosenwaks Z, Giudice LC. The insulin-related ovarian regulatory system in health and disease. Endocr Rev. 1999;20:535–82.PubMedCrossRef
35.
Zurück zum Zitat Jain A, Polotsky AJ, Rochester D, Berga SL, Loucks T, Zeitlian G, et al. Pulsatile luteinizing hormone amplitude and progesterone metabolite excretion are reduced in obese women. J Clin Endocrinol Metab. 2007;92:2468–73.PubMedCrossRef Jain A, Polotsky AJ, Rochester D, Berga SL, Loucks T, Zeitlian G, et al. Pulsatile luteinizing hormone amplitude and progesterone metabolite excretion are reduced in obese women. J Clin Endocrinol Metab. 2007;92:2468–73.PubMedCrossRef
36.
Zurück zum Zitat Diamond MP, Moley KH, Pellicer A, Vaughn WK, DeCherney AH. Effects of streptozotocin- and alloxan-induced diabetes mellitus on mouse follicular and early embryo development. J Reprod Fertil. 1989;86:1–10.PubMedCrossRef Diamond MP, Moley KH, Pellicer A, Vaughn WK, DeCherney AH. Effects of streptozotocin- and alloxan-induced diabetes mellitus on mouse follicular and early embryo development. J Reprod Fertil. 1989;86:1–10.PubMedCrossRef
37.
Zurück zum Zitat Chang AS, Dale AN, Moley KH. Maternal diabetes adversely affects preovulatory oocyte maturation, development, and granulosa cell apoptosis. Endocrinology. 2005;146:2445–53.PubMedCrossRef Chang AS, Dale AN, Moley KH. Maternal diabetes adversely affects preovulatory oocyte maturation, development, and granulosa cell apoptosis. Endocrinology. 2005;146:2445–53.PubMedCrossRef
38.
Zurück zum Zitat Colton SA, Pieper GM, Downs SM. Altered meiotic regulation in oocytes from diabetic mice. Biol Reprod. 2002;67:220–31.PubMedCrossRef Colton SA, Pieper GM, Downs SM. Altered meiotic regulation in oocytes from diabetic mice. Biol Reprod. 2002;67:220–31.PubMedCrossRef
39.
Zurück zum Zitat Ratchford AM, Esguerra CR, Moley KH. Decreased oocyte-granulosa cell gap junction communication and connexin expression in a type 1 diabetic mouse model. Mol Endocrinol. 2008;22:2643–54.PubMedCrossRef Ratchford AM, Esguerra CR, Moley KH. Decreased oocyte-granulosa cell gap junction communication and connexin expression in a type 1 diabetic mouse model. Mol Endocrinol. 2008;22:2643–54.PubMedCrossRef
40.
Zurück zum Zitat Jungheim ES, Schoeller EL, Marquard KL, Louden ED, Schaffer JE, Moley KH. Diet-induced obesity model: abnormal oocytes and persistent growth abnormalities in the offspring. Endocrinology. 2010;151:4039–46.PubMedCrossRef Jungheim ES, Schoeller EL, Marquard KL, Louden ED, Schaffer JE, Moley KH. Diet-induced obesity model: abnormal oocytes and persistent growth abnormalities in the offspring. Endocrinology. 2010;151:4039–46.PubMedCrossRef
41.
Zurück zum Zitat Wittemer C, Ohl J, Bailly M, Bettahar-Lebugle K, Nisand I. Does body mass index of infertile women have an impact on IVF procedure and outcome? J Assist Reprod Genet. 2000;17:547–52.PubMedCrossRef Wittemer C, Ohl J, Bailly M, Bettahar-Lebugle K, Nisand I. Does body mass index of infertile women have an impact on IVF procedure and outcome? J Assist Reprod Genet. 2000;17:547–52.PubMedCrossRef
42.
Zurück zum Zitat Dokras A, Baredziak L, Blaine J, Syrop C, VanVoorhis BJ, Sparks A. Obstetric outcomes after in vitro fertilization in obese and morbidly obese women. Obstet Gynecol. 2006;108:61–9.PubMedCrossRef Dokras A, Baredziak L, Blaine J, Syrop C, VanVoorhis BJ, Sparks A. Obstetric outcomes after in vitro fertilization in obese and morbidly obese women. Obstet Gynecol. 2006;108:61–9.PubMedCrossRef
43.
Zurück zum Zitat Carrell DT, Jones KP, Peterson CM, Aoki V, Emery BR, Campbell BR. Body mass index is inversely related to intrafollicular HCG concentrations, embryo quality and IVF outcome. Reprod Biomed Online. 2001;3:109–11.PubMedCrossRef Carrell DT, Jones KP, Peterson CM, Aoki V, Emery BR, Campbell BR. Body mass index is inversely related to intrafollicular HCG concentrations, embryo quality and IVF outcome. Reprod Biomed Online. 2001;3:109–11.PubMedCrossRef
44.
Zurück zum Zitat Metwally M, Cutting R, Tipton A, Skull J, Ledger WL, Li TC. Effect of increased body mass index on oocyte and embryo quality in IVF patients. Reprod Biomed Online. 2007;15:532–8.PubMedCrossRef Metwally M, Cutting R, Tipton A, Skull J, Ledger WL, Li TC. Effect of increased body mass index on oocyte and embryo quality in IVF patients. Reprod Biomed Online. 2007;15:532–8.PubMedCrossRef
45.
Zurück zum Zitat Esinler I, Bozdag G, Yarali H. Impact of isolated obesity on ICSI outcome. Reprod Biomed Online. 2008;17:583–7.PubMedCrossRef Esinler I, Bozdag G, Yarali H. Impact of isolated obesity on ICSI outcome. Reprod Biomed Online. 2008;17:583–7.PubMedCrossRef
46.
Zurück zum Zitat Sagle M, Bishop K, Ridley N, Alexander FM, Michel M, Bonney RC, et al. Recurrent early miscarriage and polycystic ovaries. BMJ. 1988;297:1027–8.PubMedCrossRef Sagle M, Bishop K, Ridley N, Alexander FM, Michel M, Bonney RC, et al. Recurrent early miscarriage and polycystic ovaries. BMJ. 1988;297:1027–8.PubMedCrossRef
47.
Zurück zum Zitat Carmina E, Lobo RA. Polycystic ovary syndrome (PCOS): arguably the most common endocrinopathy is associated with significant morbidity in women. J Clin Endocrinol Metab. 1999;84:1897–9.PubMedCrossRef Carmina E, Lobo RA. Polycystic ovary syndrome (PCOS): arguably the most common endocrinopathy is associated with significant morbidity in women. J Clin Endocrinol Metab. 1999;84:1897–9.PubMedCrossRef
48.
Zurück zum Zitat Marquard KL, Stephens SM, Jungheim ES, Ratts VS, Odem RR, Lanzendorf S, et al. Polycystic ovary syndrome and maternal obesity affect oocyte size in in vitro fertilization/intracytoplasmic sperm injection cycles. Fertil Steril. 2011;95:2146–9. Marquard KL, Stephens SM, Jungheim ES, Ratts VS, Odem RR, Lanzendorf S, et al. Polycystic ovary syndrome and maternal obesity affect oocyte size in in vitro fertilization/intracytoplasmic sperm injection cycles. Fertil Steril. 2011;95:2146–9.
49.
Zurück zum Zitat Sengoku K, Tamate K, Takuma N, Yoshida T, Goishi K, Ishikawa M. The chromosomal normality of unfertilized oocytes from patients with polycystic ovarian syndrome. Hum Reprod. 1997;12:474–7.PubMedCrossRef Sengoku K, Tamate K, Takuma N, Yoshida T, Goishi K, Ishikawa M. The chromosomal normality of unfertilized oocytes from patients with polycystic ovarian syndrome. Hum Reprod. 1997;12:474–7.PubMedCrossRef
50.
Zurück zum Zitat Heijnen EM, Eijkemans MJ, Hughes EG, Laven JS, Macklon NS, Fauser BC. A meta-analysis of outcomes of conventional IVF in women with polycystic ovary syndrome. Hum Reprod Update. 2006;12:13–21.PubMedCrossRef Heijnen EM, Eijkemans MJ, Hughes EG, Laven JS, Macklon NS, Fauser BC. A meta-analysis of outcomes of conventional IVF in women with polycystic ovary syndrome. Hum Reprod Update. 2006;12:13–21.PubMedCrossRef
51.
Zurück zum Zitat Duggal PS, Ryan NK, Van der Hoek KH, Ritter LJ, Armstrong DT, Magoffin DA, et al. Effects of leptin administration and feed restriction on thecal leucocytes in the preovulatory rat ovary and the effects of leptin on meiotic maturation, granulosa cell proliferation, steroid hormone and PGE2 release in cultured rat ovarian follicles. Reproduction. 2002;123:891–8.PubMedCrossRef Duggal PS, Ryan NK, Van der Hoek KH, Ritter LJ, Armstrong DT, Magoffin DA, et al. Effects of leptin administration and feed restriction on thecal leucocytes in the preovulatory rat ovary and the effects of leptin on meiotic maturation, granulosa cell proliferation, steroid hormone and PGE2 release in cultured rat ovarian follicles. Reproduction. 2002;123:891–8.PubMedCrossRef
52.
Zurück zum Zitat Cioffi JA, Van Blerkom J, Antczak M, Shafer A, Wittmer S, Snodgrass HR. The expression of leptin and its receptors in pre-ovulatory human follicles. Mol Hum Reprod. 1997;3:467–72.PubMedCrossRef Cioffi JA, Van Blerkom J, Antczak M, Shafer A, Wittmer S, Snodgrass HR. The expression of leptin and its receptors in pre-ovulatory human follicles. Mol Hum Reprod. 1997;3:467–72.PubMedCrossRef
53.
Zurück zum Zitat Duggal PS, Van Der Hoek KH, Milner CR, Ryan NK, Armstrong DT, Magoffin DA, et al. The in vivo and in vitro effects of exogenous leptin on ovulation in the rat. Endocrinology. 2000;141:1971–6.PubMedCrossRef Duggal PS, Van Der Hoek KH, Milner CR, Ryan NK, Armstrong DT, Magoffin DA, et al. The in vivo and in vitro effects of exogenous leptin on ovulation in the rat. Endocrinology. 2000;141:1971–6.PubMedCrossRef
54.
Zurück zum Zitat Swain JE, Dunn RL, McConnell D, Gonzalez-Martinez J, Smith GD. Direct effects of leptin on mouse reproductive function: regulation of follicular, oocyte, and embryo development. Biol Reprod. 2004;71:1446–52.PubMedCrossRef Swain JE, Dunn RL, McConnell D, Gonzalez-Martinez J, Smith GD. Direct effects of leptin on mouse reproductive function: regulation of follicular, oocyte, and embryo development. Biol Reprod. 2004;71:1446–52.PubMedCrossRef
55.
Zurück zum Zitat Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372:425–32.PubMedCrossRef Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994;372:425–32.PubMedCrossRef
56.
Zurück zum Zitat Pallares P, Garcia-Fernandez RA, Criado LM, Letelier CA, Fernandez-Toro JM, Esteban D, et al. Substantiation of ovarian effects of leptin by challenging a mouse model of obesity/type 2 diabetes. Theriogenology. 2010;73:1088–95.PubMedCrossRef Pallares P, Garcia-Fernandez RA, Criado LM, Letelier CA, Fernandez-Toro JM, Esteban D, et al. Substantiation of ovarian effects of leptin by challenging a mouse model of obesity/type 2 diabetes. Theriogenology. 2010;73:1088–95.PubMedCrossRef
57.
Zurück zum Zitat Arias-Alvarez M, Bermejo-Alvarez P, Gutierrez-Adan A, Rizos D, Lorenzo PL, Lonergan P. Effect of leptin supplementation during in vitro oocyte maturation and embryo culture on bovine embryo development and gene expression patterns. Theriogenology. 2010;75:887–96. Arias-Alvarez M, Bermejo-Alvarez P, Gutierrez-Adan A, Rizos D, Lorenzo PL, Lonergan P. Effect of leptin supplementation during in vitro oocyte maturation and embryo culture on bovine embryo development and gene expression patterns. Theriogenology. 2010;75:887–96.
58.
Zurück zum Zitat Hill MJ, Uyehara CF, Hashiro GM, Frattarelli JL. The utility of serum leptin and follicular fluid leptin, estradiol, and progesterone levels during an in vitro fertilization cycle. J Assist Reprod Genet. 2007;24:183–8.PubMedCrossRef Hill MJ, Uyehara CF, Hashiro GM, Frattarelli JL. The utility of serum leptin and follicular fluid leptin, estradiol, and progesterone levels during an in vitro fertilization cycle. J Assist Reprod Genet. 2007;24:183–8.PubMedCrossRef
59.
Zurück zum Zitat Lin Q, Poon SL, Chen J, Cheng L, HoYuen B, Leung PC. Leptin interferes with 3′,5′-cyclic adenosine monophosphate (cAMP) signaling to inhibit steroidogenesis in human granulosa cells. Reprod Biol Endocrinol. 2009;7:115.PubMedCrossRef Lin Q, Poon SL, Chen J, Cheng L, HoYuen B, Leung PC. Leptin interferes with 3′,5′-cyclic adenosine monophosphate (cAMP) signaling to inhibit steroidogenesis in human granulosa cells. Reprod Biol Endocrinol. 2009;7:115.PubMedCrossRef
60.
Zurück zum Zitat Brannian JD, Hansen KA. Leptin and ovarian folliculogenesis: implications for ovulation induction and ART outcomes. Semin Reprod Med. 2002;20:103–12.PubMedCrossRef Brannian JD, Hansen KA. Leptin and ovarian folliculogenesis: implications for ovulation induction and ART outcomes. Semin Reprod Med. 2002;20:103–12.PubMedCrossRef
61.
Zurück zum Zitat Pierre P, Froment P, Negre D, Rame C, Barateau V, Chabrolle C, et al. Role of adiponectin receptors, AdipoR1 and AdipoR2, in the steroidogenesis of the human granulosa tumor cell line, KGN. Hum Reprod. 2009;24:2890–901.PubMedCrossRef Pierre P, Froment P, Negre D, Rame C, Barateau V, Chabrolle C, et al. Role of adiponectin receptors, AdipoR1 and AdipoR2, in the steroidogenesis of the human granulosa tumor cell line, KGN. Hum Reprod. 2009;24:2890–901.PubMedCrossRef
62.
Zurück zum Zitat Fleming TP, Kwong WY, Porter R, Ursell E, Fesenko I, Wilkins A, et al. The embryo and its future. Biol Reprod. 2004;71:1046–54.PubMedCrossRef Fleming TP, Kwong WY, Porter R, Ursell E, Fesenko I, Wilkins A, et al. The embryo and its future. Biol Reprod. 2004;71:1046–54.PubMedCrossRef
63.
Zurück zum Zitat Leese HJ, Baumann CG, Brison DR, McEvoy TG, Sturmey RG. Metabolism of the viable mammalian embryo: quietness revisited. Mol Hum Reprod. 2008;14:667–72.PubMedCrossRef Leese HJ, Baumann CG, Brison DR, McEvoy TG, Sturmey RG. Metabolism of the viable mammalian embryo: quietness revisited. Mol Hum Reprod. 2008;14:667–72.PubMedCrossRef
64.
Zurück zum Zitat Leese HJ. Quiet please, do not disturb: a hypothesis of embryo metabolism and viability. Bioessays. 2002;24:845–9.PubMedCrossRef Leese HJ. Quiet please, do not disturb: a hypothesis of embryo metabolism and viability. Bioessays. 2002;24:845–9.PubMedCrossRef
65.
Zurück zum Zitat Downs SM, Mosey JL, Klinger J. Fatty acid oxidation and meiotic resumption in mouse oocytes. Mol Reprod Dev. 2009;76:844–53.PubMedCrossRef Downs SM, Mosey JL, Klinger J. Fatty acid oxidation and meiotic resumption in mouse oocytes. Mol Reprod Dev. 2009;76:844–53.PubMedCrossRef
66.
Zurück zum Zitat Dunning KR, Cashman K, Russell DL, Thompson JG, Norman RJ, Robker RL. Beta-oxidation is essential for mouse oocyte developmental competence and early embryo development. Biol Reprod. 2010;83:909–18.PubMedCrossRef Dunning KR, Cashman K, Russell DL, Thompson JG, Norman RJ, Robker RL. Beta-oxidation is essential for mouse oocyte developmental competence and early embryo development. Biol Reprod. 2010;83:909–18.PubMedCrossRef
67.
Zurück zum Zitat Yang X, Dunning KR, Wu LL, Hickey TE, Norman RJ, Russell DL, et al. Identification of perilipin-2 as a lipid droplet protein regulated in oocytes during maturation. Reprod Fertil Dev. 2010;22:1262–71.PubMedCrossRef Yang X, Dunning KR, Wu LL, Hickey TE, Norman RJ, Russell DL, et al. Identification of perilipin-2 as a lipid droplet protein regulated in oocytes during maturation. Reprod Fertil Dev. 2010;22:1262–71.PubMedCrossRef
68.
Zurück zum Zitat McEvoy TG, Coull GD, Broadbent PJ, Hutchinson JS, Speake BK. Fatty acid composition of lipids in immature cattle, pig and sheep oocytes with intact zona pellucida. J Reprod Fertil. 2000;118:163–70.PubMedCrossRef McEvoy TG, Coull GD, Broadbent PJ, Hutchinson JS, Speake BK. Fatty acid composition of lipids in immature cattle, pig and sheep oocytes with intact zona pellucida. J Reprod Fertil. 2000;118:163–70.PubMedCrossRef
69.
Zurück zum Zitat Jungheim ES, Macones GA, Odem RR, Patterson BW, Lanzendorf SE, Ratts VS, et al. Associations between free fatty acids, cumulus oocyte complex morphology and ovarian function during in vitro fertilization. Fertil Steril. 2011;95:1970–4. Jungheim ES, Macones GA, Odem RR, Patterson BW, Lanzendorf SE, Ratts VS, et al. Associations between free fatty acids, cumulus oocyte complex morphology and ovarian function during in vitro fertilization. Fertil Steril. 2011;95:1970–4.
70.
Zurück zum Zitat Wu LL, Dunning KR, Yang X, Russell DL, Lane M, Norman RJ, et al. High-fat diet causes lipotoxicity responses in cumulus-oocyte complexes and decreased fertilization rates. Endocrinology. 2010;151:5438–45.PubMedCrossRef Wu LL, Dunning KR, Yang X, Russell DL, Lane M, Norman RJ, et al. High-fat diet causes lipotoxicity responses in cumulus-oocyte complexes and decreased fertilization rates. Endocrinology. 2010;151:5438–45.PubMedCrossRef
71.
Zurück zum Zitat Borradaile NM, Han X, Harp JD, Gale SE, Ory DS, Schaffer JE. Disruption of endoplasmic reticulum structure and integrity in lipotoxic cell death. J Lipid Res. 2006;47:2726–37.PubMedCrossRef Borradaile NM, Han X, Harp JD, Gale SE, Ory DS, Schaffer JE. Disruption of endoplasmic reticulum structure and integrity in lipotoxic cell death. J Lipid Res. 2006;47:2726–37.PubMedCrossRef
72.
Zurück zum Zitat Igosheva N, Abramov AY, Poston L, Eckert JJ, Fleming TP, Duchen MR, et al. Maternal diet-induced obesity alters mitochondrial activity and redox status in mouse oocytes and zygotes. PLoS ONE. 2010;5:e10074.PubMedCrossRef Igosheva N, Abramov AY, Poston L, Eckert JJ, Fleming TP, Duchen MR, et al. Maternal diet-induced obesity alters mitochondrial activity and redox status in mouse oocytes and zygotes. PLoS ONE. 2010;5:e10074.PubMedCrossRef
73.
Zurück zum Zitat Harris SE, Leese HJ, Gosden RG, Picton HM. Pyruvate and oxygen consumption throughout the growth and development of murine oocytes. Mol Reprod Dev. 2009;76:231–8.PubMedCrossRef Harris SE, Leese HJ, Gosden RG, Picton HM. Pyruvate and oxygen consumption throughout the growth and development of murine oocytes. Mol Reprod Dev. 2009;76:231–8.PubMedCrossRef
74.
Zurück zum Zitat Harris SE, Adriaens I, Leese HJ, Gosden RG, Picton HM. Carbohydrate metabolism by murine ovarian follicles and oocytes grown in vitro. Reproduction. 2007;134:415–24.PubMedCrossRef Harris SE, Adriaens I, Leese HJ, Gosden RG, Picton HM. Carbohydrate metabolism by murine ovarian follicles and oocytes grown in vitro. Reproduction. 2007;134:415–24.PubMedCrossRef
75.
Zurück zum Zitat Cetica P, Pintos L, Dalvit G, Beconi M. Activity of key enzymes involved in glucose and triglyceride catabolism during bovine oocyte maturation in vitro. Reproduction. 2002;124:675–81.PubMedCrossRef Cetica P, Pintos L, Dalvit G, Beconi M. Activity of key enzymes involved in glucose and triglyceride catabolism during bovine oocyte maturation in vitro. Reproduction. 2002;124:675–81.PubMedCrossRef
76.
Zurück zum Zitat Hardy K, Hooper MA, Handyside AH, Rutherford AJ, Winston RM, Leese HJ. Non-invasive measurement of glucose and pyruvate uptake by individual human oocytes and preimplantation embryos. Hum Reprod. 1989;4:188–91.PubMed Hardy K, Hooper MA, Handyside AH, Rutherford AJ, Winston RM, Leese HJ. Non-invasive measurement of glucose and pyruvate uptake by individual human oocytes and preimplantation embryos. Hum Reprod. 1989;4:188–91.PubMed
77.
Zurück zum Zitat Downs SM, Humpherson PG, Martin KL, Leese HJ. Glucose utilization during gonadotropin-induced meiotic maturation in cumulus cell-enclosed mouse oocytes. Mol Reprod Dev. 1996;44:121–31.PubMedCrossRef Downs SM, Humpherson PG, Martin KL, Leese HJ. Glucose utilization during gonadotropin-induced meiotic maturation in cumulus cell-enclosed mouse oocytes. Mol Reprod Dev. 1996;44:121–31.PubMedCrossRef
78.
Zurück zum Zitat Downs SM, Gilles R, Vanderhoef C, Humpherson PG, Leese HJ. Differential response of cumulus cell-enclosed and denuded mouse oocytes in a meiotic induction model system. Mol Reprod Dev. 2006;73:379–89.PubMedCrossRef Downs SM, Gilles R, Vanderhoef C, Humpherson PG, Leese HJ. Differential response of cumulus cell-enclosed and denuded mouse oocytes in a meiotic induction model system. Mol Reprod Dev. 2006;73:379–89.PubMedCrossRef
79.
Zurück zum Zitat Sutton ML, Gilchrist RB, Thompson JG. Effects of in-vivo and in-vitro environments on the metabolism of the cumulus-oocyte complex and its influence on oocyte developmental capacity. Hum Reprod Update. 2003;9:35–48.PubMedCrossRef Sutton ML, Gilchrist RB, Thompson JG. Effects of in-vivo and in-vitro environments on the metabolism of the cumulus-oocyte complex and its influence on oocyte developmental capacity. Hum Reprod Update. 2003;9:35–48.PubMedCrossRef
80.
Zurück zum Zitat Joost HG, Bell GI, Best JD, Birnbaum MJ, Charron MJ, Chen YT, et al. Nomenclature of the GLUT/SLC2A family of sugar/polyol transport facilitators. Am J Physiol Endocrinol Metab. 2002;282:E974–6.PubMed Joost HG, Bell GI, Best JD, Birnbaum MJ, Charron MJ, Chen YT, et al. Nomenclature of the GLUT/SLC2A family of sugar/polyol transport facilitators. Am J Physiol Endocrinol Metab. 2002;282:E974–6.PubMed
81.
Zurück zum Zitat Wu X, Freeze HH. GLUT14, a duplicon of GLUT3, is specifically expressed in testis as alternative splice forms. Genomics. 2002;80:553–7.PubMedCrossRef Wu X, Freeze HH. GLUT14, a duplicon of GLUT3, is specifically expressed in testis as alternative splice forms. Genomics. 2002;80:553–7.PubMedCrossRef
82.
Zurück zum Zitat Heyner S, Smith RM, Schultz GA. Temporally regulated expression of insulin and insulin-like growth factors and their receptors in early mammalian development. Bioessays. 1989;11:171–6.PubMedCrossRef Heyner S, Smith RM, Schultz GA. Temporally regulated expression of insulin and insulin-like growth factors and their receptors in early mammalian development. Bioessays. 1989;11:171–6.PubMedCrossRef
83.
Zurück zum Zitat Carayannopoulos MO, Chi MM, Cui Y, Pingsterhaus JM, McKnight RA, Mueckler M, et al. GLUT8 is a glucose transporter responsible for insulin-stimulated glucose uptake in the blastocyst. Proc Natl Acad Sci USA. 2000;97:7313–8.PubMedCrossRef Carayannopoulos MO, Chi MM, Cui Y, Pingsterhaus JM, McKnight RA, Mueckler M, et al. GLUT8 is a glucose transporter responsible for insulin-stimulated glucose uptake in the blastocyst. Proc Natl Acad Sci USA. 2000;97:7313–8.PubMedCrossRef
84.
Zurück zum Zitat Purcell SH, Aerni-Flessner LB, Willcockson AR, Diggs-Andrews KA, Fisher SJ, Moley KH. Improved Insulin sensitivity by GLUT12 overexpression in mice. Diabetes. 2011;60:1478–82. Purcell SH, Aerni-Flessner LB, Willcockson AR, Diggs-Andrews KA, Fisher SJ, Moley KH. Improved Insulin sensitivity by GLUT12 overexpression in mice. Diabetes. 2011;60:1478–82.
85.
Zurück zum Zitat Stuart CA, Howell ME, Zhang Y, Yin D. Insulin-stimulated translocation of glucose transporter (GLUT) 12 parallels that of GLUT4 in normal muscle. J Clin Endocrinol Metab. 2009;94:3535–42.PubMedCrossRef Stuart CA, Howell ME, Zhang Y, Yin D. Insulin-stimulated translocation of glucose transporter (GLUT) 12 parallels that of GLUT4 in normal muscle. J Clin Endocrinol Metab. 2009;94:3535–42.PubMedCrossRef
86.
Zurück zum Zitat Roberts R, Stark J, Iatropoulou A, Becker DL, Franks S, Hardy K. Energy substrate metabolism of mouse cumulus-oocyte complexes: response to follicle-stimulating hormone is mediated by the phosphatidylinositol 3-kinase pathway and is associated with oocyte maturation. Biol Reprod. 2004;71:199–209.PubMedCrossRef Roberts R, Stark J, Iatropoulou A, Becker DL, Franks S, Hardy K. Energy substrate metabolism of mouse cumulus-oocyte complexes: response to follicle-stimulating hormone is mediated by the phosphatidylinositol 3-kinase pathway and is associated with oocyte maturation. Biol Reprod. 2004;71:199–209.PubMedCrossRef
87.
Zurück zum Zitat Nishimoto H, Matsutani R, Yamamoto S, Takahashi T, Hayashi KG, Miyamoto A, et al. Gene expression of glucose transporter (GLUT) 1, 3 and 4 in bovine follicle and corpus luteum. J Endocrinol. 2006;188:111–9.PubMedCrossRef Nishimoto H, Matsutani R, Yamamoto S, Takahashi T, Hayashi KG, Miyamoto A, et al. Gene expression of glucose transporter (GLUT) 1, 3 and 4 in bovine follicle and corpus luteum. J Endocrinol. 2006;188:111–9.PubMedCrossRef
88.
Zurück zum Zitat Williams SA, Blache D, Martin GB, Foot R, Blackberry MA, Scaramuzzi RJ. Effect of nutritional supplementation on quantities of glucose transporters 1 and 4 in sheep granulosa and theca cells. Reproduction. 2001;122:947–56.PubMedCrossRef Williams SA, Blache D, Martin GB, Foot R, Blackberry MA, Scaramuzzi RJ. Effect of nutritional supplementation on quantities of glucose transporters 1 and 4 in sheep granulosa and theca cells. Reproduction. 2001;122:947–56.PubMedCrossRef
89.
Zurück zum Zitat Zhou J, Bievre M, Bondy CA. Reduced GLUT1 expression in Igf1−/− null oocytes and follicles. Growth Horm IGF Res. 2000;10:111–7.PubMedCrossRef Zhou J, Bievre M, Bondy CA. Reduced GLUT1 expression in Igf1−/− null oocytes and follicles. Growth Horm IGF Res. 2000;10:111–7.PubMedCrossRef
90.
Zurück zum Zitat Kodaman PH, Behrman HR. Hormone-regulated and glucose-sensitive transport of dehydroascorbic acid in immature rat granulosa cells. Endocrinology. 1999;140:3659–65.PubMedCrossRef Kodaman PH, Behrman HR. Hormone-regulated and glucose-sensitive transport of dehydroascorbic acid in immature rat granulosa cells. Endocrinology. 1999;140:3659–65.PubMedCrossRef
91.
Zurück zum Zitat Moley KH, Chi MM, Mueckler MM. Maternal hyperglycemia alters glucose transport and utilization in mouse preimplantation embryos. Am J Physiol. 1998;275:E38–47.PubMed Moley KH, Chi MM, Mueckler MM. Maternal hyperglycemia alters glucose transport and utilization in mouse preimplantation embryos. Am J Physiol. 1998;275:E38–47.PubMed
92.
Zurück zum Zitat Moley KH, Chi MM, Knudson CM, Korsmeyer SJ, Mueckler MM. Hyperglycemia induces apoptosis in pre-implantation embryos through cell death effector pathways. Nat Med. 1998;4:1421–4.PubMedCrossRef Moley KH, Chi MM, Knudson CM, Korsmeyer SJ, Mueckler MM. Hyperglycemia induces apoptosis in pre-implantation embryos through cell death effector pathways. Nat Med. 1998;4:1421–4.PubMedCrossRef
93.
Zurück zum Zitat Chi MM, Pingsterhaus J, Carayannopoulos M, Moley KH. Decreased glucose transporter expression triggers BAX-dependent apoptosis in the murine blastocyst. J Biol Chem. 2000;275:40252–7.PubMedCrossRef Chi MM, Pingsterhaus J, Carayannopoulos M, Moley KH. Decreased glucose transporter expression triggers BAX-dependent apoptosis in the murine blastocyst. J Biol Chem. 2000;275:40252–7.PubMedCrossRef
94.
Zurück zum Zitat Wang Q, Ratchford AM, Chi MM, Schoeller E, Frolova A, Schedl T, et al. Maternal diabetes causes mitochondrial dysfunction and meiotic defects in murine oocytes. Mol Endocrinol. 2009;23:1603–12.PubMedCrossRef Wang Q, Ratchford AM, Chi MM, Schoeller E, Frolova A, Schedl T, et al. Maternal diabetes causes mitochondrial dysfunction and meiotic defects in murine oocytes. Mol Endocrinol. 2009;23:1603–12.PubMedCrossRef
95.
Zurück zum Zitat Wang Q, Frolova AI, Purcell S, Adastra K, Schoeller E, Chi MM, et al. Mitochondrial dysfunction and apoptosis in cumulus cells of type I diabetic mice. PLoS ONE. 2010;5:e15901.PubMedCrossRef Wang Q, Frolova AI, Purcell S, Adastra K, Schoeller E, Chi MM, et al. Mitochondrial dysfunction and apoptosis in cumulus cells of type I diabetic mice. PLoS ONE. 2010;5:e15901.PubMedCrossRef
96.
Zurück zum Zitat Wyman A, Pinto AB, Sheridan R, Moley KH. One-cell zygote transfer from diabetic to nondiabetic mouse results in congenital malformations and growth retardation in offspring. Endocrinology. 2008;149:466–9.PubMedCrossRef Wyman A, Pinto AB, Sheridan R, Moley KH. One-cell zygote transfer from diabetic to nondiabetic mouse results in congenital malformations and growth retardation in offspring. Endocrinology. 2008;149:466–9.PubMedCrossRef
97.
Zurück zum Zitat Riley JK, Carayannopoulos MO, Wyman AH, Chi M, Ratajczak CK, Moley KH. The PI3K/Akt pathway is present and functional in the preimplantation mouse embryo. Dev Biol. 2005;284:377–86.PubMedCrossRef Riley JK, Carayannopoulos MO, Wyman AH, Chi M, Ratajczak CK, Moley KH. The PI3K/Akt pathway is present and functional in the preimplantation mouse embryo. Dev Biol. 2005;284:377–86.PubMedCrossRef
98.
Zurück zum Zitat Riley JK, Carayannopoulos MO, Wyman AH, Chi M, Moley KH. Phosphatidylinositol 3-kinase activity is critical for glucose metabolism and embryo survival in murine blastocysts. J Biol Chem. 2006;281:6010–9.PubMedCrossRef Riley JK, Carayannopoulos MO, Wyman AH, Chi M, Moley KH. Phosphatidylinositol 3-kinase activity is critical for glucose metabolism and embryo survival in murine blastocysts. J Biol Chem. 2006;281:6010–9.PubMedCrossRef
99.
Zurück zum Zitat Chi MM, Schlein AL, Moley KH. High insulin-like growth factor 1 (IGF-1) and insulin concentrations trigger apoptosis in the mouse blastocyst via down-regulation of the IGF-1 receptor. Endocrinology. 2000;141:4784–92.PubMedCrossRef Chi MM, Schlein AL, Moley KH. High insulin-like growth factor 1 (IGF-1) and insulin concentrations trigger apoptosis in the mouse blastocyst via down-regulation of the IGF-1 receptor. Endocrinology. 2000;141:4784–92.PubMedCrossRef
100.
Zurück zum Zitat Eng GS, Sheridan RA, Wyman A, Chi MM, Bibee KP, Jungheim ES, et al. AMP kinase activation increases glucose uptake, decreases apoptosis, and improves pregnancy outcome in embryos exposed to high IGF-I concentrations. Diabetes. 2007;56:2228–34.PubMedCrossRef Eng GS, Sheridan RA, Wyman A, Chi MM, Bibee KP, Jungheim ES, et al. AMP kinase activation increases glucose uptake, decreases apoptosis, and improves pregnancy outcome in embryos exposed to high IGF-I concentrations. Diabetes. 2007;56:2228–34.PubMedCrossRef
101.
Zurück zum Zitat Pinto AB, Schlein AL, Moley KH. Preimplantation exposure to high insulin-like growth factor I concentrations results in increased resorption rates in vivo. Hum Reprod. 2002;17:457–62.PubMedCrossRef Pinto AB, Schlein AL, Moley KH. Preimplantation exposure to high insulin-like growth factor I concentrations results in increased resorption rates in vivo. Hum Reprod. 2002;17:457–62.PubMedCrossRef
102.
Zurück zum Zitat Acevedo N, Ding J, Smith GD. Insulin signaling in mouse oocytes. Biol Reprod. 2007;77:872–9.PubMedCrossRef Acevedo N, Ding J, Smith GD. Insulin signaling in mouse oocytes. Biol Reprod. 2007;77:872–9.PubMedCrossRef
103.
Zurück zum Zitat Cecconi S, Rossi G, Santilli A, Stefano LD, Hoshino Y, Sato E, et al. Akt expression in mouse oocytes matured in vivo and in vitro. Reprod Biomed Online. 2010;20:35–41.PubMedCrossRef Cecconi S, Rossi G, Santilli A, Stefano LD, Hoshino Y, Sato E, et al. Akt expression in mouse oocytes matured in vivo and in vitro. Reprod Biomed Online. 2010;20:35–41.PubMedCrossRef
104.
Zurück zum Zitat Kadakia R, Arraztoa JA, Bondy C, Zhou J. Granulosa cell proliferation is impaired in the Igf1 null ovary. Growth Horm IGF Res. 2001;11:220–4.PubMedCrossRef Kadakia R, Arraztoa JA, Bondy C, Zhou J. Granulosa cell proliferation is impaired in the Igf1 null ovary. Growth Horm IGF Res. 2001;11:220–4.PubMedCrossRef
105.
Zurück zum Zitat Minge CE, Bennett BD, Norman RJ, Robker RL. Peroxisome proliferator-activated receptor-gamma agonist rosiglitazone reverses the adverse effects of diet-induced obesity on oocyte quality. Endocrinology. 2008;149:2646–56.PubMedCrossRef Minge CE, Bennett BD, Norman RJ, Robker RL. Peroxisome proliferator-activated receptor-gamma agonist rosiglitazone reverses the adverse effects of diet-induced obesity on oocyte quality. Endocrinology. 2008;149:2646–56.PubMedCrossRef
106.
Zurück zum Zitat Fedorcsak P, Storeng R, Dale PO, Tanbo T, Abyholm T. Impaired insulin action on granulosa-lutein cells in women with polycystic ovary syndrome and insulin resistance. Gynecol Endocrinol. 2000;14:327–36.PubMedCrossRef Fedorcsak P, Storeng R, Dale PO, Tanbo T, Abyholm T. Impaired insulin action on granulosa-lutein cells in women with polycystic ovary syndrome and insulin resistance. Gynecol Endocrinol. 2000;14:327–36.PubMedCrossRef
107.
Zurück zum Zitat Clark AM, Ledger W, Galletly C, Tomlinson L, Blaney F, Wang X, et al. Weight loss results in significant improvement in pregnancy and ovulation rates in anovulatory obese women. Hum Reprod. 1995;10:2705–12.PubMed Clark AM, Ledger W, Galletly C, Tomlinson L, Blaney F, Wang X, et al. Weight loss results in significant improvement in pregnancy and ovulation rates in anovulatory obese women. Hum Reprod. 1995;10:2705–12.PubMed
108.
Zurück zum Zitat Clark AM, Thornley B, Tomlinson L, Galletley C, Norman RJ. Weight loss in obese infertile women results in improvement in reproductive outcome for all forms of fertility treatment. Hum Reprod. 1998;13:1502–5.PubMedCrossRef Clark AM, Thornley B, Tomlinson L, Galletley C, Norman RJ. Weight loss in obese infertile women results in improvement in reproductive outcome for all forms of fertility treatment. Hum Reprod. 1998;13:1502–5.PubMedCrossRef
Metadaten
Titel
The impact of obesity on egg quality
verfasst von
Scott H. Purcell
Kelle H. Moley
Publikationsdatum
01.06.2011
Verlag
Springer US
Erschienen in
Journal of Assisted Reproduction and Genetics / Ausgabe 6/2011
Print ISSN: 1058-0468
Elektronische ISSN: 1573-7330
DOI
https://doi.org/10.1007/s10815-011-9592-y

Weitere Artikel der Ausgabe 6/2011

Journal of Assisted Reproduction and Genetics 6/2011 Zur Ausgabe

Hirsutismus bei PCOS: Laser- und Lichttherapien helfen

26.04.2024 Hirsutismus Nachrichten

Laser- und Lichtbehandlungen können bei Frauen mit polyzystischem Ovarialsyndrom (PCOS) den übermäßigen Haarwuchs verringern und das Wohlbefinden verbessern – bei alleiniger Anwendung oder in Kombination mit Medikamenten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Weniger postpartale Depressionen nach Esketamin-Einmalgabe

Bislang gibt es kein Medikament zur Prävention von Wochenbettdepressionen. Das Injektionsanästhetikum Esketamin könnte womöglich diese Lücke füllen.

Bei RSV-Impfung vor 60. Lebensjahr über Off-Label-Gebrauch aufklären!

22.04.2024 DGIM 2024 Kongressbericht

Durch die Häufung nach der COVID-19-Pandemie sind Infektionen mit dem Respiratorischen Synzytial-Virus (RSV) in den Fokus gerückt. Fachgesellschaften empfehlen eine Impfung inzwischen nicht nur für Säuglinge und Kleinkinder.

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.