Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
RESEARCH ARTICLE

Effect of varying glucose and glucosamine concentration in vitro on mouse oocyte maturation and developmental competence

L. A. Frank A , M. L. Sutton-McDowall A , D. L. Russell A , X. Wang A , D. K. Feil A , R. B. Gilchrist A and J. G. Thompson A B
+ Author Affiliations
- Author Affiliations

A The Robinson Institute, Research Centre for Reproductive Health, School of Paediatrics and Reproductive Health, University of Adelaide, Adelaide, SA 5005, Australia.

B Corresponding author. Email: jeremy.thompson@adelaide.edu.au

Reproduction, Fertility and Development 25(8) 1095-1104 https://doi.org/10.1071/RD12275
Submitted: 22 August 2012  Accepted: 7 October 2012   Published: 7 November 2012

Abstract

The effects of hyper- and hypo-glycaemic conditions during the in vitro maturation of mouse cumulus–oocyte complexes on developmental competence were examined, with an emphasis on the role of the hexosamine biosynthesis pathway. A low (1 mM) glucose concentration achieved optimal oocyte competence (3-fold higher blastocyst development rate compared with high (30 mM) glucose, P < 0.05). In addition, glucose supplementation during only the first hour after release from the follicle was necessary and sufficient to support oocyte maturation and embryo development to the blastocyst stage. Glucosamine (a known hyperglycaemic mimetic and specific activator of the hexosamine pathway) was able to substitute for glucose during this first hour, indicating that flux through the hexosamine pathway is essential for oocyte competence. In the absence of glucose throughout the maturation period, glucosamine was not able to increase developmental competence, and at higher concentrations (2.5 and 5 mM) had a detrimental effect on MII and blastocyst development rates, compared with controls (P < 0.05). These experiments underscore the importance of glucose metabolic pathways during in vitro maturation and support the concept that excess flux through the hexosamine pathway has detrimental consequences.

Additional keywords : culture medium, cumulus cells, embryo, hexosamine biosynthesis pathway.


References

Ali, A., and Sirard, M. A. (2002). Effect of the absence or presence of various protein supplements on further development of bovine oocytes during in vitro maturation. Biol. Reprod. 66, 901–905.
Effect of the absence or presence of various protein supplements on further development of bovine oocytes during in vitro maturation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XitlCltLs%3D&md5=eebc9f0f9295e3f45db4d3ee03108938CAS | 11906907PubMed |

Becerra, J. E., Khoury, M. J., Cordero, J. F., and Erickson, J. D. (1990). Diabetes mellitus during pregnancy and the risks for specific birth defects: a population-based case-control study. Pediatrics 85, 1–9.
| 1:STN:280:DyaK3c7hsFWluw%3D%3D&md5=075cd51b76d3d19dde978207355dec9dCAS | 2404255PubMed |

Chang, A. S., Dale, A. N., and Moley, K. H. (2005). Maternal diabetes adversely affects preovulatory oocyte maturation, development and granulosa cell apoptosis. Endocrinology 146, 2445–2453.
Maternal diabetes adversely affects preovulatory oocyte maturation, development and granulosa cell apoptosis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXjslamtbk%3D&md5=ef12a32bc6d700c8e5517369a92b5726CAS | 15718275PubMed |

Chen, L., Wert, S. E., Hendrix, E. M., Russell, P. T., Cannon, M., and Larsen, W. J. (1990). Hyaluronic acid synthesis and gap junction endocytosis are necessary for normal expansion of the cumulus mass. Mol. Reprod. Dev. 26, 236–247.
Hyaluronic acid synthesis and gap junction endocytosis are necessary for normal expansion of the cumulus mass.Crossref | GoogleScholarGoogle Scholar | 2115793PubMed |

Chen, L., Russell, P. T., and Larsen, W. J. (1993). Functional significance of cumulus expansion in the mouse: roles for the preovulatory synthesis of hyaluronic acid within the cumulus mass. Mol. Reprod. Dev. 34, 87–93.
Functional significance of cumulus expansion in the mouse: roles for the preovulatory synthesis of hyaluronic acid within the cumulus mass.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3sXhsVCgs74%3D&md5=c049164ae0318c888160a80b25fe10eaCAS | 8418823PubMed |

Clark, A. R., Stokes, Y. M., and Thompson, J. G. (2011). Estimation of glucose uptake by ovarian follicular cells. Ann. Biomed. Eng. 39, 2654–2667.
Estimation of glucose uptake by ovarian follicular cells.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BC3MfjvVOitQ%3D%3D&md5=d252e218e590bd96edbcaa64d87bc691CAS | 21769539PubMed |

Colton, S. A., Pieper, G. M., and Downs, S. M. (2002). Altered meiotic regulation in oocytes from diabetic mice. Biol. Reprod. 67, 220–231.
Altered meiotic regulation in oocytes from diabetic mice.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XkvV2itb8%3D&md5=d3a7a57bc211eeb9415f72fe54449cd0CAS | 12080021PubMed |

Cornblath, M., and Schwartz, R. (1976). Disorders of carbohydrate metabolism in infancy. In ‘Major Problems in Clinical Pediatrics Vol. 3.’ 1976/01/01 edn. (Saunders: Philadelphia.)

Downs, S. M., and Mastropolo, A. M. (1994). The participation of energy substrates in the control of meiotic maturation in murine oocytes. Dev. Biol. 162, 154–168.
The participation of energy substrates in the control of meiotic maturation in murine oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2cXhvVyitLs%3D&md5=9b2c901f69561a8113c811e81b6c4bbdCAS | 8125183PubMed |

Downs, S. M., and Utecht, A. M. (1999). Metabolism of radiolabelled glucose by mouse oocytes and oocyte–cumulus cell complexes. Biol. Reprod. 60, 1446–1452.
Metabolism of radiolabelled glucose by mouse oocytes and oocyte–cumulus cell complexes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1MXjsVeisLs%3D&md5=40a29e21043de48843a9804dc3dc55b9CAS | 10330104PubMed |

Downs, S. M., Humpherson, P. G., Martin, K. L., and Leese, H. J. (1996). Glucose utilization during gonadotrophin-induced meiotic maturation in cumulus cell-enclosed mouse oocytes. Mol. Reprod. Dev. 44, 121–131.
Glucose utilization during gonadotrophin-induced meiotic maturation in cumulus cell-enclosed mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK28XivV2mt7g%3D&md5=0957b800a3425a7a490daa950260e7b5CAS | 8722700PubMed |

Downs, S. M., Humpherson, P. G., and Leese, H. J. (1998). Meiotic induction in cumulus cell-enclosed mouse oocytes: involvement of the pentose phosphate pathway. Biol. Reprod. 58, 1084–1094.
Meiotic induction in cumulus cell-enclosed mouse oocytes: involvement of the pentose phosphate pathway.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXit1KhtL0%3D&md5=ea5c6377cea453bbfebebb6c43dc267cCAS | 9546744PubMed |

Dunne, F.P., Brydon, P., Smith, T., Essex, M., Nicholson, H., and Dunn, J. (1999). Pre-conception diabetes care in insulin-dependent diabetes mellitus. QJM: monthly journal of the Association of Physicians 92, 175–176.
Pre-conception diabetes care in insulin-dependent diabetes mellitus.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK1M3lsl2htQ%3D%3D&md5=7b1d7dc97b7babcd9630019cf8f9c0c6CAS | 10326077PubMed |

Eppig, J. J. (1981). Regulation by sulfated glycosaminoglycans of the expansion of cumuli oophori isolated from mice. Biol. Reprod. 25, 599–608.
Regulation by sulfated glycosaminoglycans of the expansion of cumuli oophori isolated from mice.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL3MXmtFGgt7s%3D&md5=08981c746dfcf8e766545ce03e26d60dCAS | 6272891PubMed |

Eppig, J. J., Schultz, R. M., O’Brien, M., and Chesnel, F. (1994). Relationship between the developmental programs controlling nuclear and cytoplasmic maturation of mouse oocytes. Dev. Biol. 164, 1–9.
Relationship between the developmental programs controlling nuclear and cytoplasmic maturation of mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK2c3pvFSjtA%3D%3D&md5=d1de9078c9c637a6c451a6a84120c1ccCAS | 8026614PubMed |

Eppig, J. J., Hosoe, M., O’Brien, M. J., Pendola, F. M., Requena, A., and Watanabe, S. (2000). Conditions that affect acquisition of developmental competence by mouse oocytes in vitro: FSH, insulin, glucose and ascorbic acid. Mol. Cell. Endocrinol. 163, 109–116.
Conditions that affect acquisition of developmental competence by mouse oocytes in vitro: FSH, insulin, glucose and ascorbic acid.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3cXlvV2ms7o%3D&md5=d9266b465af36908704d57399588143dCAS | 10963882PubMed |

Fagbohun, C. F., and Downs, S. M. (1992). Requirement for glucose in ligand-stimulated meiotic maturation of cumulus cell-enclosed mouse oocytes. J. Reprod. Fertil. 96, 681–697.
Requirement for glucose in ligand-stimulated meiotic maturation of cumulus cell-enclosed mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3sXpvVyqsw%3D%3D&md5=c741f71f787cbcdb2550e90d3e620cc1CAS | 1339848PubMed |

Farrell, T., Neale, L., and Cundy, T. (2002). Congenital anomalies in the offspring of women with type 1, type 2 and gestational diabetes. Diabet. Med. 19, 322–326.
Congenital anomalies in the offspring of women with type 1, type 2 and gestational diabetes.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD383gvFeqsQ%3D%3D&md5=f23ad81d8bcd5e8b6b4622d412a5bad9CAS | 11943005PubMed |

Funahashi, H., Koike, T., and Sakai, R. (2008). Effect of glucose and pyruvate on nuclear and cytoplasmic maturation of porcine oocytes in a chemically-defined medium. Theriogenology 70, 1041–1047.
Effect of glucose and pyruvate on nuclear and cytoplasmic maturation of porcine oocytes in a chemically-defined medium.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhtFajurjK&md5=628f81baa6d6f6f4993c1f22dd0f2ac2CAS | 18657854PubMed |

Greene, M. F. (1999). Spontaneous abortions and major malformations in women with diabetes mellitus. Semin. Reprod. Endocrinol. 17, 127–136.
Spontaneous abortions and major malformations in women with diabetes mellitus.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK1MvlslKksw%3D%3D&md5=b1b27cbaebe72104aad5124304fe074aCAS | 10528364PubMed |

Guo, X., and Gao, S. (2009). Pins homolog LGN regulates meiotic spindle organization in mouse oocytes. Cell Res. 19, 838–848.
Pins homolog LGN regulates meiotic spindle organization in mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXot12ltbc%3D&md5=04b9762631bcf644fe652a898f72273fCAS | 19434098PubMed |

Gutnisky, C., Dalvit, G. C., Pintos, L. N., Thompson, J. G., Beconi, M. T., and Cetica, P. D. (2007). Influence of hyaluronic acid synthesis and cumulus mucification on bovine oocyte in vitro maturation, fertilisation and embryo development. Reprod. Fertil. Dev. 19, 488–497.
Influence of hyaluronic acid synthesis and cumulus mucification on bovine oocyte in vitro maturation, fertilisation and embryo development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXjtFKnsLg%3D&md5=e812165019d4f2fe29f038fd67f3ca99CAS | 17394798PubMed |

Harris, S. E., Gopichandran, N., Picton, H. M., Leese, H. J., and Orsi, N. M. (2005). Nutrient concentrations in murine follicular fluid and the female reproductive tract. Theriogenology 64, 992–1006.
Nutrient concentrations in murine follicular fluid and the female reproductive tract.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXmvVGjsbw%3D&md5=2308de1d60050d36de2a97c47e11b5caCAS | 16054501PubMed |

Hashimoto, S., Minami, N., Yamada, M., and Imai, H. (2000). Excessive concentration of glucose during in vitro maturation impairs the developmental competence of bovine oocytes after in vitro fertilization: relevance to intracellular reactive oxygen species and glutathione contents. Mol. Reprod. Dev. 56, 520–526.
Excessive concentration of glucose during in vitro maturation impairs the developmental competence of bovine oocytes after in vitro fertilization: relevance to intracellular reactive oxygen species and glutathione contents.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3cXkslKlur8%3D&md5=5a55b54aec94843bb22998db99112a3cCAS | 10911402PubMed |

Hendryx, J. T., and Wordinger, R. J. (1979). The effects of decreased glucose concentrations on the in vitro development of the post-blastocyst mouse embryo in a fetal calf serum- or bovine serum albumin-supplemented medium. Experientia 35, 1508–1510.
The effects of decreased glucose concentrations on the in vitro development of the post-blastocyst mouse embryo in a fetal calf serum- or bovine serum albumin-supplemented medium.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL3cXjvVGqtg%3D%3D&md5=aa3ffc210ca92c3343697fe5f88cb85cCAS | 510505PubMed |

Herrick, J. R., Lane, M., Gardner, D. K., Behboodi, E., Memili, E., Blash, S., Echelard, Y., and Krisher, R. L. (2006). Metabolism, protein content and in vitro embryonic development of goat cumulus–oocyte complexes matured with physiological concentrations of glucose and l-lactate. Mol. Reprod. Dev. 73, 256–266.
Metabolism, protein content and in vitro embryonic development of goat cumulus–oocyte complexes matured with physiological concentrations of glucose and l-lactate.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xjslyntw%3D%3D&md5=8ef7e1734cd732a9c700c43a5a8db6a7CAS | 16250005PubMed |

Hizaki, H., Segi, E., Sugimoto, Y., Hirose, M., Saji, T., Ushikubi, F., Matsuoka, T., Noda, Y., Tanaka, T., Yoshida, N., Narumiya, S., and Ichikawa, A. (1999). Abortive expansion of the cumulus and impaired fertility in mice lacking the prostaglandin E receptor subtype EP(2). Proc. Natl. Acad. Sci. USA 96, 10501–10506.
Abortive expansion of the cumulus and impaired fertility in mice lacking the prostaglandin E receptor subtype EP(2).Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1MXlvFemsrg%3D&md5=0a67420b68153b5cdad7531ec073ea44CAS | 10468638PubMed |

Jamnongjit, M., Gill, A., and Hammes, S. R. (2005). Epidermal growth factor receptor signalling is required for normal ovarian steroidogenesis and oocyte maturation. Proc. Natl. Acad. Sci. USA 102, 16257–16262.
Epidermal growth factor receptor signalling is required for normal ovarian steroidogenesis and oocyte maturation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXht1Cgur3K&md5=46d06e376997c51ee982540b36b0ddfaCAS | 16260720PubMed |

Kakar, M. A., Maddocks, S., Lorimer, M. F., Kleemann, D. O., Rudiger, S. R., Hartwich, K. M., and Walker, S. K. (2005). The effect of peri-conception nutrition on embryo quality in the superovulated ewe. Theriogenology 64, 1090–1103.
The effect of peri-conception nutrition on embryo quality in the superovulated ewe.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD2MvlvF2muw%3D%3D&md5=32d2dbae8ab2c21414e409760a6e5e39CAS | 16125553PubMed |

Kanji, M. I., Toews, M. L., and Carper, W. R. (1976). A kinetic study of glucose-6-phosphate dehydrogenase. J. Biol. Chem. 251, 2258–2262.
| 1:CAS:528:DyaE28XktFKhs74%3D&md5=264875dcc74a71f6f7e760cd46cf31b6CAS |

Kawashima, I., Liu, Z., Mullany, L. K., Mihara, T., Richards, J. S., and Shimada, M. (2012). EGF-like factors induce expansion of the cumulus cell–oocyte complexes by activating calpain-mediated cell movement. Endocrinology 153, 3949–3959.
EGF-like factors induce expansion of the cumulus cell–oocyte complexes by activating calpain-mediated cell movement.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhtFGktbvF&md5=56cbca01b568dd24608b7f185632402bCAS | 22673225PubMed |

Khurana, N. K., and Niemann, H. (2000). Effects of oocyte quality, oxygen tension, embryo density, cumulus cells and energy substrates on cleavage and morula/blastocyst formation of bovine embryos. Theriogenology 54, 741–756.
Effects of oocyte quality, oxygen tension, embryo density, cumulus cells and energy substrates on cleavage and morula/blastocyst formation of bovine embryos.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD3M%2FovVGrtA%3D%3D&md5=9437dfe3bb3430944ec13158291ffef1CAS | 11101035PubMed |

Kimura, K., Iwata, H., and Thompson, J. G. (2008). The effect of glucosamine concentration on the development and sex ratio of bovine embryos. Anim. Reprod. Sci. 103, 228–238.
The effect of glucosamine concentration on the development and sex ratio of bovine embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhsVSmtLfL&md5=911f3e52001d3726295ca884e71651f2CAS | 17198747PubMed |

Lapolla, A., Dalfra, M. G., and Fedele, D. (2008). Pregnancy complicated by type 2 diabetes: an emerging problem. Diabetes Res. Clin. Pract. 80, 2–7.
Pregnancy complicated by type 2 diabetes: an emerging problem.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXktVKhu7g%3D&md5=7dc87f7eb00dc91e17e553167e5c35a6CAS | 18201793PubMed |

Lea, R. G., McCracken, J. E., McIntyre, S. S., Smith, W., and Baird, J. D. (1996). Disturbed development of the preimplantation embryo in the insulin-dependent diabetic BB/E rat. Diabetes 45, 1463–1470.
Disturbed development of the preimplantation embryo in the insulin-dependent diabetic BB/E rat.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK28XmsVyrurk%3D&md5=bfa412617d1db9c143d66203a623cdb1CAS | 8866548PubMed |

Luciano, A. M., Modina, S., Vassena, R., Milanesi, E., Lauria, A., and Gandolfi, F. (2004). Role of intracellular cyclic adenosine 3′,5′-monophosphate concentration and oocyte–cumulus cells communications on the acquisition of the developmental competence during in vitro maturation of bovine oocyte. Biol. Reprod. 70, 465–472.
Role of intracellular cyclic adenosine 3′,5′-monophosphate concentration and oocyte–cumulus cells communications on the acquisition of the developmental competence during in vitro maturation of bovine oocyte.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXnsl2juw%3D%3D&md5=391df76b3c4f6073d0cf20a223bd5b0cCAS | 14568913PubMed |

Marshall, S., Bacote, V., and Traxinger, R. R. (1991). Discovery of a metabolic pathway mediating glucose-induced desensitization of the glucose transport system. Role of hexosamine biosynthesis in the induction of insulin resistance. J. Biol. Chem. 266, 4706–4712.
| 1:CAS:528:DyaK3MXhs1WltL0%3D&md5=38dc74a7dac082cbc219315801d64c11CAS | 2002019PubMed |

Marshall, S., Nadeau, O., and Yamasaki, K. (2005). Glucosamine-induced activation of glycogen biosynthesis in isolated adipocytes. Evidence for a rapid allosteric control mechanism within the hexosamine biosynthesis pathway. J. Biol. Chem. 280, 11018–11024.
Glucosamine-induced activation of glycogen biosynthesis in isolated adipocytes. Evidence for a rapid allosteric control mechanism within the hexosamine biosynthesis pathway.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXisVOqur0%3D&md5=d7521f4ef0ca733e0b53bd021a962353CAS | 15647256PubMed |

Merriman, J. A., Whittingham, D. G., and Carroll, J. (1998). The effect of follicle-stimulating hormone and epidermal growth factor on the developmental capacity of in vitro-matured mouse oocytes. Hum. Reprod. 13, 690–695.
The effect of follicle-stimulating hormone and epidermal growth factor on the developmental capacity of in vitro-matured mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXivF2jt7Y%3D&md5=1ee5fb026a858ad70cd73c13864e181dCAS | 9572435PubMed |

Mikkelsen, A. L., Host, E., Blaabjerg, J., and Lindenberg, S. (2001). Maternal serum supplementation in culture medium benefits maturation of immature human oocytes. Reprod. Biomed. Online 3, 112–116.
Maternal serum supplementation in culture medium benefits maturation of immature human oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXos1Gktb0%3D&md5=f301f38c434a783fe84853efca13c1a1CAS | 12513873PubMed |

Moley, K. H., Vaughn, W. K., DeCherney, A. H., and Diamond, M. P. (1991). Effect of diabetes mellitus on mouse pre-implantation embryo development. J. Reprod. Fertil. 93, 325–332.
Effect of diabetes mellitus on mouse pre-implantation embryo development.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK387lslSlsw%3D%3D&md5=e0a7a06809954f29001871233b929f8cCAS | 1787451PubMed |

Pantaleon, M., Scott, J., and Kaye, P. L. (2008). Nutrient sensing by the early mouse embryo: hexosamine biosynthesis and glucose signaling during preimplantation development. Biol. Reprod. 78, 595–600.
Nutrient sensing by the early mouse embryo: hexosamine biosynthesis and glucose signaling during preimplantation development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXjvVaitbo%3D&md5=12a03adbb2053c244953821acd383e7eCAS | 18046015PubMed |

Pantaleon, M., Tan, H. Y., Kafer, G. R., and Kaye, P. L. (2010). Toxic effects of hyperglycaemia are mediated by the hexosamine signalling pathway and O-linked glycosylation in early mouse embryos. Biol. Reprod. 82, 751–758.
Toxic effects of hyperglycaemia are mediated by the hexosamine signalling pathway and O-linked glycosylation in early mouse embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXjslelt7g%3D&md5=df1faed222429acecea53d2ff4139050CAS | 20032283PubMed |

Ray, J. G., O’Brien, T. E., and Chan, W. S. (2001). Preconception care and the risk of congenital anomalies in the offspring of women with diabetes mellitus: a meta-analysis. QJM 94, 435–444.
Preconception care and the risk of congenital anomalies in the offspring of women with diabetes mellitus: a meta-analysis.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD3Mvos1Oqsg%3D%3D&md5=4890a1c02a7fa4ca19886648adbaf1a5CAS | 11493721PubMed |

Reizel, Y., Elbaz, J., and Dekel, N. (2010). Sustained activity of the EGF receptor is an absolute requisite for LH-induced oocyte maturation and cumulus expansion. Mol. Endocrinol. 24, 402–411.
Sustained activity of the EGF receptor is an absolute requisite for LH-induced oocyte maturation and cumulus expansion.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhslejsrs%3D&md5=754d1277c454534d8304d3409dcde375CAS | 20009084PubMed |

Rose, T. A., and Bavister, B. D. (1992). Effect of oocyte maturation medium on in vitro development of in vitro-fertilized bovine embryos. Mol. Reprod. Dev. 31, 72–77.
Effect of oocyte maturation medium on in vitro development of in vitro-fertilized bovine embryos.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK383it1GqtQ%3D%3D&md5=b3229ce5adff004c688860a05c6e2e14CAS | 1562330PubMed |

Rose-Hellekant, T. A., Libersky-Williamson, E. A., and Bavister, B. D. (1998). Energy substrates and amino acids provided during in vitro maturation of bovine oocytes alter acquisition of developmental competence. Zygote 6, 285–294.
Energy substrates and amino acids provided during in vitro maturation of bovine oocytes alter acquisition of developmental competence.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1MXhtVyqu7o%3D&md5=e2ae3e415fb9ea964adca71e276747bfCAS | 9921638PubMed |

Russell, D. L., and Robker, R. L. (2007). Molecular mechanisms of ovulation: co-ordination through the cumulus complex. Hum. Reprod. Update 13, 289–312.
Molecular mechanisms of ovulation: co-ordination through the cumulus complex.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXmtlCisbc%3D&md5=f8e463f96a73430c0443a97d3b031645CAS | 17242016PubMed |

Sadler, T. W., Hunter, E. S., Balkan, W., and Horton, W. E. (1988). Effects of maternal diabetes on embryogenesis. Am. J. Perinatol. 5, 319–326.
Effects of maternal diabetes on embryogenesis.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaL1M%2FgtVShsA%3D%3D&md5=5834fd72b96b7561615972fd57384352CAS | 3166639PubMed |

Salustri, A., Yanagishita, M., and Hascall, V. C. (1989). Synthesis and accumulation of hyaluronic acid and proteoglycans in the mouse cumulus cell–oocyte complex during follicle-stimulating hormone-induced mucification. J. Biol. Chem. 264, 13840–13847.
| 1:CAS:528:DyaL1MXlt1OgsLg%3D&md5=0e00d3194fbb257f16c179ff088b380aCAS | 2503506PubMed |

Sato, H., Iwata, H., Hayashi, T., Kimura, K., Kuwayama, T., and Monji, Y. (2007). The effect of glucose on the progression of the nuclear maturation of pig oocytes. Anim. Reprod. Sci. 99, 299–305.
The effect of glucose on the progression of the nuclear maturation of pig oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXjslGkuro%3D&md5=f23bc532e8a3ceec7e3eb865951d7abbCAS | 16784824PubMed |

Sayeski, P. P., and Kudlow, J. E. (1996). Glucose metabolism to glucosamine is necessary for glucose stimulation of transforming growth factor-alpha gene transcription. J. Biol. Chem. 271, 15237–15243.
Glucose metabolism to glucosamine is necessary for glucose stimulation of transforming growth factor-alpha gene transcription.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK28XjvVWmu7c%3D&md5=4da93092e2b502564974304828282122CAS | 8663078PubMed |

Schelbach, C. J. (2011) ‘An Investigation into the Effect of Glucosamine on Reproductive Outcomes in the Mouse’. (University of Adelaide: Adelaide).

Schelbach, C. J., Kind, K. L., Lane, M., and Thompson, J. G. (2010). Mechanisms contributing to the reduced developmental competence of glucosamine-exposed mouse oocytes. Reprod. Fertil. Dev. 22, 771–779.
Mechanisms contributing to the reduced developmental competence of glucosamine-exposed mouse oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXlsVCqt7k%3D&md5=445a95480dd8ef1d7afabfd3cb3c75bcCAS | 20450829PubMed |

Schroeder, A. C., Schultz, R. M., Kopf, G. S., Taylor, F. R., Becker, R. B., and Eppig, J. J. (1990). Fetuin inhibits zona pellucida hardening and conversion of ZP2 to ZP2f during spontaneous mouse oocyte maturation in vitro in the absence of serum. Biol. Reprod. 43, 891–897.
Fetuin inhibits zona pellucida hardening and conversion of ZP2 to ZP2f during spontaneous mouse oocyte maturation in vitro in the absence of serum.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3cXmt1ymtbw%3D&md5=4b11fe6df7b3aed6014d2e792521cdfcCAS | 1705446PubMed |

Sutton, M. L., Gilchrist, R. B., and Thompson, J. G. (2003). 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 9, 35–48.
Effects of in vivo and in vitro environments on the metabolism of the cumulus–oocyte complex and its influence on oocyte developmental capacity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXjtVelt7Y%3D&md5=9ebd7426b7806b3a80f3af517260eaf0CAS | 12638780PubMed |

Sutton-McDowall, M. L., Gilchrist, R. B., and Thompson, J. G. (2004). Cumulus expansion and glucose utilisation by bovine cumulus–oocyte complexes during in vitro maturation: the influence of glucosamine and follicle-stimulating hormone. Reproduction 128, 313–319.
Cumulus expansion and glucose utilisation by bovine cumulus–oocyte complexes during in vitro maturation: the influence of glucosamine and follicle-stimulating hormone.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXot1KitLk%3D&md5=cca0c749897e2e1e952e0bc3b98bcf73CAS | 15333782PubMed |

Sutton-McDowall, M. L., Gilchrist, R. B., and Thompson, J. G. (2005). Effect of hexoses and gonadotrophin supplementation on bovine oocyte nuclear maturation during in vitro maturation in a synthetic follicle fluid medium. Reprod. Fertil. Dev. 17, 407–415.
Effect of hexoses and gonadotrophin supplementation on bovine oocyte nuclear maturation during in vitro maturation in a synthetic follicle fluid medium.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXjtFOktbs%3D&md5=495f8e3267cf80bd95d9d644857e276dCAS | 15899152PubMed |

Sutton-McDowall, M. L., Mitchell, M., Cetica, P., Dalvit, G., Pantaleon, M., Lane, M., Gilchrist, R. B., and Thompson, J. G. (2006). Glucosamine supplementation during in vitro maturation inhibits subsequent embryo development: possible role of the hexosamine pathway as a regulator of developmental competence. Biol. Reprod. 74, 881–888.
Glucosamine supplementation during in vitro maturation inhibits subsequent embryo development: possible role of the hexosamine pathway as a regulator of developmental competence.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xjsl2jur8%3D&md5=6346693bebf8341947914828cbbf172dCAS | 16436527PubMed |

Sutton-McDowall, M. L., Gilchrist, R. B., and Thompson, J. G. (2010). The pivotal role of glucose metabolism in determining oocyte developmental competence. Reproduction 139, 685–695.
The pivotal role of glucose metabolism in determining oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXltFajtr0%3D&md5=e999bd2e5438715c307566da1c288f51CAS | 20089664PubMed |

Tirone, E., D’Alessandris, C., Hascall, V. C., Siracusa, G., and Salustri, A. (1997). Hyaluronan synthesis by mouse cumulus cells is regulated by interactions between follicle-stimulating hormone (or epidermal growth factor) and a soluble oocyte factor (or transforming growth factor beta1). J. Biol. Chem. 272, 4787–4794.
Hyaluronan synthesis by mouse cumulus cells is regulated by interactions between follicle-stimulating hormone (or epidermal growth factor) and a soluble oocyte factor (or transforming growth factor beta1).Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXhsValtrg%3D&md5=a7e74d3fdd0074709227853e475179b5CAS | 9030534PubMed |

van de Sandt, J. J., Schroeder, A. C., and Eppig, J. J. (1990). Culture media for mouse oocyte maturation affect subsequent embryonic development. Mol. Reprod. Dev. 25, 164–171.
Culture media for mouse oocyte maturation affect subsequent embryonic development.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK3c7otVCgsA%3D%3D&md5=a2745eff7614e3972017d8dd2e53309bCAS | 2310566PubMed |

Vanderhyden, B. C., Caron, P. J., Buccione, R., and Eppig, J. J. (1990). Developmental pattern of the secretion of cumulus expansion-enabling factor by mouse oocytes and the role of oocytes in promoting granulosa cell differentiation. Dev. Biol. 140, 307–317.
Developmental pattern of the secretion of cumulus expansion-enabling factor by mouse oocytes and the role of oocytes in promoting granulosa cell differentiation.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK3czitlersw%3D%3D&md5=972eb18bc9a9404cb48f5a99e6bde168CAS | 2115479PubMed |

Virk, J., Li, J., Vestergaard, M., Obel, C., Lu, M., and Olsen, J. (2010). Early life disease programming during the preconception and prenatal period: making the link between stressful life events and type-1 diabetes. PLoS ONE 5, e11523.
Early life disease programming during the preconception and prenatal period: making the link between stressful life events and type-1 diabetes.Crossref | GoogleScholarGoogle Scholar | 20634978PubMed |

Wahabi, H. A., Alzeidan, R. A., Bawazeer, G. A., Alansari, L. A., and Esmaeil, S. A. (2010). Preconception care for diabetic women for improving maternal and fetal outcomes: a systematic review and meta-analysis. BMC Pregnancy Childbirth 10, 63.
Preconception care for diabetic women for improving maternal and fetal outcomes: a systematic review and meta-analysis.Crossref | GoogleScholarGoogle Scholar | 20946676PubMed |

Wang, Q., Ratchford, A. M., Chi, M. M., Schoeller, E., Frolova, A., Schedl, T., and Moley, K. H. (2009). Maternal diabetes causes mitochondrial dysfunction and meiotic defects in murine oocytes. Mol. Endocrinol. 23, 1603–1612.
Maternal diabetes causes mitochondrial dysfunction and meiotic defects in murine oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhtlSitb%2FF&md5=c58453597793c6010509ddb97ac1163bCAS | 19574447PubMed |

Wells, L., Whelan, S. A., and Hart, G. W. (2003). O-GlcNAc: a regulatory post-translational modification. Biochem. Biophys. Res. Commun. 302, 435–441.
O-GlcNAc: a regulatory post-translational modification.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXhs1Khuro%3D&md5=c8eca46301803ef97dc796e9d324d71aCAS | 12615051PubMed |

Wyman, A., Pinto, A. B., Sheridan, R., and Moley, K. H. (2008). One-cell zygote transfer from diabetic to non-diabetic mouse results in congenital malformations and growth retardation in offspring. Endocrinology 149, 466–469.
One-cell zygote transfer from diabetic to non-diabetic mouse results in congenital malformations and growth retardation in offspring.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXht1yqsLc%3D&md5=a37a9891d61651c55e901863933008e9CAS | 18039778PubMed |

Yang, X., Ongusaha, P. P., Miles, P. D., Havstad, J. C., Zhang, F., So, W. V., Kudlow, J. E., Michell, R. H., Olefsky, J. M., Field, S. J., and Evans, R. M. (2008). Phosphoinositide signalling links O-GlcNAc transferase to insulin resistance. Nature 451, 964–969.
Phosphoinositide signalling links O-GlcNAc transferase to insulin resistance.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXit1yns74%3D&md5=33a1f877782474aba4cd70d5b91816d3CAS | 18288188PubMed |

Zachara, N. E., and Hart, G. W. (2004). O-GlcNAc a sensor of cellular state: the role of nucleocytoplasmic glycosylation in modulating cellular function in response to nutrition and stress. Biochim. Biophys. Acta 1673, 13–28.
O-GlcNAc a sensor of cellular state: the role of nucleocytoplasmic glycosylation in modulating cellular function in response to nutrition and stress.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXltl2htLc%3D&md5=6ae7222a4d53243a680a5d6730edc68cCAS | 15238246PubMed |

Zhang, Z., Liew, C. W., Handy, D. E., Zhang, Y., Leopold, J. A., Hu, J., Guo, L., Kulkarni, R. N., Loscalzo, J., and Stanton, R. C. (2010). High glucose inhibits glucose-6-phosphate dehydrogenase, leading to increased oxidative stress and beta-cell apoptosis. FASEB J. 24, 1497–1505.
High glucose inhibits glucose-6-phosphate dehydrogenase, leading to increased oxidative stress and beta-cell apoptosis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXlslShsrg%3D&md5=231915109b39f1d8989e1a3af69c3d03CAS | 20032314PubMed |