Skip to main content Accessibility help
×
Hostname: page-component-77c89778f8-cnmwb Total loading time: 0 Render date: 2024-07-16T11:09:57.318Z Has data issue: false hasContentIssue false

28 - Cellular origin of age-related aneuploidy in mammalian oocytes

from Section 5 - Pathology

Published online by Cambridge University Press:  05 October 2013

Ursula Eichenlaub-Ritter
Affiliation:
Faculty of Biology, Institute of Gene Technology/Microbiology, University of Bielefeld, Bielefeld, Germany
Roger Gosden
Affiliation:
Jamestowne Bookworks,Williamsburg, VA, USA
Alan Trounson
Affiliation:
California Institute for Regenerative Medicine
Roger Gosden
Affiliation:
Center for Reproductive Medicine and Infertility, Cornell University, New York
Ursula Eichenlaub-Ritter
Affiliation:
Universität Bielefeld, Germany
Get access

Summary

Introduction

Separation of chromosomes in meiosis is a well-guarded process such that errors in chromosome segregation are rare events. For instance, only 1 in every 100000 divisions in yeast is associated with non-disjunction. Aneuploidy in germ cells of mammals like the mouse is generally much higher, in the range 0.5–1% [1]. Furthermore, there is a gender-specific difference in susceptibility to meiotic errors during germ cell formation in mammals, particularly in humans. On average, only 1–4% of sperm in healthy men have numerical chromosomal aberrations, while on average about 20% of all human meiosis II oocytes are aneuploid [1–4]. The correlation between the incidence of the birth of a trisomic child with Down's syndrome and maternal age was first recognized in 1933 by Penrose [5], confirmed by chromosomal analysis of spontaneous abortions and live births and, since the introduction of assisted reproduction, by evidence from polar bodies, oocytes, and embryos (e.g., [3, 6–8]). However, the cause(s) of the extraordinary susceptibility of aging oocytes to meiotic errors was obscure until recently.

Meiotic stages at which errors may occur

In typical mitosis there is division of sister chromatids derived from replication of each chromosome, and each pair therefore carries the same alleles along their arms (Figure 28.1A). In contrast, in meiosis I the two originally paternally and maternally derived homologs, each containing two sister chromatids, separate during first meiotic division (termed reductional division, Figure 28.1B, Ciii). They are normally physically attached to each other by at least one chiasma from recombination between sister chromatids of parental homologs (indicated by X in Figure 28.1B, Ci, Ciii). Chiasmata are held in place by cohesion between sister chromatid arms and centromeres (Figure 28.1Ci–Ciii) placed on chromatids before S-phase (Figure 28.1Ci), which is maintained until anaphase I (Figure 28.1Ciii, B) (reviewed in [9]). The paternal and maternal alleles along chromatid arms of recombined chromosomes switch left and right of a chiasma or exchange (indicated by different coloring in Figure 28.1B, Ci, E–J). Hence, the distribution of polymorphisms can be used to trace the origin and recombinational history of a chromosome in a zygote or child.

Type
Chapter
Information
Biology and Pathology of the Oocyte
Role in Fertility, Medicine and Nuclear Reprograming
, pp. 330 - 345
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Pacchierotti, F, Adler, ID, Eichenlaub-Ritter, U, et al. Gender effects on the incidence of aneuploidy in mammalian germ cells. Environ Res 2007; 104: 46–69.CrossRefGoogle ScholarPubMed
Templado, C, Vidal, F, Estop, A.Aneuploidy in human spermatozoa. Cytogenet Genome Res 2011; 133: 91–9.CrossRefGoogle ScholarPubMed
Hassold, T, Hall, H, Hunt, P.The origin of human aneuploidy: where we have been, where we are going. Hum Mol Genet 2007; 16(Spec No. 2): R203–8.CrossRefGoogle ScholarPubMed
Hassold, T, Hunt, P.Maternal age and chromosomally abnormal pregnancies: what we know and what we wish we knew. Curr Opin Pediatr 2009; 21(6): 703–8.CrossRefGoogle ScholarPubMed
Penrose, LS.The relative effects of paternal and maternal age in mongolism. J Genet 1933; 27: 219–24.CrossRefGoogle Scholar
Pellestor, F, Andréo, B, Arnal, F, et al. Maternal aging and chromosomal abnormalities: new data drawn from in vitro unfertilized human oocytes. Hum Genet 2003; 112: 195–203.Google ScholarPubMed
Handyside, AH, Montag, M, Magli, MC, et al. Multiple meiotic errors caused by predivision of chromatids in women of advanced maternal age undergoing in vitro fertilisation. Eur J Hum Genet 2012; 349: 795–808. .Google Scholar
Gabriel, AS, Thornhill, AR, Ottolini, CS, et al. Array comparative genomic hybridisation on first polar bodies suggests that non-disjunction is not the predominant mechanism leading to aneuploidy in humans. J Med Genet 2011; 48: 433–7.CrossRefGoogle Scholar
Nasmyth, K.Cohesin: a catenase with separate entry and exit gates?Nat Cell Biol 2011; 13: 1170–7.CrossRefGoogle ScholarPubMed
Kudo, NR, Anger, M, Peters, AH, et al. Role of cleavage by separase of the Rec8 kleisin subunit of cohesin during mammalian meiosis I. J Cell Sci 2009; 122: 2686–98.CrossRefGoogle ScholarPubMed
Ishiguro, T, Tanaka, K, Sakuno, T, et al. Shugoshin-PP2A counteracts casein-kinase-1-dependent cleavage of Rec8 by separase. Nat Cell Biol 2010; 12: 500–6.CrossRefGoogle ScholarPubMed
Tanno, Y, Kitajima, TS, Honda, T, et al. Phosphorylation of mammalian Sgo2 by Aurora B recruits PP2A and MCAK to centromeres. Genes Dev 2010; 24: 2169–79.CrossRefGoogle ScholarPubMed
Xu, Z, Cetin, B, Anger, M, et al. Structure and function of the PP2A-shugoshin interaction. Mol Cell 2009; 35: 426–41.CrossRefGoogle ScholarPubMed
Sakuno, T, Tada, K, Watanabe, Y.Kinetochore geometry defined by cohesion within the centromere. Nature 2009; 458: 852–8.CrossRefGoogle ScholarPubMed
Eichenlaub-Ritter, U, Staubach, N, Trapphoff, T.Chromosomal and cytoplasmic context determines predisposition to maternal age-related aneuploidy: brief overview and update on MCAK in mammalian oocytes. Biochem Soc Trans 2010; 38: 1681–6.CrossRefGoogle ScholarPubMed
Henderson, SA, Edwards, RG.Chiasma frequency and maternal age in mammals. Nature 1968; 218: 22–8.CrossRefGoogle ScholarPubMed
Nicolaidis, P, Petersen, MB. Origin and mechanisms of non-disjunction in human autosomal trisomies. Hum Reprod 1998; 13: 313–19.CrossRefGoogle ScholarPubMed
Bugge, M, Collins, A, Hertz, JM, et al. Non-disjunction of chromosome 13. Hum Mol Genet 2007; 16: 2004–10.CrossRefGoogle ScholarPubMed
Oliver, TR, Feingold, E, Yu, K, et al. New insights into human nondisjunction of chromosome 21 in oocytes. PLoS Genet 2008; 4: e1000033.CrossRefGoogle ScholarPubMed
Hussin, J, Roy-Gagnon, MH, Gendron, R, et al. Age-dependent recombination rates in human pedigrees. PLoS Genet 2011; 7: e1002251.CrossRefGoogle ScholarPubMed
Kong, A, Barnard, J, Gudbjartsson, DF, et al. Recombination rate and reproductive success in humans. Nat Genet 2004; 36: 1203–6.CrossRefGoogle ScholarPubMed
Hultén, MA, Patel, S, Jonasson, J, et al. On the origin of the maternal age effect in trisomy 21 Down syndrome: the Oocyte Mosaicism Selection model. Reproduction 2010; 139: 1–9.CrossRefGoogle ScholarPubMed
Vialard, F, Lombroso, R, Bergere, M, et al. Oocyte aneuploidy mechanisms are different in two situations of increased chromosomal risk: older patients and patients with recurrent implantation failure after in vitro fertilization. Fertil Steril 2007; 87: 1333–9.CrossRefGoogle ScholarPubMed
Morelli, MA, Cohen, PE. Not all germ cells are created equal: aspects of sexual dimorphism in mammalian meiosis. Reproduction 2005; 130: 761–81.CrossRefGoogle ScholarPubMed
Hunt, PA, Hassold, TJ. Sex matters in meiosis. Science 2002; 296: 2181–3.CrossRefGoogle ScholarPubMed
Yuan, L, Liu, JG, Hoja, MR, et al. Female germ cell aneuploidy and embryo death in mice lacking the meiosis-specific protein SCP3. Science 2002; 296: 1115–18.CrossRefGoogle ScholarPubMed
Bolor, H, Mori, T, Nishiyama, S, et al. Mutations of the SYCP3 gene in women with recurrent pregnancy loss. Am J Hum Genet 2009; 84: 14–20.CrossRefGoogle ScholarPubMed
Mizutani, E, Suzumori, N, Ozaki, Y, et al. SYCP3 mutation may not be associated with recurrent miscarriage caused by aneuploidy. Hum Reprod 2011; 26: 1259–66.CrossRefGoogle Scholar
Revenkova, E, Herrmann, K, Adelfalk, C, et al. Oocyte cohesin expression restricted to predictyate stages provides full fertility and prevents aneuploidy. Curr Biol 2010; 20: 1529–33.CrossRefGoogle ScholarPubMed
Hodges, CA, Revenkova, E, Jessberger, R, et al. SMC1beta-deficient female mice provide evidence that cohesins are a missing link in age-related nondisjunction. Nat Genet 2005; 37: 1351–5.CrossRefGoogle ScholarPubMed
Brunet, S, Dumont, J, Lee, KW, et al. Meiotic regulation of TPX2 protein levels governs cell cycle progression in mouse oocytes. PLoS One 2008; 3:3338.CrossRefGoogle ScholarPubMed
Breuer, M, Kolano, A, Kwon, M, et al. HURP permits MTOC sorting for robust meiotic spindle bipolarity, similar to extra centrosome clustering in cancer cells. J Cell Biol 2010; 191: 1251–60.CrossRefGoogle ScholarPubMed
Eichenlaub-Ritter, U, Vogt, E, Yin, H, et al. Spindles, mitochondria and redox potential in ageing oocytes. Reprod Biomed Online 2004; 8: 45–58.CrossRefGoogle ScholarPubMed
Battaglia, DE, Goodwin, P, Klein, A, et al. Influence of maternal age on meiotic spindle assembly in oocytes from naturally cycling women. Hum Reprod 1996; 11: 2217–22.CrossRefGoogle ScholarPubMed
Hamatani, T, Falco, G, Carter, MG, et al. Age-associated alteration of gene expression patterns in mouse oocytes. Hum Mol Genet 2004; 13: 2263–78.CrossRefGoogle ScholarPubMed
Steuerwald, N, Cohen, J, Herrera, RJ, et al. Association between spindle assembly checkpoint expression and maternal age in human oocytes. Mol Hum Reprod 2001; 7: 49–55.CrossRefGoogle ScholarPubMed
Steuerwald, NM, Bermúdez, MG, Wells, D, et al. Maternal age-related differential global expression profiles observed in human oocytes. Reprod Biomed Online 2007; 14: 700–8.CrossRefGoogle ScholarPubMed
Pan, H, O’Brien, MJ, Wigglesworth, K, et al. Transcript profiling during mouse oocyte development and the effect of gonadotropin priming and developmentin vitro. Dev Biol 2005; 286: 493–506.CrossRefGoogle ScholarPubMed
Grøndahl, ML, Yding, Andersen C, Bogstad, J, et al. Gene expression profiles of single human mature oocytes in relation to age. Hum Reprod 2010; 25: 957–68.CrossRefGoogle Scholar
Fragouli, E, Bianchi, V, Patrizio, P, et al. Transcriptomic profiling of human oocytes: association of meiotic aneuploidy and altered oocyte gene expression. Mol Hum Reprod 2010; 16: 570–82.CrossRefGoogle ScholarPubMed
Kitajima, TS, Ohsugi, M, Ellenberg, J.Complete kinetochore tracking reveals error-prone homologous chromosome biorientation in mammalian oocytes. Cell 2011; 146: 568–81.CrossRefGoogle ScholarPubMed
Lane, SI, Yun, Y, Jones, KT. Timing of anaphase-promoting complex activation in mouse oocytes is predicted by microtubule-kinetochore attachment but not by bivalent alignment or tension. Development 2012; 139: 1947–55.CrossRefGoogle ScholarPubMed
Avo, Santos M, van de Werken, C, de Vries, M, et al. A role for Aurora C in the chromosomal passenger complex during human preimplantation embryo development. Hum Reprod 2011; 26: 1868–81.Google Scholar
Vogt, E, Kipp, A, Eichenlaub-Ritter, U. Aurora kinase B, epigenetic state of centromeric heterochromatin and chiasma resolution in oocytes. Reprod Biomed Online 2009; 19: 352–68.CrossRefGoogle ScholarPubMed
Vogt, E, Sanhaji, M, Klein, W, et al. MCAK is present at centromeres, midspindle and chiasmata and involved in silencing of the spindle assembly checkpoint in mammalian oocytes. Mol Hum Reprod 2010; 16: 665–84.CrossRefGoogle ScholarPubMed
Illingworth, C, Pirmadjid, N, Serhal, P, et al. MCAK regulates chromosome alignment but is not necessary for preventing aneuploidy in mouse oocyte meiosis I. Development 2010; 137: 2133–8.CrossRefGoogle Scholar
Musacchio, A.Spindle assembly checkpoint: the third decade. Philos Trans R Soc Lond B Biol Sci 2011; 366: 3595–604.CrossRefGoogle ScholarPubMed
Homer, HA, McDougall, A, Levasseur, M, et al. Mad2 prevents aneuploidy and premature proteolysis of cyclin B and securin during meiosis I in mouse oocytes. Genes Dev 2005; 19: 202–7.CrossRefGoogle ScholarPubMed
Eichenlaub-Ritter, U, Boll, I.Nocodazole sensitivity, age-related aneuploidy, and alterations in the cell cycle during maturation of mouse oocytes. Cytogenet Cell Genet 1989; 52: 170–6.CrossRefGoogle ScholarPubMed
Volarcik, K, Sheean, L, Goldfarb, J, et al. The meiotic competence of in-vitro matured human oocytes is influenced by donor age: evidence that folliculogenesis is compromised in the reproductively aged ovary. Hum Reprod 1998; 13: 154–60.CrossRefGoogle ScholarPubMed
Seli, E, Robert, C, Sirard, MA. OMICS in assisted reproduction: possibilities and pitfalls. Mol Hum Reprod 2010; 16: 513–30.CrossRefGoogle ScholarPubMed
Salisbury, J, Hutchison, KW, Wigglesworth, K, et al. Probe-level analysis of expression microarrays characterizes isoform-specific degradation during mouse oocyte maturation. PLoS One 2009; 4: e7479.CrossRefGoogle ScholarPubMed
Baker, DJ, Jeganathan, KB, Cameron, JD, et al. BubR1 insufficiency causes early onset of aging-associated phenotypes and infertility in mice. Nat Genet 2004; 36: 744–9.CrossRefGoogle ScholarPubMed
Li, M, Li, S, Yuan, J, et al. Bub3 is a spindle assembly checkpoint protein regulating chromosome segregation during mouse oocyte meiosis. PLoS One 2009; 4: e7701.CrossRefGoogle ScholarPubMed
Hached, K, Xie, SZ, Buffin, E, et al. Mps1 at kinetochores is essential for female mouse meiosis I. Development 2011; 138: 2261–71.CrossRefGoogle ScholarPubMed
LeMaire-Adkins, R, Radke, K, Hunt, PA. Lack of checkpoint control at the metaphase/anaphase transition: a mechanism of meiotic nondisjunction in mammalian females. J Cell Biol 1997; 139:1611–19.CrossRefGoogle ScholarPubMed
Kouznetsova, A, Lister, L, Nordenskjold, M, et al. Bi-orientation of achiasmatic chromosomes in meiosis I oocytes contributes to aneuploidy in mice. Nat Genet 2007; 39: 966–8.CrossRefGoogle ScholarPubMed
Vogt, E, Kirsch-Volders, M, Parry, J, et al. Spindle formation, chromosome segregation and the spindle checkpoint in mammalian oocytes and susceptibility to meiotic error. Mutat Res 2008; 651: 14–29.CrossRefGoogle ScholarPubMed
Pacchierotti, F, Eichenlaub-Ritter, U.Environmental hazard in the aetiology of somatic and germ cell aneuploidy. Cytogenet Genome Res 2011; 133: 254–68.CrossRefGoogle ScholarPubMed
Hoffmann, S, Maro, B, Kubiak, JZ, et al. A single bivalent efficiently inhibits cyclin B1 degradation and polar body extrusion in mouse oocytes indicating robust SAC during female meiosis I. PLoS One 2011; 6: e27143.CrossRefGoogle ScholarPubMed
Kolano, A, Brunet, S, Silk, AD, et al. Error-prone mammalian female meiosis from silencing the spindle assembly checkpoint without normal interkinetochore tension. Proc Natl Acad Sci USA 2012: 109: E1858–67. .CrossRefGoogle ScholarPubMed
Nagaoka, SI, Hodges, CA, Albertini, DF, et al. Oocyte-specific differences in cell-cycle control create an innate susceptibility to meiotic errors. Curr Biol 2011; 21: 651–7.CrossRefGoogle ScholarPubMed
Duncan, FE, Chiang, T, Schultz, RM, et al. Evidence that a defective spindle assembly checkpoint is not the primary cause of maternal age-associated aneuploidy in mouse eggs. Biol Reprod 2009; 81: 768–76.CrossRefGoogle Scholar
Eichenlaub-Ritter, U, Wieczorek, M, Lüke, S, et al. Age related changes in mitochondrial function and new approaches to study redox regulation in mammalian oocytes in response to age or maturation conditions. Mitochondrion 2011; 11: 783–96.CrossRefGoogle ScholarPubMed
Feeney, KM, Wasson, CW, Parish, JL. Cohesin: a regulator of genome integrity and gene expression. Biochem J 2010; 428: 147–61.CrossRefGoogle ScholarPubMed
Chiang, T, Schultz, RM, Lampson, MA. Age-dependent susceptibility of chromosome cohesion to premature separase activation in mouse oocytes. Biol Reprod 2011; 85: 1279–83.CrossRefGoogle ScholarPubMed
Chiang, T, Duncan, FE, Schindler, K, et al. Evidence that weakened centromere cohesion is a leading cause of age-related aneuploidy in oocytes. Curr Biol 2010; 20: 1522–8.CrossRefGoogle ScholarPubMed
Lister, LM, Kouznetsova, A, Hyslop, LA, et al. Age-related meiotic segregation errors in mammalian oocytes are preceded by depletion of cohesin and Sgo2. Curr Biol 2010; 20: 1511–21.CrossRefGoogle ScholarPubMed
Merriman, JA, Jennings, PC, McLaughlin, EA, et al. Effect of aging on superovulation efficiency, aneuploidy rates, and sister chromatid cohesion in mice aged up to 15 months. Biol Reprod 2012; 86: 49.CrossRefGoogle ScholarPubMed
Tachibana-Konwalski, K, Godwin, J, van der Weyden, L, et al. Rec8-containing cohesin maintains bivalents without turnover during the growing phase of mouse oocytes. Genes Dev 2010 24: 2505–16.CrossRefGoogle ScholarPubMed
Garcia-Cruz, R, Brieno, MA, Roig, I, et al. Dynamics of cohesin proteins REC8, STAG3, SMC1 beta and SMC3 are consistent with a role in sister chromatid cohesion during meiosis in human oocytes. Hum Reprod 2010; 25: 2316–27.CrossRefGoogle ScholarPubMed
Hornak, M, Jeseta, M, Musilova, P, et al. Frequency of aneuploidy related to age in porcine oocytes. PLoS One 2011; 6: e18892.CrossRefGoogle ScholarPubMed
Angell, R.First-meiotic-division nondisjunction in human oocytes. Am J Hum Genet 1997; 61: 23–32.CrossRefGoogle ScholarPubMed
van den Berg, IM, Eleveld, C, van der Hoeven, M, et al. Defective deacetylation of histone 4 K12 in human oocytes is associated with advanced maternal age and chromosome misalignment. Hum Reprod 2011; 26: 1181–90.CrossRefGoogle ScholarPubMed
Cukurcam, S, Betzendahl, I, Michel, G, et al. Influence of follicular fluid meiosis-activating sterol on aneuploidy rate and precocious chromatid segregation in aged mouse oocytes. Hum Reprod 2007; 22: 815–28.CrossRefGoogle ScholarPubMed
Van Blerkom, J, Davis, P, Thalhammer, V. Regulation of mitochondrial polarity in mouse and human oocytes: the influence of cumulus derived nitric oxide. Mol Hum Reprod 2008; 14: 431–44.CrossRef
Yu, Y, Dumollard, R, Rossbach, A, et al. Redistribution of mitochondria leads to bursts of ATP production during spontaneous mouse oocyte maturation. J Cell Physiol 2010; 224: 672–80.CrossRefGoogle ScholarPubMed
de Bruin, JP, Dorland, M, Spek, ER, et al. Age-related changes in the ultrastructure of the resting follicle pool in human ovaries. Biol Reprod 2004; 70: 419–24.CrossRefGoogle ScholarPubMed
Thouas, GA, Trounson, AO, Jones, GM. Effect of female age on mouse oocyte developmental competence following mitochondrial injury. Biol Reprod 2005; 73: 366–73.CrossRefGoogle ScholarPubMed
Tatone, C, Heizenrieder, T, Di Emidio, G, et al. Evidence that carbonyl stress by methylglyoxal exposure induces DNA damage and spindle aberrations, affects mitochondrial integrity in mammalian oocytes and contributes to oocyte ageing. Hum Reprod 2011; 26: 1843–59.CrossRefGoogle ScholarPubMed
Liu, L, Keefe, DL. Defective cohesin is associated with age-dependent misaligned chromosomes in oocytes. Reprod Biomed Online 2008; 1 6: 103–12.CrossRefGoogle Scholar
Selesniemi, K, Lee, HJ, Tilly, JL. Moderate caloric restriction initiated in rodents during adulthood sustains function of the female reproductive axis into advanced chronological age. Aging Cell 2008; 7: 622–9.CrossRefGoogle ScholarPubMed
Liu, J, Liu, M, Ye, X, et al. Delay in oocyte aging in mice by the antioxidant N-acetyl-L-cysteine (NAC). Hum Reprod 2012; 27: 1411–20.CrossRefGoogle Scholar
Wang, Q, Ratchford, AM, Chi, MM, et al. Maternal diabetes causes mitochondrial dysfunction and meiotic defects in murine oocytes. Mol Endocrinol 2009; 23: 1603–12.CrossRefGoogle ScholarPubMed
Li, Q, Miao, DQ, Zhou, P, et al. Glucose metabolism in mouse cumulus cells prevents oocyte aging by maintaining both energy supply and the intracellular redox potential. Biol Reprod 2011; 84: 1111–8.CrossRefGoogle ScholarPubMed
Roberts, R, Iatropoulou, A, Ciantar, D, et al. Follicle-stimulating hormone affects metaphase I chromosome alignment and increases aneuploidy in mouse oocytes maturedin vitro. Biol Reprod 2005; 72: 107–18.CrossRefGoogle ScholarPubMed
Xu, YW, Peng, YT, Wang, B, et al. High follicle-stimulating hormone increases aneuploidy in human oocytes maturedin vitro. Fertil Steril 2011; 95: 99–104.CrossRefGoogle ScholarPubMed
Baart, EB, Martini, E, Eijkemans, MJ, et al. Milder ovarian stimulation for in-vitro fertilization reduces aneuploidy in the human preimplantation embryo: a randomized controlled trial. Hum Reprod 2007; 22: 980–8.CrossRefGoogle ScholarPubMed
Kline, JK, Kinney, AM, Levin, B, et al. Trisomic pregnancy and elevated FSH: implications for the oocyte pool hypothesis. Hum Reprod 2011; 26: 1537–50.CrossRefGoogle ScholarPubMed
Haadsma, ML, Mooij, TM, Groen, H, et al. A reduced size of the ovarian follicle pool is associated with an increased risk of a trisomic pregnancy in IVF-treated women. Hum Reprod 2010; 25: 552–8.CrossRefGoogle ScholarPubMed
Fragouli, E, Bianchi, V, Patrizio, P, et al. Transcriptomic profiling of human oocytes: association of meiotic aneuploidy and altered oocyte gene expression. Mol Hum Reprod 2010; 16: 570–82.CrossRefGoogle ScholarPubMed
Guglielmino, MR, Santonocito, M, Vento, M, et al. TAp73 is downregulated in oocytes from women of advanced reproductive age. Cell Cycle 2011; 10: 3253–6.CrossRefGoogle ScholarPubMed
Demant, M, Trapphoff, T, Fröhlich, T, et al. Vitrification at the pre-antral stage transiently alters inner mitochondrial membrane potential but proteome of in vitro grown and matured mouse oocytes appears unaffected. Hum Reprod 2012; 27: 1096–111.CrossRefGoogle ScholarPubMed
Susiarjo, M, Hassold, TJ, Freeman, E, et al. Bisphenol A exposure in utero disrupts early oogenesis in the mouse. PLoS Genet 2007; 3: e5.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×