Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-mp689 Total loading time: 0 Render date: 2024-04-18T07:11:39.273Z Has data issue: false hasContentIssue false

4 - Follicle formation and oocyte death

from Section 2 - Life cycle

Published online by Cambridge University Press:  05 October 2013

Melissa E. Pepling
Affiliation:
Department of Biology, Syracuse University, Syracuse, NY, 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

This chapter will focus on the development of primordial germ cells (PGCs) into oocytes and their subsequent assembly into primordial follicles that is essential for reproductive success (Figure 4.1). In the mouse, PGCs migrate to the genital ridge and begin dividing rapidly by mitosis [1]. These oogonia remain connected, through incomplete cytokinesis, in clusters of synchronously dividing cells known as germline cysts [2]. The oogonia then begin to enter meiosis and arrest in the diplotene stage of prophase I. Around the same time, germ cell cysts begin to break apart [3]. As these cysts separate, many oocytes are lost by apoptosis while others are surrounded by a single layer of granulosa cells, forming primordial follicles [4]. It is believed that improper regulation of cyst breakdown and primordial follicle formation can lead to fertility disorders such as premature ovarian insufficiency and primary amenorrhea, where an early depletion of oocytes leads to infertility [5, 6]. In addition, aberrant regulation of oocyte death as primordial follicles form may be the underlying cause of ovarian dysgerminoma, or germ cell tumors [7]. Little is known about what molecules regulate cyst breakdown, primordial follicle formation, and oocyte death. Elucidation of mechanisms regulating cyst breakdown, oocyte numbers, and primordial follicle formation is important because it will lead to the development of early screening and interventions for infertility and germ cell cancers.

Type
Chapter
Information
Biology and Pathology of the Oocyte
Role in Fertility, Medicine and Nuclear Reprograming
, pp. 38 - 49
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

Peters, H.Migration of gonocytes into the mammalian gonad and their differentiation. Philos Trans R Soc Lond B Biol Sci 1970; 259: 91–101.CrossRefGoogle ScholarPubMed
Pepling, ME, Spradling, AC.Female mouse germ cells form synchronously dividing cysts. Development 1998; 125: 3323–8.Google ScholarPubMed
Pepling, ME, Sundman, EA, Patterson, NL, et al. Differences in oocyte development and estradiol sensitivity among mouse strains. Reproduction 2010; 139: 349–57.CrossRefGoogle ScholarPubMed
Pepling, ME, Spradling, AC.The mouse ovary contains germ cell cysts that undergo programmed breakdown to form follicles. Dev Biol 2001; 234: 339–51.CrossRefGoogle Scholar
Kezele, P, Skinner, MK.Regulation of ovarian primordial follicle assembly and development by estrogen and progesterone: endocrine model of follicle assembly. Endocrinology 2003; 144: 3329–37.CrossRefGoogle ScholarPubMed
Yen, SSC, Jaffe, RB.Reproductive Endocrinology: Physiology, Pathophysiology, and Clinical Management, 3rd edn. Philadelphia: Saunders, 1991.Google Scholar
Berek, JS, Novak, E.Berek & Novak's Gynecology, 14th edn. Philadelphia: Lippincott Williams & Wilkins, 2007.Google Scholar
Monk, M, McLaren, A.X-chromosome activity in foetal germ cells of the mouse. J Embryol Exp Morphol 1981; 63: 75–84.Google ScholarPubMed
Witschi, E.Migration of the germ cells of human embryos from the yolk sac to the primitive gonadal folds. In: Contributions to Embryology,Vol. 32. Washington DC: Carnegie Institution. 1948; 67–80.Google Scholar
de Cuevas, M, Lilly, MA, Spradling, AC.Germline cyst formation in Drosophila. Annu Rev Genet 1997; 31: 405–28.CrossRefGoogle ScholarPubMed
Greenbaum, MP, Yan, W, Wu, MH, et al. TEX14 is essential for intercellular bridges and fertility in male mice. Proc Natl Acad Sci USA 2006; 103: 4982–7.CrossRefGoogle ScholarPubMed
Greenbaum, MP, Iwamori, N, Agno, JE, Matzuk, MM.Mouse TEX14 is required for embryonic germ cell intercellular bridges but not female fertility. Biol Reprod 2009; 80: 449–57.CrossRefGoogle Scholar
Mork, L, Tang, H, Batchvarov, I, Capel, B.Mouse germ cell clusters form by aggregation as well as clonal divisions. Mech Dev 2012; 128: 591–6.CrossRefGoogle ScholarPubMed
Gondos, B, Bhiraleus, P, Hobel, CJ.Ultrastructural observations on germ cells in human fetal ovaries. Am J Obstet Gynecol 1971; 110: 644–52.CrossRefGoogle ScholarPubMed
McLaren, A.Germ and somatic cell lineages in the developing gonad. Mol Cell Endocrinol 2000; 163: 3–9.CrossRefGoogle ScholarPubMed
Bullejos, M, Koopman, P.Germ cells enter meiosis in a rostro-caudal wave during development of the mouse ovary. Mol Reprod Dev 2004; 68: 422–8.CrossRefGoogle Scholar
Menke, DB, Koubova, J, Page, DC.Sexual differentiation of germ cells in XX mouse gonads occurs in an anterior-to-posterior wave. Dev Biol 2003; 262: 303–12.CrossRefGoogle Scholar
Bowles, J, Knight, D, Smith, C, et al. Retinoid signaling determines germ cell fate in mice. Science 2006; 312: 596–600.CrossRefGoogle ScholarPubMed
Koubova, J, Menke, DB, Zhou, Q, et al. Retinoic acid regulates sex-specific timing of meiotic initiation in mice. Proc Natl Acad Sci USA 2006; 103: 2474–9.CrossRefGoogle ScholarPubMed
Baltus, AE, Menke, DB, Hu, YC, et al. In germ cells of mouse embryonic ovaries, the decision to enter meiosis precedes premeiotic DNA replication. Nat Genet 2006; 38: 1430–4.CrossRefGoogle ScholarPubMed
Borum, K.Oogenesis in the mouse. A study of the origin of the mature ova. Exp Cell Res 1961; 45: 39–47.CrossRefGoogle Scholar
Shin, YH, Choi, Y, Erdin, SU, et al. Hormad1 mutation disrupts synaptonemal complex formation, recombination, and chromosome segregation in mammalian meiosis. PLoS Genet 2010; 6: e1001190.CrossRefGoogle ScholarPubMed
Byskov, AG.Differentiation of mammalian embryonic gonad. Physiol Rev 1986; 66: 71–117.CrossRefGoogle ScholarPubMed
Liu, CF, Liu, C, Yao, HH.Building pathways for ovary organogenesis in the mouse embryo. Curr Top Dev Biol 2010; 90: 263–90.CrossRefGoogle ScholarPubMed
Mork, L, Maatouk, DM, McMahon, JA, et al. Temporal differences in granulosa cell specification in the ovary reflect distinct follicle fates in mice. Biol Reprod 2012; 86: 37.CrossRefGoogle ScholarPubMed
Merchant-Larios, H, Chimal-Monroy, J.The ontogeny of primordial follicles in the mouse ovary. Prog Clin Biol Res 1989; 296: 55–63.Google ScholarPubMed
Mazaud, S, Guyot, R, Guigon, CJ, et al. Basal membrane remodeling during follicle histogenesis in the rat ovary: contribution of proteinases of the MMP and PA families. Dev Biol 2005; 277: 403–16.CrossRefGoogle ScholarPubMed
Baker, TG.Oogenesis and ovarian development. In: Balin, H, Glasser, S, eds. Reproductive Biology. Amsterdam: Excerpta Medica. 1972; 398–405.Google Scholar
Pepling, ME.From primordial germ cell to primordial follicle: mammalian female germ cell development. Genesis 2006; 44: 622–32.CrossRefGoogle ScholarPubMed
De Felici, M, Di Carlo, A, Pesce, M, et al. Bcl-2 and Bax regulation of apoptosis in germ cells during prenatal oogenesis in the mouse embryo. Cell Death Differ 1999; 6: 908–15.CrossRefGoogle Scholar
McClellan, KA, Gosden, R, Taketo, T.Continuous loss of oocytes throughout meiotic prophase in the normal mouse ovary. Dev Biol 2003; 258: 334–48.CrossRefGoogle ScholarPubMed
Nandedkar, T, Dharma, S, Modi, D, Dsouza, S.Differential gene expression in transition of primordial to preantral follicles in mouse ovary. Soc Reprod Fertil Suppl 2007; 63: 57–67.Google ScholarPubMed
Coucouvanis, EC, Sherwood, SW, Carswell-Crumpton, C, Spack, EG, Jones, PP.Evidence that the mechanism of prenatal germ cell death in the mouse is apoptosis. Exp Cell Res 1993; 209: 238–47.CrossRefGoogle ScholarPubMed
Lobascio, AM, Klinger, FG, Scaldaferri, ML, Farini, D, De Felici, M.Analysis of programmed cell death in mouse fetal oocytes. Reproduction 2007; 134: 241–52.CrossRefGoogle ScholarPubMed
De Felici, M, Lobascio, AM, Klinger, FG.Cell death in fetal oocytes: many players for multiple pathways. Autophagy 2008; 4: 240–2.CrossRefGoogle ScholarPubMed
Edinger, AL, Thompson, CB.Death by design: apoptosis, necrosis and autophagy. Curr Opin Cell Biol 2004; 16: 663–9.CrossRefGoogle ScholarPubMed
Codogno, P, Meijer, AJ.Autophagy and signaling: their role in cell survival and cell death. Cell Death Differ 2005; 12 Suppl 2: 1509–18.CrossRefGoogle ScholarPubMed
Pesce, M, Farrace, MG, Amendola, A, Piacentini, M, De Felici, M.Stem cell factor regulation of apoptosis in mouse primordial germ cells. In: Tilly, J, Strauss, JF, eds. Cell Death in Reproductive Physiology. New York: Verlag. 1997; 19–31.CrossRefGoogle Scholar
Rodrigues, P, Limback, D, McGinnis, LK, Plancha, CE, Albertini, DF.Multiple mechanisms of germ cell loss in the perinatal mouse ovary. Reproduction 2009; 137: 709–20.CrossRefGoogle ScholarPubMed
Elvin, JA, Yan, C, Matzuk, MM.Oocyte-expressed TGF-beta superfamily members in female fertility. Mol Cell Endocrinol 2000; 159: 1–5.CrossRefGoogle ScholarPubMed
Yan, C, Wang, P, DeMayo, J, et al. Synergistic roles of bone morphogenetic protein 15 and growth differentiation factor 9 in ovarian function. Mol Endocrinol 2001; 15: 854–66.CrossRefGoogle ScholarPubMed
Jefferson, W, Newbold, R, Padilla-Banks, E, Pepling, M.Neonatal genistein treatment alters ovarian differentiation in the mouse: inhibition of oocyte nest breakdown and increased oocyte survival. Biol Reprod 2006; 74: 161–8.CrossRefGoogle ScholarPubMed
Kent, HA.Polyovular follicles and multinucleate ova in the ovaries of young mice. Anat Rec 1960; 137: 521–4.CrossRefGoogle ScholarPubMed
Wang, J, Roy, SK.Growth differentiation factor-9 and stem cell factor promote primordial follicle formation in the hamster: modulation by follicle-stimulating hormone. Biol Reprod 2004; 70: 577–85.CrossRefGoogle ScholarPubMed
Schindler, R, Nilsson, E, Skinner, MK.Induction of ovarian primordial follicle assembly by connective tissue growth factor CTGF. PLoS One 2010; 5: e12979.CrossRefGoogle ScholarPubMed
Soyal, SM, Amleh, A, Dean, J.FIGalpha, a germ cell-specific transcription factor required for ovarian follicle formation. Development 2000; 127: 4645–54.Google ScholarPubMed
Rajkovic, A, Pangas, SA, Ballow, D, Suzumori, N, Matzuk, MM.NOBOX deficiency disrupts early folliculogenesis and oocyte-specific gene expression. Science 2004; 305: 1157–9.CrossRefGoogle ScholarPubMed
Kerr, B, Garcia-Rudaz, C, Dorfman, M, Paredes, A, Ojeda, SR.NTRK1 and NTRK2 receptors facilitate follicle assembly and early follicular development in the mouse ovary. Reproduction 2009; 138: 131–40.CrossRefGoogle ScholarPubMed
Trombly, DJ, Woodruff, TK, Mayo, KE.Suppression of Notch signaling in the neonatal mouse ovary decreases primordial follicle formation. Endocrinology 2009; 150: 1014–24.CrossRefGoogle ScholarPubMed
Di Carlo, A, De Felici, M.A role for E-cadherin in mouse primordial germ cell development. Dev Biol 2000; 226: 209–19.CrossRefGoogle ScholarPubMed
Wang, C, Roy, SK.Expression of E-cadherin and N-cadherin in perinatal hamster ovary: possible involvement in primordial follicle formation and regulation by follicle-stimulating hormone. Endocrinology 2010; 151: 2319–30.CrossRefGoogle ScholarPubMed
Chen, Y, Jefferson, WN, Newbold, RR, Padilla-Banks, E, Estradiol, Pepling ME., progesterone, and genistein inhibit oocyte nest breakdown and primordial follicle assembly in the neonatal mouse ovary in vitro and in vivo. Endocrinology 2007; 148: 3580–90.CrossRefGoogle ScholarPubMed
Karavan, JR, Pepling, ME.Effects of estrogenic compounds on neonatal oocyte development. Reprod Toxicol 2012; 34: 51–6.CrossRefGoogle ScholarPubMed
Chen, Y, Breen, K, Pepling, ME.Estrogen can signal through multiple pathways to regulate oocyte cyst breakdown and primordial follicle assembly in the neonatal mouse ovary. J Endocrinol 2009; 202: 407–17.CrossRefGoogle ScholarPubMed
Uda, M, Ottolenghi, C, Crisponi, L, et al. Foxl2 disruption causes mouse ovarian failure by pervasive blockage of follicle development. Hum Mol Genet 2004; 13: 1171–81.CrossRefGoogle ScholarPubMed
Hahn, KL, Johnson, J, Beres, BJ, Howard, S, Wilson-Rawls, J.Lunatic fringe null female mice are infertile due to defects in meiotic maturation. Development 2005; 132: 817–28.CrossRefGoogle ScholarPubMed
Nilsson, EE, Schindler, R, Savenkova, MI, Skinner, MK.Inhibitory actions of anti-Mullerian hormone (AMH) on ovarian primordial follicle assembly. PLoS One 2011; 6: e20087.CrossRefGoogle ScholarPubMed
Bristol-Gould, SK, Kreeger, PK, Selkirk, CG, et al. Postnatal regulation of germ cells by activin: the establishment of the initial follicle pool. Dev Biol 2006; 298: 132–48.CrossRefGoogle ScholarPubMed
McMullen, ML, Cho, BN, Yates, CJ, Mayo, KE.Gonadal pathologies in transgenic mice expressing the rat inhibin alpha-subunit. Endocrinology 2001; 142: 5005–14.CrossRefGoogle ScholarPubMed
Kimura, F, Bonomi, LM, Schneyer, AL.Follistatin regulates germ cell nest breakdown and primordial follicle formation. Endocrinology 2011; 152: 697–706.CrossRefGoogle ScholarPubMed
Brown, C, LaRocca, J, Pietruska, J, et al. Subfertility caused by altered follicular development and oocyte growth in female mice lacking PKB alpha/Akt1. Biol Reprod 2010; 82: 246–56.CrossRefGoogle ScholarPubMed
Liu, H, Luo, LL, Qian, YS, et al. FOXO3a is involved in the apoptosis of naked oocytes and oocytes of primordial follicles from neonatal rat ovaries. Biochem Biophys Res Commun 2009; 381: 722–7.CrossRefGoogle ScholarPubMed
John, GB, Shirley, LJ, Gallardo, TD, Castrillon, DH.Specificity of the requirement for Foxo3 in primordial follicle activation. Reproduction 2007; 133: 855–63.CrossRefGoogle ScholarPubMed
Rajareddy, S, Reddy, P, Du, C, et al. p27kip1 (cyclin-dependent kinase inhibitor 1B) controls ovarian development by suppressing follicle endowment and activation and promoting follicle atresia in mice. Mol Endocrinol 2007; 21: 2189–202.CrossRefGoogle ScholarPubMed
Adhikari, D, Zheng, W, Shen, Y, et al. Tsc/mTORC1 signaling in oocytes governs the quiescence and activation of primordial follicles. Hum Mol Genet 2010; 19: 397–410.CrossRefGoogle ScholarPubMed
Reddy, P, Liu, L, Adhikari, D, et al. Oocyte-specific deletion of Pten causes premature activation of the primordial follicle pool. Science 2008; 319: 611–13.CrossRefGoogle ScholarPubMed
, Lan ZJ, Xu, X, Cooney, AJ.Differential oocyte-specific expression of Cre recombinase activity in GDF-9-iCre, Zp3cre, and Msx2Cre transgenic mice. Biol Reprod 2004; 71: 1469–74.Google Scholar
Dissen, GA, Romero, C, Hirshfield, AN, Ojeda, SR.Nerve growth factor is required for early follicular development in the mammalian ovary. Endocrinology 2001; 142: 2078–86.CrossRefGoogle ScholarPubMed
Spears, N, Molinek, MD, Robinson, LL, et al. The role of neurotrophin receptors in female germ-cell survival in mouse and human. Development 2003; 130: 5481–91.CrossRefGoogle ScholarPubMed
Benedict, JC, Lin, TM, Loeffler, IK, Peterson, RE, Flaws, JA.Physiological role of the aryl hydrocarbon receptor in mouse ovary development. Toxicol Sci 2000; 56: 382–8.CrossRefGoogle ScholarPubMed
Robles, R, Morita, Y, Mann, KK, et al. The aryl hydrocarbon receptor, a basic helix-loop-helix transcription factor of the PAS gene family, is required for normal ovarian germ cell dynamics in the mouse. Endocrinology 2000; 141: 450–3.CrossRefGoogle ScholarPubMed
Wang, N, Zhang, P, Guo, X, Zhou, Z, Sha, J.Hnrnpk, a protein differentially expressed in immature rat ovarian development, is required for normal primordial follicle assembly and development. Endocrinology 2011; 152: 1024–35.CrossRefGoogle ScholarPubMed
Fuchs, Y, Steller, H.Programmed cell death in animal development and disease. Cell 2011; 147: 742–58.CrossRefGoogle ScholarPubMed
Conradt, B.Genetic control of programmed cell death during animal development. Annu Rev Genet 2009; 43: 493–523.CrossRefGoogle ScholarPubMed
Bergeron, L, Perez, GI, Macdonald, G, et al. Defects in regulation of apoptosis in caspase-2-deficient mice. Genes Dev 1998; 12: 1304–14.CrossRefGoogle ScholarPubMed
Hanoux, V, Pairault, C, Bakalska, M, Habert, R, Livera, G.Caspase-2 involvement during ionizing radiation-induced oocyte death in the mouse ovary. Cell Death Differ 2007; 14: 671–81.CrossRefGoogle ScholarPubMed
Troy, CM, Shelanski, ML.Caspase-2 redux. Cell Death Differ 2003; 10: 101–7.CrossRefGoogle ScholarPubMed
Hsu, SY, Hsueh, AJ.Tissue-specific Bcl-2 protein partners in apoptosis: an ovarian paradigm. Physiol Rev 2000; 80: 593–614.CrossRefGoogle ScholarPubMed
Kim, MR, Tilly, JL.Current concepts in Bcl-2 family member regulation of female germ cell development and survival. Biochim Biophys Acta 2004; 1644: 205–10.CrossRefGoogle Scholar
Cory, S, Adams, JM.The Bcl-2 family: regulators of the cellular life-or-death switch. Nat Rev Cancer 2002; 2: 647–56.CrossRefGoogle ScholarPubMed
Ratts, VS, Flaws, JA, Klop, R, Sorenson, CM, Tilly, JL.Ablation of bcl-2 gene expression decreases the number of oocytes and primordial follicles established in the postnatal female mouse gonad. Endocrinology 1995; 136: 3665–8.CrossRefGoogle Scholar
Veis, DJ, Sorenson, CM, Shutter, JR, Korsmeyer, SJ.Bcl-2-deficient mice demonstrate fulminant lymphoid apoptosis, polycystic kidneys, and hypopigmented hair. Cell 1993; 75: 229–40.CrossRefGoogle ScholarPubMed
Flaws, JA, Hirshfield, AN, Hewitt, JA, Babus, JK, Furth, PA.Effect of bcl-2 on the primordial follicle endowment in the mouse ovary. Biol Reprod 2001; 64: 1153–9.CrossRefGoogle ScholarPubMed
Perez, GI, Robles, R, Knudson, CM, et al. Prolongation of ovarian lifespan into advanced chronological age by Bax-deficiency. Nat Genet 1999; 21: 200–3.CrossRefGoogle ScholarPubMed
Greenfeld, CR, Pepling, ME, Babus, JK, Furth, PA, Flaws, JA.BAX regulates follicular endowment in mice. Reproduction 2007; 133: 865–76.CrossRefGoogle ScholarPubMed
Gawriluk, TR, Hale, AN, Flaws, JA, et al. Autophagy is a cell survival program for female germ cells in the murine ovary. Reproduction 2011; 141: 759–65.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
×