Skip to main content Accessibility help
×
Hostname: page-component-6d856f89d9-26vmc Total loading time: 0 Render date: 2024-07-16T08:14:27.925Z Has data issue: false hasContentIssue false

Chapter 28 - Molecular and Cellular Integrity of Cultured Follicles

from Section 8 - In Vitro Follicle Culture

Published online by Cambridge University Press:  27 March 2021

Jacques Donnez
Affiliation:
Catholic University of Louvain, Brussels
S. Samuel Kim
Affiliation:
University of Kansas School of Medicine
Get access

Summary

According to the most recent cancer statistics, more than 870,000 new diagnosis of cancer are expected in the US female population in 2018, with the three most common cancers in women being breast, lung, and colorectal cancers [1].

Several improvements have been made in the early diagnosis and treatment of infant and adults cancer and these advances have resulted in greatly increased life expectancy and chances of survival. Nevertheless, some oncological treatments, although leading to cancer cure rates higher than 90%, have a detrimental effect in the reproductive potential of children and young women, resulting in a population at high-risk of developing premature ovarian insufficiency (POI) and therefore infertility [2].

In order to prevent the risk of facing this outcome, fertility preservation options are offered to these patients in order to protect their fertility potential prior to gonadotoxic treatment. Among the available options, ovarian tissue cryopreservation and transplantation is the only method suitable for prepubertal girls and adult women who require urgent treatment.

Type
Chapter
Information
Fertility Preservation
Principles and Practice
, pp. 323 - 331
Publisher: Cambridge University Press
Print publication year: 2021

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

Smitz, J, Cortvrindt, R. Oocyte in-vitro maturation and follicle culture: current clinical achievements and future directions. Hum Reprod, 1999;14(Suppl 1):145161.Google Scholar
Rodrigues, P, Limback, D, McGinnis, LK, Plancha, CE, Albertini, DF. Oogenesis: prospects and challenges for the future. J Cell Physiol, 2008;216(2):355365.Google Scholar
Rosendahl, M, Andersen, CY, Ernst, E et al. Ovarian function after removal of an entire ovary for cryopreservation of pieces of cortex prior to gonadotoxic treatment: a follow-up study. Hum Reprod, 2008;23(11):24752483.Google Scholar
Sanchez, M, Alama, P, Gadea, B et al. Fresh human orthotopic ovarian cortex transplantation: long-term results. Hum Reprod, 2007;22(3):786791.CrossRefGoogle ScholarPubMed
Senbon, S, Ishii, K, Fukumi, Y, Miyano, T. Fertilization and development of bovine oocytes grown in female SCID mice. Zygote, 2005;13(4):309315.Google Scholar
Telfer, EE, Binnie, JP, McCaffery, FH, Campbell, BK. In vitro development of oocytes from porcine and bovine primary follicles. Mol Cell Endocrinol, 2000;163(1–2):117123.Google Scholar
Van Den Hurk, R, Abir, R, Telfer, EE, Bevers, MM. Primate and bovine immature oocytes and follicles as sources of fertilizable oocytes. Hum Reprod Update, 2000;6(5):457474.Google Scholar
Hutt, KJ, Albertini, DF. An oocentric view of folliculogenesis and embryogenesis. Reprod Biomed Online, 2007;14(6):758764.Google Scholar
Jayawardana, BC, Shimizu, T, Nishimoto, H et al. Hormonal regulation of expression of growth differentiation factor-9 receptor type I and II genes in the bovine ovarian follicle. Reproduction, 2006;131(3):545553.Google Scholar
Walters, KA, Binnie, JP, Campbell, BK, Armstrong, DG, Telfer, EE. The effects of IGF-I on bovine follicle development and IGFBP-2 expression are dose and stage dependent. Reproduction, 2006;131(3):515523.Google Scholar
Blondin, P, Bousquet, D, Twagiramungu, H, Barnes, F, Sirard, MA. Manipulation of follicular development to produce developmentally competent bovine oocytes. Biol Reprod, 2002;66(1):28382843.CrossRefGoogle ScholarPubMed
Chian, RC, Chung, JT, Downey, BR, Tan, SL. Maturational and developmental competence of immature oocytes retrieved from bovine ovaries at different phases of folliculogenesis. Reprod Biomed Online, 2002;4(2):127132.Google Scholar
Fortune, JE, Kito, S, Byrd, DD. Activation of primordial follicles in vitro. J Reprod Fertil, 1999;54:439448.Google Scholar
Harada, M, Miyano, T, Matsumura, K et al. Bovine oocytes from early antral follicles grow to meiotic competence in vitro: effect of FSH and hypoxanthine. Theriogenology, 1997;48(5):743755.CrossRefGoogle ScholarPubMed
Amorim, CA, Van, Langendonckt, A, David, A, Dolmans, MM, Donnez, J. Survival of human pre-antral follicles after cryopreservation of ovarian tissue, follicular isolation and in vitro culture in a calcium alginate matrix. Hum Reprod, 2009;24(1):9299.CrossRefGoogle Scholar
Heise, M, Koepsel, R, Russell, AJ, McGee, EA. Calcium alginate microencapsulation of ovarian follicles impacts FSH delivery and follicle morphology. Reprod Biol Endocrinol, 2005;3:47.Google Scholar
Johnson, LD, Albertini, DF, McGinnis, LK, Biggers, JD. Chromatin organization, meiotic status and meiotic competence acquisition in mouse oocytes from cultured ovarian follicles. J Reprod Fertil, 1995;104(2):277284.Google Scholar
Lenie, S, Cortvrindt, R, Adriaenssens, T, Smitz, J.A reproducible two-step culture system for isolated primary mouse ovarian follicles as single functional units. Biol Reprod, 2004;71(5):17301738.Google Scholar
Loret de, Mola JR, Barnhart, K, Kopf, GS et al. Comparison of two culture systems for the in-vitro growth and maturation of mouse preantral follicles. Clin Exp Obstet Gynecol, 2004;31(1):1519.Google Scholar
Vigo, D, Villani, S, Faustini, M et al. Follicle-like model by granulosa cell encapsulation in a barium alginate-protamine membrane. Tissue Eng, 2005;11(5–6):709714.Google Scholar
Heise, MK, Koepsel, R, McGee, EA, Russell, AJ. Dynamic oxygen enhances oocyte maturation in long-term follicle culture. Tissue Eng Part C Methods, 2009;15(3):323332.CrossRefGoogle ScholarPubMed
Keros, V, Xella, S, Hultenby, K et al. Vitrification versus controlled-rate freezing in cryopreservation of human ovarian tissue. Hum Reprod, 2009;24(7):16701683.CrossRefGoogle ScholarPubMed
Maman, E, Prokopis, K, Levron, J et al. Does controlled ovarian stimulation prior to chemotherapy increase primordial follicle loss and diminish ovarian reserve? An animal study. Hum Reprod, 2009;24(1):206210.CrossRefGoogle ScholarPubMed
Bromfield, JJ, Coticchio, G, Hutt, K et al. Meiotic spindle dynamics in human oocytes following slow-cooling cryopreservation. Hum Reprod, 2009;24(9):21142123.Google Scholar
Fair, T, Hyttel, P, Greve, T. Bovine oocyte diameter in relation to maturational competence and transcriptional activity. Mol Reprod Dev, 1995;42(4):437442.CrossRefGoogle ScholarPubMed
Humblot, P, Holm, P, Lonergan, P et al. Effect of stage of follicular growth during superovulation on developmental competence of bovine oocytes. Theriogenology, 2005;63(4):11491166.CrossRefGoogle ScholarPubMed
Albertini, DF, Combelles, CM, Benecchi, E, Carabatsos, MJ. Cellular basis for paracrine regulation of ovarian follicle development. Reproduction, 2001;121(5):647653.Google Scholar
Feary, ES, Juengel, JL, Smith, P et al. Patterns of expression of messenger RNAs encoding GDF9, BMP15, TGFBR1, BMPR1B, and BMPR2 during follicular development and characterization of ovarian follicular populations in ewes carrying the Woodlands FecX2W mutation. Biol Reprod, 2007;77(6):990998.Google Scholar
Fouladi-Nashta, AA, Campbell, KH. Dissociation of oocyte nuclear and cytoplasmic maturation by the addition of insulin in cultured bovine antral follicles. Reproduction, 2006;131(3):449460.Google Scholar
Iwata, H, Hashimoto, S, Ohota, M et al. Effects of follicle size and electrolytes and glucose in maturation medium on nuclear maturation and developmental competence of bovine oocytes. Reproduction, 2004;127(2):159164.Google Scholar
Pavlok, A, Lapathitis, G, Cech, S et al. Simulation of intrafollicular conditions prevents GVBD in bovine oocytes: a better alternative to affect their developmental capacity after two-step culture. Mol Reprod Dev, 2005;71(2):197208.CrossRefGoogle ScholarPubMed
Albertini, DF. Regulation of meiotic maturation in the mammalian oocyte: Inteplay between exogenous cues and the microtubule cytoskeleton. Bioessays, 1992;14(2):97103.CrossRefGoogle Scholar
Allworth, AE, Albertini, DF. Meiotic maturation in cultured bovine oocytes is accompanied by remodeling of the cumulus cell cytoskeleton. Dev Biol, 1993;158(1):101112.Google Scholar
Combelles, CM, Carabatsos, MJ, Kumar, TR, Matzuk, MM, Albertini, DF. Hormonal control of somatic cell oocyte interactions during ovarian follicle development. Mol Reprod Dev, 2004;69(3):347355.Google Scholar
Berkholtz, CB, Shea, LD, Woodruff, TK. Extracellular matrix functions in follicle maturation. Semin Reprod Med, 2006;24(4):262269.CrossRefGoogle ScholarPubMed
Dong, J, Albertini, DF, Nishimori, K et al. Growth differentiation factor-9 is required during early ovarian folliculogenesis. Nature, 1996;383(6600):531535.CrossRefGoogle ScholarPubMed
Carabatsos, MJ, Elvin, J, Matzuk, MM, Albertini, DF. Characterization of oocyte and follicle development in growth differentiation factor-9-deficient mice. Dev Biol, 1998;204(2):373384.Google Scholar
Albertini, DF. Oocyte–granulosa cell interactions. In Blerkom, JV (ed.) Essential IVF: Reviews of Topical Issues in Clinical In Vitro Fertilization. Boston: Kluwer Academic Publishers. 2002, 4358.Google Scholar
Carabatsos, MJ, Sellitto, C, Goodenough, DA, Albertini, DF. Oocyte–granulosa cell heterologous gap junctions are required for the coordination of nuclear and cytoplasmic meiotic competence. Dev Biol, 2000;226(2):167.Google Scholar
Mattson, BA, Albertini, DF. Oogenesis: chromatin and microtubule dynamics during meiotic prophase. Mol Reprod Dev, 1990;25(4):374383.CrossRefGoogle ScholarPubMed
McGinnis, LK, Kinsey, WH, Albertini, DF. Functions of Fyn kinase in the completion of meiosis in mouse oocytes. Dev Biol, 2009;327(2):280287.Google Scholar
Parrott, JA, Skinner, MK. Direct actions of kit-ligand on theca cell growth and differentiation during follicle development. Endocrinology, 1997;138(9):38193827.Google 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(4):709720.Google Scholar
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(4):671681.Google Scholar

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
×