Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-25wd4 Total loading time: 0 Render date: 2024-04-27T21:04:29.965Z Has data issue: false hasContentIssue false

Chapter 32 - Survival of Primordial Follicles

Tips and Tricks

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 European Society of Human Reproduction and Embryology (ESHRE) Working Group on Oocyte Cryopreservation [], between 2010 and 2014, cancer patients in Europe underwent similar numbers of ovarian tissue cryopreservation procedures (~4,400) and oocyte cryopreservation procedures (~3,800). This milestone in the field of ovarian tissue cryopreservation is responsible for a new surge of interest in primordial follicles and their requirements for survival and development, as they represent more than 90% of the follicle population to be cryopreserved. When patients cannot have their frozen-thawed ovarian tissue reimplanted due to the risk of transmission of malignant cells, these follicles can be isolated and grafted inside a bioengineered ovary (see Chapter 36) or grown in vitro (see Chapter 31). We will focus on the latter strategy, more specifically on the main elements that primordial follicles need to survive in vitro.

Type
Chapter
Information
Fertility Preservation
Principles and Practice
, pp. 364 - 380
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

The ESHRE Working Group on Oocyte Cryopreservation in Europe, Shenfield, F, de Mouzon, J, Scaravelli, G et al. Oocyte and ovarian tissue cryopreservation in European countries: statutory background, practice, storage and use. Hum Reprod Open, 2017;2017:hox003.Google Scholar
Depalo, R, Nappi, L, Loverro, G et al. Evidence of apoptosis in human primordial and primary follicles. Hum Reprod, 2003;18:26782682.Google Scholar
McLaughlin, EA, McIver, SC. Awakening the oocyte: controlling primordial follicle development. Reproduction, 2009;137:111.Google Scholar
Otala, M, Erkkilä, K, Tuuri, T et al. Cell death and its supression in human ovarian tissue culture. Mol Hum Reprod, 2002;8:228236.Google Scholar
Isachenko, V, Montag, M, Isachenko, E et al. Effective method for in-vitro culture of cryopreserved human ovarian tissue. Reprod Biomed Online, 2006;13:228234.CrossRefGoogle ScholarPubMed
Sadeu, JC, Smitz, J. Growth differentiation factor-9 and anti-Müllerian hormone expression in cultured human follicles from frozen–thawed ovarian tissue. Reprod Biomed Online, 2008;17:537548.Google Scholar
McLaughlin, M, Albertini, DF, Wallace, WHB, Anderson, RA, Telfer, EE. Metaphase II oocytes from human unilaminar follicles grown in a multi-step culture system. Mol Hum Reprod, 2018:24:135142.Google Scholar
Hovatta, O, Silye, R, Abir, R, Krausz, T, Winston, RML. Extracellular matrix improves survival of both stored and fresh human primordial and primary ovarian follicles in long-term culture. Hum Reprod, 1997;12:10321036.Google Scholar
Wright, CS, Hovatta, O, Margara, R et al. Effects of follicle-stimulating hormone and serum substitution on the in-vitro growth of human ovarian follicles. Hum Reprod, 1999;14:15551562.Google Scholar
Hovatta, O, Wright, C, Krausz, T, Hardy, K, Winston, RML. Human primordial, primary and secondary ovarian follicles in long-term culture: effect of partial isolation. Hum Reprod, 1999;14:25192524.Google Scholar
Louhio, H, Hovatta, O, Sjöberg, J, Tuuri, T. The effects of insulin, and insulin-like growth factors I and II on human ovarian follicles in long-term culture. Mol Hum Reprod, 2000;6:694698.Google Scholar
Rahimi, G, Isachenko, E, Sauer, H et al. Measurement of apoptosis in long-term cultures of human ovarian tissue. Reproduction, 2001;122:657663.Google Scholar
Hreinsson, JG, Scott, JE, Rasmussen, C et al. Growth differentiation factor-9 promotes the growth, development, and survival of human ovarian follicles in organ culture. J Clin Endocrinol Metab, 2002;87:316321.Google Scholar
Isachenko, E, Isachenko, V, Rahimi, G, Nawroth, F. Cryopreservation of human ovarian tissue by direct plunging into liquid nitrogen. Eur J Obstet Gynecol Reprod Biol, 2003;108:186193.Google Scholar
Biron-Shental, T, Fisch, B, Van Den Hurk, R et al. Survival of frozen-thawed human ovarian fetal follicles in long-term organ culture. Fertil Steril, 2004;81:716719.Google Scholar
Otala, M, Mäkinen, S, Tuuri, T et al. Effects of testosterone, dihydrotestosterone, and 17β-estradiol on human ovarian tissue survival in culture. Fertil Steril, 2004;82:10771085.Google Scholar
Scott, JE, Carlsson, IB, Bavister, BD, Hovatta, O. Human ovarian tissue cultures: extracellular matrix composition, coating density and tissue dimensions. Reprod Biomed Online, 2004;9:287293.Google Scholar
Scott, JE, Zhang, P, Hovatta, O. benefits of 8-bromo-guanosine 3’,5’-cyclic monophosphate (8-br-cGMP) in human ovarian cortical tissue culture. Reprod Biomed Online, 2004;8:319324.Google Scholar
Zhang, P, Louhio, H, Tuuri, T et al. In vitro effect of cyclic adenosine 3’,5’-monophosphate (cAMP) on early human ovarian follicles. J Assist Reprod Genet, 2004;21:301306.Google Scholar
Schmidt, KLT, Kryger-Baggasen, N, Byskov, AG, Andersen, CY. Anti-Müllerian hormone initiates growth of human primordial follicles in vitro. Mol Cell Endocrinol, 2005;234:8793.Google Scholar
Carlsson, IB, Laitinen, MPE, Scott, JE et al. Kit ligand and c-kit are expressed during early human ovarian follicular development and their interaction is required for the survival of follicles in long-term culture. Reproduction, 2006;131:641649.Google Scholar
Carlsson, IB, Scott, JE, Visser, JA et al. Anti-Müllerian hormone inhibits initiation of growth of human primordial ovarian follicles in vitro. Hum Reprod, 2006;21:22232227.Google Scholar
Sadeu, JC, Cortvrindt, R, Ron-El, R, Kasterstein, E, Smitz, J. Morphological and ultrastructural evaluation of cultured frozen-thawed human fetal ovarian tissue. Fertil Steril, 2006;85:11301141.Google Scholar
Isachenko, V, Isachenko, E, Reinsberg, J et al. Cryopreservation of human ovarian tissue: comparison of rapid and conventional freezing. Cryobiology, 2007;55:261268.Google Scholar
Morimoto, Y, Oku, Y, Sonoda, M et al. High oxygen atmosphere improves human follicle development in organ cultures of ovarian cortical tissues in vitro. Hum Reprod, 2007;22:31703177.Google Scholar
Telfer, EE, McLaughlin, M, Ding, C, Joo Thong, K. A two-step serum-free culture system supports development of human oocytes from primordial follicles in the presence of activin. Hum Reprod, 2008;23:11511158.Google Scholar
Garor, R, Abir, R, Erman, A et al. Effects of basic fibroblast growth factor on in vitro development of human ovarian primordial follicles. Fertil Steril, 2009;91:19671975.Google Scholar
Kedem, A, Hourvitz, A, Fisch, B et al. Alginate scaffold for organ culture of cryopreserved-thawed human ovarian cortical follicles. J Assist Reprod Genet, 2011;28:761769.Google Scholar
Kedem, A, Fisch, B, Garor, R et al. Development of human primordial follicles in vitro, with seemingly more beneficial effects of GDF9. J Clin Endocrinol Metab, 2011;96:E1246-E1254.Google Scholar
McLaughlin, M, Patrizio, P, Kayisli, U et al. mTOR kinase inhibition results in oocyte loss characterized by empty follicles in human ovarian cortical strips cultured in vitro. Fertil Steril, 2011;96:11541159.Google Scholar
Lerer-Serfaty, G, Samara, N, Risch, B et al. Attempted application of bioengineered/biosynthetic supporting matrices with phosphatidylinositol-trisphosphate-enhancing substances to organ culture of human primordial follicles. J Assist Reprod Genet, 2013;30:12791288.CrossRefGoogle ScholarPubMed
Anderson, RA, McLaughlin, M, Wallace, WHB, Albertini, DF, Telfer, EE. The immature human ovary shows loss of abnormal follicles and increasing follicle developmental competence through childhood and adolescence. Hum Reprod, 2014;29:97106.Google Scholar
Laronda, MM, Duncan, FE, Hornick, JE et al. Alginate encapsulation supports the growth and differentiation of human primordial follicles within ovarian cortical tissue. J Assist Reprod Genet, 2014;31:10131028.Google Scholar
Lande, Y, Fisch, B, Tsur, A et al. Short-term exposure of human ovarian follicles to cyclophosphamide metabolites seems to promote follicular activation in vitro. Reprod Biomed Online, 2017;34:104114.Google Scholar
Younis, AJ, Lerer-Serfaty, G, Stav, D et al. Extracellular-like matrices and leukemia inhibitory factor for in vitro culture of human primordial follicles. Reprod Fertil Dev, 2017;29:19821994.Google Scholar
Talevi, R, Sudhakaran, S, Barbato, V et al. Is oxygen availability a limiting factor for in vitro folliculogenesis? PLoS One, 2018;13:e0192501.Google Scholar
Schubert, B, Smitz, J. In vitro culture of human primordial follicles. In Chian, RC, Quinn, P (eds.) Fertility Cryopreservation. Cambridge: Cambridge University. 2010, 200212.Google Scholar
Da Silva-Buttkus, P, Marcelli, G, Franks, S, Stark, J, Hardy, K. Inferring biological mechanisms from spatial analysis: prediction of a local inhibitor in the ovary. Proc Natl Acad Sci, 2009;106:456461.Google Scholar
Chiti, MC, Dolmans, MM, Mortiaux, L et al. A novel fibrin-based artificial ovary prototype resembling human ovarian tissue in terms of architecture and rigidity. J Assist Reprod Genet, 2018;35:4148.Google Scholar
Schmidt, KL, Byskov, AG, Nyboe, Andersen A, Müller, J, Yding, Andersen C. Density and distribution of primordial follicles in single pieces of cortex from 21 patients and in individual pieces of cortex from three entire human ovaries. Hum Reprod, 2003;18:11581164.Google Scholar
Chambers, EL, Gosden, RG, Yap, C, Picton, HM. In situ identification of follicles in ovarian cortex as a tool for quantifying follicle density, viability and developmental potential in strategies to preserve female fertility. Hum Reprod, 2010;25:25592568.Google Scholar
Peters, ITA, Stegehuis, PL, Peek, R et al. Noninvasive detection of metastases and follicle density in ovarian tissue using full-field optical coherence tomography. Clin Cancer Res, 2016;22:55065513.Google Scholar
Guérard, M, Zeller, A, Singer, T, Gocke, E. In vitro genotoxicity of neutral red after photo-activation and metabolic activation in the Ames test, the micronucleous test and the comet assay. Mutat Res, 2012;746:1520.Google Scholar
Fischer, BB, Krieger-Liszkay, A, Eggen, RIL. Photosensitizers neutral red (type I) and rose bengal (type II) cause light-dependent toxicity in Chlamydomonas reinhardtii and induce the Gpxh gene via increased singlet oxygen formation. Environ Sci Technol, 2004;38:63076313.Google Scholar
Langbeen, A, Jorssen, EPA, Granata, N et al. Effects of neutral red assisted viability assessment on the cryotolerance of isolated bovine preantral follicles. J Assist Reprod Genet, 2014;31:17271736.CrossRefGoogle ScholarPubMed
Bulgarelli, DL, Ting, AY, Gordon, BJ, de Sá Rosa-e-Silva, ACJ, Zelinski, MB. Development of macaque secondary follicles exposed to neutral red prior to 3-dimensional culture. J Assist Reprod Genet, 2018;35:7179.Google Scholar
Abir, R, Fisch, B, Nitke, S et al. Morphological study of fully and partially isolated early human follicles. Fertil Steril, 2001;75:141146.Google Scholar
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;24:9299.Google Scholar
Vanacker, J, Camboni, A, Dath, C et al. Enzymatic isolation of human primordial and primary ovarian follicles with Liberase DH: protocol for application in a clinical setting. Fertil Steril, 2011;96:379–383.e3.Google Scholar
Camboni, A, Van Langendonckt, A, Donnez, J et al. Alginate beads as a tool to handle, cryopreserve and culture isolated human primordial/primary follicles. Cryobiology, 2013;67:6469.Google Scholar
Vanacker, J, Luyckx, V, Amorim, C et al. Should we isolate human preantral follicles before or after cryopreservation of ovarian tissue? Fertil Steril, 2013;99:13631368.e2.Google Scholar
Abir, R, Roizman, P, Fisch, B et al. Pilot study of isolated early human follicles cultured in collagen gels for 24 hours. Hum Reprod, 1999;14:12991301.Google Scholar
Dolmans, MM, Michaux, N, Camboni, A et al. Evaluation of Liberase, a purified enzyme blend, for the isolation of human primordial and primary ovarian follicles. Hum Reprod, 2006;21:413420.Google Scholar
Schmidt, VM, Isachenko, V, Rappl, G et al. Comparison of the enzymatic efficiency of Liberase TM and tumor dissociation enzyme: effect on the viability of cells digested from fresh and cryopreserved human ovarian cortex. Reprod Biol Endocrinol, 2018;16:PMC5985056.Google Scholar
Chiti, MC, Dolmans, MM, Hobeika, M et al. A modified and tailored human follicle isolation procedure improves follicle recovery and survival. J Ovarian Res, 2017;10:71.Google Scholar
Roy, SK, Treacy, BJ. Isolation and long-term culture of human preantral follicles. Fertil Steril, 1993;59:783790.Google Scholar
Fabbri, R, Pasquinelli, G, Keane, D et al. Culture of cryopreserved ovarian tissue: state of the art in 2008. Fertil Steril, 2009;91:16191629.Google Scholar
Paulini, F, Vilela, JM, Chiti, MC et al. Survival and growth of human preantral follicles after cryopreservation of ovarian tissue, follicle isolation and short-term xenografting. Reprod Biomed Online, 2016;33:425432.Google Scholar
Kim, CH, Cheon, YP, Lee, YJ et al. The effect of fibroblast co-culture on in vitro maturation of mouse preantral follicles. Dev Reprod, 2013;17:269274.Google Scholar
Tagler, D, Tu, T, Smith, RM et al. Embryonic fibroblasts enable the culture of primary ovarian follicles within alginate hydrogels. Tissue Eng Part A, 2012;18:12291238.CrossRefGoogle ScholarPubMed
Tingen, CM, Kiesewetter, SE, Jozefik, J et al. A macrophage and theca cell-enriched stromal cell population influences growth and survival of immature murine follicles in vitro. Reproduction, 2011;141:809820.Google Scholar
Hornick, JE, Duncan, FE, Shea, LD, Woodruff, TK. Multiple follicle culture supports primary follicle growth through paracrine-acting signals. Reproduction, 2013;145:1932.Google Scholar
Guzel, Y, Oktem, O. Understanding follicle growth in vitro: Are we getting closer to obtaining mature oocytes from in vitro-grown follicles in human? Mol Reprod Dev, 2017;84:544559.Google Scholar
Picton, HM, Gosden, RG. In vitro growth of human primordial follicles from frozen-banked ovarian tissue. Mol Cell Endocrinol, 2000;166:2735.Google Scholar
Xu, M, Barrett, SL, West-Farrell, E et al. In vitro grown human ovarian follicles from cancer patients support oocyte growth. Hum Reprod, 2009;24:25312540.Google Scholar
Amorim, CA, Shikanov, A. The artificial ovary: current status and future perspectives. Future Oncol, 2016;12:23232332.Google Scholar
Ouni, E, Vertommen, D, Chiti, MC, Dolmans, MM, Amorim, CA. A draft map of the human ovarian proteome for tissue engineering and clinical applications. Mol Cell Proteomics, 2019;18(1):S 159–S173 DOI:10.1074/mcp.RA117.000469.Google Scholar
Caliari, SR, Burdick, JA. A practical guide to hydrogels for cell culture. Nat Methods, 2016;13:405414.CrossRefGoogle ScholarPubMed
Sun, J, Tan, H. Alginate-based biomaterials for regenerative medicine applications. Materials, 2013;6:12851309.Google Scholar
Stein, H, Wilensky, M, Tsafrir, Y et al. Production of bioactive, post-translationally modified, heterotrimeric, human recombinant type-I collagen in transgenic tobacco. Biomacromolecules, 2009;10:26402645.Google Scholar
Peled, E, Boss, J, Bejar, J, Zinman, C, Seliktar, D. A novel poly(ethylene glycol)-fibrinogen hydrogel for tibial segmental defect repair in a rat model. J Biomed Mater Res A, 2007;80:874884.Google Scholar
Smitz, J, Dolmans, MM, Donnez, J et al. Current achievements and future research directions in ovarian tissue culture, in vitro follicle development and transplantation: implications for fertility preservation. Hum Reprod Update, 2010;16:395414.CrossRefGoogle ScholarPubMed
Gigli, I, Byrd, DD, Fortune, JE. Effects of oxygen tension and supplements to the culture medium on activation and development of bovine follicles in vitro. Theriogenology, 2006;66:344353.Google Scholar
Meyvantsson, I, Beebe, DJ. Cell culture in microfluidic systems. Annu Rev Anal Chem, 2008;1:423449.Google Scholar
Mehling, M, Tay, S. Microfluidic cell culture. Curr Opin Biotechnol, 2014;25:95102.Google Scholar
Nagashima, JB, El Assal, R, Songsasen, N, Demirci, U. Evaluation of an ovary-on-a-chip in large mammalian models: Species specificity and influence of follicle isolation status. J Tissue Eng Regen Med, 2018;12:e1926e1935.Google Scholar
Aziz, AUR, Fu, M, Deng, J et al. A microfluidic device for culturing an encapsulated ovarian follicle. Micromachines, 2017;8:335.Google Scholar
Navis, AR. Hanging drop tissue culture. Embryo Project Encyclopedia (2007–11-08). ISSN: 1940–5030 http://embryo.asu.edu/handle/10776/1720Google Scholar
Choi, JK, Agarwal, P, He, X. In vitro culture of early secondary preantral follicles in hanging drop of ovarian cell-conditioned medium to obtain MII oocytes from outbred deer mice. Tissue Eng Part A, 2013;19:26262637.Google Scholar
Millet, LJ, Gillette, MU. Over a century of neuron culture: from the hanging drop to microfluidic devices. Yale J Biol Med, 2012;85:501521.Google Scholar
Wycherley, G, Downey, D, Kane, MT, Hynes, AC. A novel follicle culture system markedly increases follicle volume, cell number and oestradiol secretion. Reproduction, 2004;127:669677.Google Scholar
He, X, Toth, TL. In vitro culture of ovarian follicles from Peromyscus. Semin Cell Dev Biol, 2017;61:140149.Google Scholar
Wang, W, Tang, Y, Ni, L et al. A modified protocol for in vitro maturation of mouse oocytes from secondary preantral follicles. Adv Biosci Biotechnol, 2012;3:17194.Google Scholar
Peroglio, M, Gaspar, D, Zeugolis, DI, Alini, M. Relevance of bioreactors and whole tissue cultures for the translation of new therapies to humans. J Orthop Res, 2018;36:1021.Google Scholar
Zanotelli, MR, Henningsen, JD, Hopkins, PM et al. An ovarian bioreactor for in vitro culture of the whole bovine ovary: a preliminary report. J Ovarian Res, 2016;9:47.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
×