Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-wq484 Total loading time: 0 Render date: 2024-04-25T11:04:37.562Z Has data issue: false hasContentIssue false

Chapter 16 - The Prospects of Infertility Treatment Using “Artificial” Eggs

Published online by Cambridge University Press:  02 December 2021

Gianpiero D. Palermo
Affiliation:
Cornell Institute of Reproductive Medicine, New York
Zsolt Peter Nagy
Affiliation:
Reproductive Biology Associates, Atlanta, GA
Get access

Summary

The female germline undergoes a unique sequence of differentiation processes that finally endows the eggs with totipotency. The reconstitution in vitro of oogenesis using pluripotent stem cells, which eventually produces “artificial eggs,” has long been sought in reproductive biology and medicine, as this would contribute not only to a better understanding of the basic mechanisms underlying totipotency, but also to an alternative source of gametes for reproduction. This chapter introduces a scientific background, the current status and the prospects of artificial eggs in mice and humans.

Type
Chapter
Information
Manual of Intracytoplasmic Sperm Injection in Human Assisted Reproduction
With Other Advanced Micromanipulation Techniques to Edit the Genetic and Cytoplasmic Content of the Oocyte
, pp. 154 - 160
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

Saitou, M., and Yamaji, M. (2010). Germ cell specification in mice: signaling, transcription regulation, and epigenetic consequences. Reproduction 139, 931942.CrossRefGoogle ScholarPubMed
Sasaki, H., and Matsui, Y. (2008). Epigenetic events in mammalian germ-cell development: reprogramming and beyond. Nat Rev Genet 9, 129140.CrossRefGoogle ScholarPubMed
Pedersen, T., and Peters, H. (1968). Proposal for a classification of oocytes and follicles in the mouse ovary. J Reprod Fertil 17, 555557.Google Scholar
Ohinata, Y., Ohta, H., Shigeta, M., Yamanaka, K., Wakayama, T., and Saitou, M. (2009). A signaling principle for the specification of the germ cell lineage in mice. Cell 137, 571584.Google Scholar
Hayashi, K., Ohta, H., Kurimoto, K., Aramaki, S., and Saitou, M. (2011). Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 146, 519532.CrossRefGoogle ScholarPubMed
Hayashi, K., Ogushi, S., Kurimoto, K., Shimamoto, S., Ohta, H., and Saitou, M. (2012). Offspring from oocytes derived from in vitro primordial germ cell-like cells in mice. Science 338, 971975.Google Scholar
Irie, N., Weinberger, L., Tang, W.W., Kobayashi, T., Viukov, S., Manor, Y.S., Dietmann, S., Hanna, J.H., and Surani, M.A. (2015). SOX17 is a critical specifier of human primordial germ cell fate. Cell 160, 253268.Google Scholar
Sasaki, K., Yokobayashi, S., Nakamura, T., Okamoto, I., Yabuta, Y., Kurimoto, K., Ohta, H., Moritoki, Y., Iwatani, C., Tsuchiya, H., et al. (2015). Robust In Vitro Induction of Human Germ Cell Fate from Pluripotent Stem Cells. Cell Stem Cell 17, 178194.CrossRefGoogle ScholarPubMed
Hikabe, O., Hamazaki, N., Nagamatsu, G., Obata, Y., Hirao, Y., Hamada, N., Shimamoto, S., Imamura, T., Nakashima, K., Saitou, M., et al. (2016). Reconstitution in vitro of the entire cycle of the mouse female germ line. Nature 539, 299303.CrossRefGoogle ScholarPubMed
Eppig, J.J., and O’Brien, M.J. (1996). Development in vitro of mouse oocytes from primordial follicles. Biol Reprod 54, 197207.Google Scholar
Morohaku, K., Tanimoto, R., Sasaki, K., Kawahara-Miki, R., Kono, T., Hayashi, K., Hirao, Y., and Obata, Y. (2016). Complete in vitro generation of fertile oocytes from mouse primordial germ cells. Proc Nat Acad Sci USA 113, 90219026.CrossRefGoogle ScholarPubMed
Hayashi, K., Hikabe, O., Obata, Y., and Hirao, Y. (2017). Reconstitution of mouse oogenesis in a dish from pluripotent stem cells. Nat Protoc 12, 17331744.Google Scholar
Hayashi, K., and Saitou, M. (2013a). Generation of eggs from mouse embryonic stem cells and induced pluripotent stem cells. Nat Protoc 8, 15131524.Google Scholar
Hayashi, K., and Saitou, M. (2013b). Stepwise differentiation from naive state pluripotent stem cells to functional primordial germ cells through an epiblast-like state. Methods Mol Biol 1074, 175183.CrossRefGoogle ScholarPubMed
Clark, A.T., Rodriguez, R.T., Bodnar, M.S., Abeyta, M.J., Cedars, M.I., Turek, P.J., Firpo, M.T., and Reijo Pera, R.A. (2004). Human STELLAR, NANOG, and GDF3 genes are expressed in pluripotent cells and map to chromosome 12p13, a hotspot for teratocarcinoma. Stem Cells 22, 169179.CrossRefGoogle ScholarPubMed
Kee, K., Gonsalves, J.M., Clark, A.T., and Pera, R.A. (2006). Bone morphogenetic proteins induce germ cell differentiation from human embryonic stem cells. Stem Cells Dev 15, 831837.Google Scholar
Sugawa, F., Arauzo-Bravo, M.J., Yoon, J., Kim, K.P., Aramaki, S., Wu, G., Stehling, M., Psathaki, O.E., Hubner, K., and Scholer, H.R. (2015). Human primordial germ cell commitment in vitro associates with a unique PRDM14 expression profile. EMBO J 34, 10091024.Google Scholar
McLaughlin, M., Albertini, D.F., Wallace, W.H.B., Anderson, R.A., and Telfer, E.E. (2018). Metaphase II oocytes from human unilaminar follicles grown in a multi-step culture system. Mol Hum Reprod 24, 135142.Google Scholar
Skory, R.M., Xu, Y., Shea, L.D., and Woodruff, T.K. (2015). Engineering the ovarian cycle using in vitro follicle culture. Hum Reprod 30, 13861395.Google Scholar
Telfer, E.E., and Zelinski, M.B. (2013). Ovarian follicle culture: advances and challenges for human and nonhuman primates. Fertil Steril 99, 15231533.Google Scholar
Yin, H., Kristensen, S.G., Jiang, H., Rasmussen, A., and Andersen, C.Y. (2016). Survival and growth of isolated pre-antral follicles from human ovarian medulla tissue during long-term 3D culture. Hum Reprod 31, 15311539.CrossRefGoogle ScholarPubMed
Yu, R.R., Cheng, A.T., Lagenaur, L.A., Huang, W., Weiss, D.E., Treece, J., Sanders-Beer, B.E., Hamer, D.H., Lee, P.P., Xu, Q., et al. (2009). A Chinese rhesus macaque (Macaca mulatta) model for vaginal Lactobacillus colonization and live microbicide development. J Med Primatol 38, 125136.Google Scholar
Xiao, S., Zhang, J., Romero, M.M., Smith, K.N., Shea, L.D., and Woodruff, T.K. (2015). In vitro follicle growth supports human oocyte meiotic maturation. Sci Rep 5, 17323.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
×