14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Fertility preservation in boys: recent developments and new insights

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          BACKGROUND

          Infertility is an important side effect of treatments used for cancer and other non-malignant conditions in males. This may be due to the loss of spermatogonial stem cells (SSCs) and/or altered functionality of testicular somatic cells (e.g. Sertoli cells, Leydig cells). Whereas sperm cryopreservation is the first-line procedure to preserve fertility in post-pubertal males, this option does not exist for prepubertal boys. For patients unable to produce sperm and at high risk of losing their fertility, testicular tissue freezing is now proposed as an alternative experimental option to safeguard their fertility.

          OBJECTIVE AND RATIONALE

          With this review, we aim to provide an update on clinical practices and experimental methods, as well as to describe patient management inclusion strategies used to preserve and restore the fertility of prepubertal boys at high risk of fertility loss.

          SEARCH METHODS

          Based on the expertise of the participating centres and a literature search of the progress in clinical practices, patient management strategies and experimental methods used to preserve and restore the fertility of prepubertal boys at high risk of fertility loss were identified. In addition, a survey was conducted amongst European and North American centres/networks that have published papers on their testicular tissue banking activity.

          OUTCOMES

          Since the first publication on murine SSC transplantation in 1994, remarkable progress has been made towards clinical application: cryopreservation protocols for testicular tissue have been developed in animal models and are now offered to patients in clinics as a still experimental procedure. Transplantation methods have been adapted for human testis, and the efficiency and safety of the technique are being evaluated in mouse and primate models. However, important practical, medical and ethical issues must be resolved before fertility restoration can be applied in the clinic.Since the previous survey conducted in 2012, the implementation of testicular tissue cryopreservation as a means to preserve the fertility of prepubertal boys has increased. Data have been collected from 24 co-ordinating centres worldwide, which are actively offering testis tissue cryobanking to safeguard the future fertility of boys. More than 1033 young patients (age range 3 months to 18 years) have already undergone testicular tissue retrieval and storage for fertility preservation.

          LIMITATIONS, REASONS FOR CAUTION

          The review does not include the data of all reproductive centres worldwide. Other centres might be offering testicular tissue cryopreservation. Therefore, the numbers might be not representative for the entire field in reproductive medicine and biology worldwide. The key ethical issue regarding fertility preservation in prepubertal boys remains the experimental nature of the intervention.

          WIDER IMPLICATIONS

          The revised procedures can be implemented by the multi-disciplinary teams offering and/or developing treatment strategies to preserve the fertility of prepubertal boys who have a high risk of fertility loss.

          STUDY FUNDING/COMPETING INTEREST(S)

          The work was funded by ESHRE. None of the authors has a conflict of interest.

          Related collections

          Most cited references149

          • Record: found
          • Abstract: found
          • Article: not found

          Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells.

          The generation of properly functioning gametes in vitro requires reconstitution of the multistepped pathway of germ cell development. We demonstrate here the generation of primordial germ cell-like cells (PGCLCs) in mice with robust capacity for spermatogenesis. PGCLCs were generated from embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) through epiblast-like cells (EpiLCs), a cellular state highly similar to pregastrulating epiblasts but distinct from epiblast stem cells (EpiSCs). Reflecting epiblast development, EpiLC induction from ESCs/iPSCs is a progressive process, and EpiLCs highly competent for the PGC fate are a transient entity. The global transcription profiles, epigenetic reprogramming, and cellular dynamics during PGCLC induction from EpiLCs meticulously capture those associated with PGC specification from the epiblasts. Furthermore, we identify Integrin-β3 and SSEA1 as markers that allow the isolation of PGCLCs with spermatogenic capacity from tumorigenic undifferentiated cells. Our findings provide a paradigm for the first step of in vitro gametogenesis. Copyright © 2011 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Long-term proliferation in culture and germline transmission of mouse male germline stem cells.

            Spermatogenesis is a complex process that originates in a small population of spermatogonial stem cells. Here we report the in vitro culture of spermatogonial stem cells that proliferate for long periods of time. In the presence of glial cell line-derived neurotrophic factor, epidermal growth factor, basic fibroblast growth factor, and leukemia inhibitory factor, gonocytes isolated from neonatal mouse testis proliferated over a 5-month period (>10(14)-fold) and restored fertility to congenitally infertile recipient mice following transplantation into seminiferous tubules. Long-term spermatogonial stem cell culture will be useful for studying spermatogenesis mechanism and has important implications for developing new technology in transgenesis or medicine.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              The adult human testis transcriptional cell atlas

              Human adult spermatogenesis balances spermatogonial stem cell (SSC) self-renewal and differentiation, alongside complex germ cell-niche interactions, to ensure long-term fertility and faithful genome propagation. Here, we performed single-cell RNA sequencing of ~6500 testicular cells from young adults. We found five niche/somatic cell types (Leydig, myoid, Sertoli, endothelial, macrophage), and observed germline-niche interactions and key human-mouse differences. Spermatogenesis, including meiosis, was reconstructed computationally, revealing sequential coding, non-coding, and repeat-element transcriptional signatures. Interestingly, we identified five discrete transcriptional/developmental spermatogonial states, including a novel early SSC state, termed State 0. Epigenetic features and nascent transcription analyses suggested developmental plasticity within spermatogonial States. To understand the origin of State 0, we profiled testicular cells from infants, and identified distinct similarities between adult State 0 and infant SSCs. Overall, our datasets describe key transcriptional and epigenetic signatures of the normal adult human testis, and provide new insights into germ cell developmental transitions and plasticity.
                Bookmark

                Author and article information

                Journal
                Hum Reprod Open
                Hum Reprod Open
                hropen
                Human Reproduction Open
                Oxford University Press
                2399-3529
                2020
                06 June 2020
                06 June 2020
                : 2020
                : 3
                : hoaa016
                Affiliations
                [1 ] Biology of the Testis , Research Laboratory for Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium
                [2 ] NORDFERTIL Research Lab Stockholm , Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institutet and Karolinska University Hospital, Solna, Sweden
                [3 ] Division of Haematology-Oncology and Stem Cell Transplantation , New Children’s Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
                [4 ] MRC Centre for Reproductive Health , The Queen’s Medical Research Institute, The University of Edinburgh; and the Edinburgh Royal Hospital for Sick Children, Edinburgh, UK
                [5 ] Center for Reproductive Medicine , Amsterdam UMC, Amsterdam Reproduction and Development Research Institute, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
                [6 ] Bioethics Institute Ghent , Ghent University, 9000 Ghent, Belgium
                [7 ] Normandie Univ , UNIROUEN, EA 4308 “Gametogenesis and Gamete Quality”, Rouen University Hospital, Biology of Reproduction-CECOS Laboratory, F 76000, Rouen, France
                [8 ] Department of Gynecology and Andrology , Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
                [9 ] Department of Paediatric Oncology and Haematology , Children’s Hospital Oxford, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
                [10 ] Department of Oncology Pathology , Karolinska Institutet, Solna, Sweden
                [11 ] Section of Reproductive Medicine , Division of Gynecology and Reproduction, Karolinska University Hospital, Stockholm, Sweden
                [12 ] Magee-Womens Research Institute , University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
                [13 ] Centre of Reproductive Medicine and Andrology , Institute of Reproductive and Regenerative Biology, University of Münster, Münster, Germany
                [14 ] Reproductive Medicine Unit , Hospital Universitario y Politécnico La Fe, Valencia, Spain
                [15 ] Departement of Obstetrics and Gynacology , Division Reproductive Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
                [16 ] Laboratory of Reproductive Biology , The Juliane Marie Centre for Women, Children and Reproduction, University Hospital of Copenhagen, Denmark
                [17 ] Children’s Medical Center , Landspítali University Hospital, Reykjavik, Iceland and Faculty of Medicine, University of Iceland, Reykjavik, Iceland
                Author notes
                Correspondence address. Biology of the Testis, Research Laboratory for Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Laarbeeklaan 103, 1090 Brussels, Belgium. Tel: +32 2 474 9213; E-mail: Ellen.Goossens@ 123456vub.be

                Co-senior authors

                Representative of the ESHRE special interest group (SIG) Andrology

                Representative of the ESHRE SIG Fertility Preservation ESHRE Pages content is not externally peer reviewed. The manuscript has been approved by the Executive Committee of ESHRE.

                Representative of the ESHRE SIG Fertility Preservation

                Author information
                http://orcid.org/0000-0001-7601-9689
                Article
                hoaa016
                10.1093/hropen/hoaa016
                7275639
                32529047
                22866355-85e1-4eba-b5d8-c1705a55a9b3
                © The Author(s) 2020. Published by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology.

                This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License ( http://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact journals.permissions@oup.com

                History
                : 22 January 2020
                : 22 January 2020
                : 14 February 2020
                Page count
                Pages: 18
                Funding
                Funded by: FNRS-Televie;
                Award ID: 7.4596.13, 7.4554.14F and 7.6511.16
                Funded by: Netherlands Organisation for Health Research and Development, DOI 10.13039/501100001826;
                Award ID: 116003002
                Funded by: Swedish Childhood Cancer Foundation, DOI 10.13039/501100006313;
                Award ID: TJ2016-0093
                Funded by: European Society of Human Reproduction and Embryology; MRC Centre for Reproductive Health funded by MRC Centre Grant;
                Categories
                ESHRE Pages

                cryopreservation,in vitro spermatogenesis,fertility preservation,fertility restoration,prepubertal boys,spermatogonial stem cell,testicular tissue freezing,testis,transplantation

                Comments

                Comment on this article