1
|
Krishnan S, Daly MP, Kipping R, Harrison C. A systematic review of interventions to improve male knowledge of fertility and fertility-related risk factors. HUM FERTIL 2024; 27:2328066. [PMID: 38497245 DOI: 10.1080/14647273.2024.2328066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/02/2024] [Indexed: 03/19/2024]
Abstract
Male infertility is a global health concern. The effectiveness of interventions developed to improve males' knowledge of fertility regulation and fertility-related risk factors remains unclear. This systematic review aimed to synthesize and evaluate the evidence for these interventions. Four databases were searched from inception to June 2023. Eligible studies examined interventions to increase fertility knowledge among presumed fertile males aged ≥16 years of age. Conference abstracts, protocols and studies without sex-disaggregated results for males were excluded. A narrative synthesis without meta-analysis was performed. A total of 4884 records were identified. Five studies (reported in six publications), all conducted in high-income countries, were included. Two were randomized control trials, and three were experimental studies. Interventions were delivered in person by a health professional (n = 3), online and via a mobile app. All studies showed a significant improvement in knowledge of fertility or fertility-related risk factors from baseline to follow-up. The largest improvement was observed for secondary and vocational students. A moderate, long-term retainment of knowledge was observed at two-year follow-up in one study. Available evidence suggests interventions to improve males' fertility knowledge are effective, particularly for younger, less educated males.
Collapse
Affiliation(s)
- Srinithy Krishnan
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Michael P Daly
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ruth Kipping
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - China Harrison
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- The National Institute for Health and Care Research Applied Research Collaboration West (NIHR ARC West) at University Hospitals Bristol and Weston NHS Foundation Trust, UK
- National Institute for Health and Care Research, Health Protection Research Unit (NIHR HPRU) in Behavioural Science and Evaluation, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| |
Collapse
|
2
|
Jayasinghe YL, Ginsburg E. Oncofertility in Children and Adolescents. Obstet Gynecol Clin North Am 2024; 51:711-730. [PMID: 39510740 DOI: 10.1016/j.ogc.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Major improvements have been seen in oncofertility care over the last decade. Clinical ethics frameworks support new populations, including children, having access to novel fertility preservation techniques. Oncofertility consultation requires a multidisciplinary approach. Clinicians need to develop competencies in infertility risk-assessment, counseling regarding fertility preservation procedures and alternate family planning options, and managing individualised supportive care needs to minimize medical and psychological morbidity.
Collapse
Affiliation(s)
- Yasmin L Jayasinghe
- Department of Obstetrics Gynecology and Newborn Health, Royal Womens Hospital, University of Melbourne, Australia; Department of Gynecology, Royal Children's Hospital, Melbourne, Australia; Murdoch Children's Research Institute, Melbourne, Australia.
| | - Elizabeth Ginsburg
- Department of Reproductive Endocrinology and Infertility, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Department of Obstetrics Gynecology & Reproductive Biology, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| |
Collapse
|
3
|
Bier S, Kliesch S. [Cryopreservation of human spermatozoa or testicular tissue for fertility preservation]. UROLOGIE (HEIDELBERG, GERMANY) 2024; 63:1089-1096. [PMID: 39441429 DOI: 10.1007/s00120-024-02456-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Loss of reproductive capacity due to treatments for malignant or non-malignant diseases or even as a result of diseases themselves significantly impacts patients' quality of life. Cryopreservation of sperm from ejaculate is a well-established procedure for preserving the fertility of these patients and thus improving their quality of life in the long term. If cryopreservation of sperm from ejaculate is not possible, either because ejaculation cannot occur or no sperm can be found in the ejaculate, the preferred treatment option is (microsurgical) testicular sperm extraction (mTESE). Testicular sperm and ejaculated spermatozoa can be cryopreserved and later used for intracytoplasmic sperm injection (ICSI) treatment. The use of cryopreserved sperm for fertility treatment does not carry an increased risk of malformations in the offspring. If gonadotoxic therapy is necessary in pre- or early pubertal boys, the only option to preserve fertility in the long term is to cryopreserve spermatogonial stem cells from testicular tissue as part of the Androprotect© network. This is an experimental approach which has been available since 2012 across Germany and which is accompanied by intensive scientific work ( www.androprotect.de ).
Collapse
Affiliation(s)
- Simone Bier
- Abteilung für Klinische und Operative Andrologie, Centrum für Reproduktionsmedizin und Andrologie, WHO-Kooperationszentrum, EAA-Ausbildungszentrum, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude D11 (Domagkstraße 11), 48149, Münster, Deutschland
| | - Sabine Kliesch
- Abteilung für Klinische und Operative Andrologie, Centrum für Reproduktionsmedizin und Andrologie, WHO-Kooperationszentrum, EAA-Ausbildungszentrum, Universitätsklinikum Münster, Albert-Schweitzer-Campus 1, Gebäude D11 (Domagkstraße 11), 48149, Münster, Deutschland.
| |
Collapse
|
4
|
Delgouffe E, Paturlanne JM, Kovacevic A, Keselj I, Ammar O, Gaikwad AS, Saritas G, Egeberg D, Marcu D, de la Iglesia A. From spermatogonia to spermatozoa: Filling gaps in andrology at the 16th Network of Young Researchers in Andrology meeting. Andrology 2024. [PMID: 39301841 DOI: 10.1111/andr.13763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
The 16th Network of Young Researchers in Andrology meeting, hosted at the Sleepwell Hostel in Brussels, Belgium, was the first Network of Young Researchers in Andrology meeting as the young arm of the European Academy of Andrology. Over three days, this vibrant event provided a valuable platform for early-career researchers in andrology to present and discuss their research. With 41 attendees from 12 different countries, the meeting featured a diverse scientific program including keynote lectures from six world-leading experts, covering a broad range of topics in andrology. The 16th Network of Young Researchers in Andrology meeting showcased advancements in fertility preservation, single-cell applications, in vitro testis modeling, and epigenetics. Networking opportunities were a key highlight, featuring a scientific speed-dating session and a networking dinner designed to foster meaningful connections and collaborations among participants. The meeting concluded with a workshop on the science of sleep, offering attendees practical strategies to enhance their rest and well-being. Overall, the 16th Network of Young Researchers in Andrology meeting significantly advanced the audience's knowledge, strengthened the network of young researchers, and underlined Network of Young Researchers in Andrology's commitment to supporting and collaborating with emerging scientists in the andrology community.
Collapse
Affiliation(s)
- Emily Delgouffe
- Genetics, reproduction, and development (GRAD) Research Group, Biology of the Testis (BITE) Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Juan Manuel Paturlanne
- Centre of Reproductive Medicine and Andrology, Institute of Reproductive and Regenerative Biology, University of Münster, Munster, Germany
| | - Andjela Kovacevic
- Department of Developmental Pathology, Medical Faculty, Institute of Pathology, University of Bonn, Bonn, Germany
| | - Isidora Keselj
- Laboratory of Reproductive Endocrinology and Signaling, Laboratory for Chronobiology and Aging, CeRES, Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - Omar Ammar
- Ar-Razzi Private Hospital, IVF Centre, Ramadi, Iraq
- Department of Obstetrics and Gynaecology, College of Medicine, University of Anbar, Ramadi, Iraq
| | - Avinash S Gaikwad
- Centre of Medical Genetics, Institute of Reproductive Genetics, University of Münster, Munster, Germany
| | - Gülizar Saritas
- Department of Growth and Reproduction, Copenhagen University Hospital, Copenhagen, Denmark
| | | | - Daniel Marcu
- School of Biological Science, University of East Anglia, Norwich, UK
| | - Alberto de la Iglesia
- Department of Cellular Plasticity and Reproduction, Université Paris Cité, INSERM, CNRS, Institut Cochin, Paris, France
| |
Collapse
|
5
|
Bashiri Z, Hosseini SJ, Salem M, Koruji M. In vivo and in vitro sperm production: an overview of the challenges and advances in male fertility restoration. Clin Exp Reprod Med 2024; 51:171-180. [PMID: 38525520 PMCID: PMC11372308 DOI: 10.5653/cerm.2023.06569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/14/2023] [Indexed: 03/26/2024] Open
Abstract
Male infertility can be caused by genetic anomalies, endocrine disorders, inflammation, and exposure to toxic chemicals or gonadotoxic treatments. Therefore, several recent studies have concentrated on the preservation and restoration of fertility to enhance the quality of life for affected individuals. It is currently recommended to biobank the tissue extracted from testicular biopsies to provide a later source of spermatogonial stem cells (SSCs). Another successful approach has been the in vitro production of haploid male germ cells. The capacity of SSCs to transform into sperm, as in testicular tissue transplantation, SSC therapy, and in vitro or ex vivo spermatogenesis, makes them ideal candidates for in vivo fertility restoration. The transplantation of SSCs or testicular tissue to regenerate spermatogenesis and create embryos has been achieved in nonhuman mammal species. Although the outcomes of human trials have yet to be released, this method may soon be approved for clinical use in humans. Furthermore, regenerative medicine techniques that develop tissue or cells on organic or synthetic scaffolds enriched with bioactive molecules have also gained traction. All of these methods are now in different stages of experimentation and clinical trials. However, thanks to rigorous studies on the safety and effectiveness of SSC-based reproductive treatments, some of these techniques may be clinically available in upcoming decades.
Collapse
Affiliation(s)
- Zahra Bashiri
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Omid Fertility and Infertility Clinic, Hamedan, Iran
| | - Seyed Jamal Hosseini
- Biomedical Engineering Department, Amirkabir University of Technology, Tehran, Iran
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Salem
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Gizer M, Önen S, Korkusuz P. The Evolutionary Route of in vitro Human Spermatogenesis: What is the Next Destination? Stem Cell Rev Rep 2024; 20:1406-1419. [PMID: 38684571 PMCID: PMC11319530 DOI: 10.1007/s12015-024-10726-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Malfunction in spermatogenesis due to genetic diseases, trauma, congenital disorders or gonadotoxic treatments results in infertility in approximately 7% of males. The behavior of spermatogonial stem cells (SSCs) within three-dimensional, multifactorial, and dynamic microenvironment implicates a niche that serves as a repository for fertility, since can serve as a source of mature and functional male germ cells. Current protocols enable reprogramming of mature somatic cells into induced pluripotent stem cells (iPSCs) and their limited differentiation to SSCs within the range of 0-5%. However, the resulting human iPSC-derived haploid spermatogenic germ cell yield in terms of number and functionality is currently insufficient for transfer to infertility clinic as a therapeutic tool. In this article, we reviewed the evolution of experimental culture platforms and introduced a novel iPSCs-based approach for in vitro spermatogenesis based on a niche perspective bearing cellular, chemical, and physical factors that provide the complex arrangement of testicular seminiferous tubules embedded within a vascularized stroma. We believe that bioengineered organoids supported by smart bio-printed tubules and microfluidic organ-on-a-chip systems offer efficient, precise, personalized platforms for autologous pluripotent stem cell sources to undergo the spermatogenetic cycle, presenting a promising tool for infertile male patients with complete testicular aplasia.
Collapse
Affiliation(s)
- Merve Gizer
- Department of Stem Cell Sciences, Graduate School of Health Sciences, Hacettepe University, 06100, Ankara, Turkey
- METU MEMS Center, 06530, Ankara, Turkey
| | | | - Petek Korkusuz
- METU MEMS Center, 06530, Ankara, Turkey.
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100, Ankara, Turkey.
| |
Collapse
|
7
|
Huang C, Ji XR, Huang ZH, Wang RJ, Fan LQ, Zhu WB, Luo Q, Qing-Li. Global status of research on fertility preservation in male patients with cancer: A bibliometric and visual analysis. Heliyon 2024; 10:e33621. [PMID: 39040288 PMCID: PMC11260990 DOI: 10.1016/j.heliyon.2024.e33621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Background Recently, male fertility preservation before cancer treatment has become more prevalent. The research in this field has progressed over time, with some studies having a major impact and providing guidance for further research. However, the trends and hotspots of research on fertility preservation in male cancer patients may have changed; exploring them is essential for relevant research progress. Design We extracted relevant studies from the Web of Science Core Collection database, capturing information on the countries of study, affiliations, authors, keywords, as well as co-citations of references and journals. To identify publication trends, research strengths, key subjects, prominent topics, and emerging areas, we conducted a bibliometric analysis using CiteSpace. Results We included 3201 articles on fertility preservation in male cancer patients published over January 1999 to December 2023 were included. Although the relevant research growth rate was slow initially, the number of publications increased annually. Of all study countries, the United States, Germany, and Japan reported the earliest studies; the United States published the highest number of relevant studies. The US institutions remained at the forefront for all 25 years, and the US researcher Ashok Agarwal published the most articles. Literature co-citation analyses indicated a transformation in the study participants; they comprised a younger demographic (i.e., a large number of adolescent male patients underwent fertility preservation); moreover, fertility preservation techniques evolved from sperm cryopreservation to testicular tissue cryopreservation. Research on reproductive outcomes of sperm cryopreservation was the recent hotspot in male fertility preservation research, and the impact of immunotherapy and checkpoint inhibitors on male fertility requires further research. Conclusions Male fertility preservation will be a major future research focus, with closer connections and collaborations between countries and organizations. Our results present the historical data on the development of research on male fertility preservation in cancer patients, providing relevant insights for future research and development in this study area.
Collapse
Affiliation(s)
- Chuan Huang
- The Institute of Reproductive and Stem Cell Engineering, Basic Medicine College, Central South University, Changsha, Hunan, China
- Human Sperm Bank, The Reproductive & Genetic Hospital of CITIC-XIANGYA, Changsha, China
| | - Xi-Ren Ji
- Human Sperm Bank, The Reproductive & Genetic Hospital of CITIC-XIANGYA, Changsha, China
| | - Zeng-Hui Huang
- The Institute of Reproductive and Stem Cell Engineering, Basic Medicine College, Central South University, Changsha, Hunan, China
- Human Sperm Bank, The Reproductive & Genetic Hospital of CITIC-XIANGYA, Changsha, China
| | - Rui-Jun Wang
- The Institute of Reproductive and Stem Cell Engineering, Basic Medicine College, Central South University, Changsha, Hunan, China
| | - Li-Qing Fan
- The Institute of Reproductive and Stem Cell Engineering, Basic Medicine College, Central South University, Changsha, Hunan, China
- Human Sperm Bank, The Reproductive & Genetic Hospital of CITIC-XIANGYA, Changsha, China
| | - Wen-Bing Zhu
- The Institute of Reproductive and Stem Cell Engineering, Basic Medicine College, Central South University, Changsha, Hunan, China
- Human Sperm Bank, The Reproductive & Genetic Hospital of CITIC-XIANGYA, Changsha, China
| | - Qiang Luo
- Human Sperm Bank, The Reproductive & Genetic Hospital of CITIC-XIANGYA, Changsha, China
| | - Qing-Li
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Asadi-Azarbaijani B, Oskam IC, Jahnukainen K. A 12-year overview of fertility preservation practice in Nordic pediatric oncology centers. J Cancer Surviv 2024:10.1007/s11764-024-01627-x. [PMID: 38871993 DOI: 10.1007/s11764-024-01627-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE Fertility preservation is the only option to safeguard fertility following gonadotoxic treatments. This study aimed to provide an updated status on fertility preservation for pediatric cancer patients in the Nordic countries. METHODS A questionnaire consisting of 14 questions was sent to directors of 18 main pediatric oncology centers in the Nordic countries in 2010 and 2022. We received information regarding indications, guidelines, counseling, and available fertility preservation options. RESULTS The response rates were 89% in 2010 and 72% in 2022. The results reveal an increase in clinical practice guidelines on fertility preservation for cancer patients, from 25% in 2010 to 70% in 2022. Counseling on fertility preservation options in 2022 was more specific and offered to most patients who fulfilled indications for fertility preservation (from 19 to 77%). Sperm cryopreservation continues to be the predominant fertility preservation method for pubertal boys in the Nordic countries. However, there has been a notable increase in the availability of testicular tissue preservation for prepubertal boys (0 to 62%). A similar increase in the offer of ovarian tissue preservation for prepubertal girls (0 to 92%) was observed among pediatric cancer patients. CONCLUSIONS The past decade has shown commendable advancements in fertility preservation for pediatric cancer patients in the Nordic countries. IMPLICATIONS FOR CANCER SURVIVORS As fertility care evolves globally, continuous assessment of regional practices and challenges is imperative to enhance the quality of care and life for pediatric cancer survivors in the Nordic regions.
Collapse
Affiliation(s)
| | - Irma C Oskam
- The Livestock Production Research Centre, Norwegian University of Life Sciences, Aas, Norway
| | - Kirsi Jahnukainen
- Department of Pediatrics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
9
|
Mohammadi A, Shabani R, Bashiri Z, Rafiei S, Asgari H, Koruji M. Therapeutic potential of exosomes in spermatogenesis regulation and male infertility. Biol Cell 2024; 116:e2300127. [PMID: 38593304 DOI: 10.1111/boc.202300127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Spermatogenesis is a fundamental process crucial for male reproductive health and fertility. Exosomes, small membranous vesicles released by various cell types, have recently garnered attention for their role in intercellular communication. OBJECTIVE This review aims to comprehensively explore the role of exosomes in regulating spermatogenesis, focusing on their involvement in testicular development and cell-to-cell communication. METHODS A systematic examination of literature was conducted to gather relevant studies elucidating the biogenesis, composition, and functions of exosomes in the context of spermatogenesis. RESULTS Exosomes play a pivotal role in orchestrating the complex signaling networks required for proper spermatogenesis. They facilitate the transfer of key regulatory molecules between different cell populations within the testes, including Sertoli cells, Leydig cells, and germ cells. CONCLUSION The emerging understanding of exosome-mediated communication sheds light on novel mechanisms underlying spermatogenesis regulation. Further research in this area holds promise for insights into male reproductive health and potential therapeutic interventions.
Collapse
Affiliation(s)
- Amirhossein Mohammadi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ronak Shabani
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Reproductive Sciences and Technology Research Center, Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Omid Fertility & Infertility Clinic, Hamedan, Iran
| | - Sara Rafiei
- Department of Botany and Plant Sciences, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Hamidreza Asgari
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
朱 慧, 黄 薇. [Reproductive Strategies for Declining Fertility: Fertility Preservation]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:507-512. [PMID: 38948295 PMCID: PMC11211799 DOI: 10.12182/20240560204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Indexed: 07/02/2024]
Abstract
There is a global trend of declining fertility among people of childbearing age and mankind is confronted with great challenges of fertility problems. As a result, fertility preservation technology has emerged. Fertility preservation involves interventions and procedures aimed at preserving the patients' chances of having children when their fertility may have been impaired by their medical conditions or the treatments thereof, for example, chemotherapy and/or radiotherapy for cancer. The changes in patients' fertility can be temporary or permanent damage. Fertility preservation can help people diagnosed with cancer or other non-malignant diseases. More and more fertility preservation methods are being used to preserve the fertility of cancer patients and protect their reproductive organs from gonadotoxicity. Fertility preservation may be appropriate for young patients with early-stage cancers and good prognosis before they undergo treatments (chemotherapy and/or radiotherapy) that can negatively affect their fertility. It is also appropriate for patients with chronic conditions or those who have encountered environmental exposures that affect their gonadal function. Fertility preservation methods include oocyte cryopreservation, embryo cryopreservation, and ovarian tissue cryopreservation (OTC) for women and sperm freezing and testicular tissue freezing for men. The survival rates of children and adolescents diagnosed with malignant tumors have been steadily increasing as a result of advances in cancer treatments. Cryopreservation of oocytes and sperm is recognized as a well-established and successful strategy for fertility preservation in pubertal patients. OTC is the sole option for prepubertal girls. On the other hand, cryopreservation of immature testicular tissue remains the only alternative for prepubertal boys, but the technology is still in the experimental stage. A review showed that the utilization rate of cryopreserved semen ranged from 2.6% to 21.5%. In the case of cryopreserved female reproductive materials, the utilization rate ranged from 3.1% to 8.7% for oocytes, approximately from 9% to 22.4% for embryos, and from 6.9% to 30.3% for ovarian tissue. When patients have needs for fertility treatment, cryopreserved vitrified oocytes are resuscitated and in vitro fertilization-embryo transfer (IVF-ET) was performed to help patients accomplish their reproductive objectives, with the live birth rate (LBR) being 32%. On the other hand, when cryopreserved embryos are resuscitated and transferred, the LBR was 41%. OTC has the advantage of restoring natural fertility and presents a LBR of 33%, compared with the LBR of 19% among 266 IVF patients. In addition, OTC has the benefit of restoring the endocrine function. It has been observed that the shortest recovery time of the first menstruation after transplantation was 3.9 months, and the recovery rate of ovarian function reached 100%. To date, a growing number of cancer survivors and patients with other diseases are benefiting from fertility preservation measures. In the face of declining human fertility, fertility preservation provides a new approach to human reproduction. Fertility preservation should be applied in line with the ethical principles so as to fully protect the rights and interests of patients and their offsprings.
Collapse
Affiliation(s)
- 慧莉 朱
- 四川大学华西第二医院 生殖医学科 (成都 610041)Department of Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 出生缺陷与相关妇儿疾病教育部重点实验室(四川大学) (成都 610041)Key Laboratory of Birth Defects and Related Maternal and Child Diseases of the Ministry of Education, Sichuan University, Chengdu 610041, China
| | - 薇 黄
- 四川大学华西第二医院 生殖医学科 (成都 610041)Department of Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- 出生缺陷与相关妇儿疾病教育部重点实验室(四川大学) (成都 610041)Key Laboratory of Birth Defects and Related Maternal and Child Diseases of the Ministry of Education, Sichuan University, Chengdu 610041, China
- 国家卫生健康委时间生物学重点实验室(四川大学) (成都 610041)National Health Commission Key Laboratory of Chronobiology, Sichuan University , Chengdu 610041, China
| |
Collapse
|
11
|
Volkova N, Yukhta M, Goltsev A. DNA fragmentation, antioxidant activity and histological structure of cryopreserved testicular tissue depending on sexual maturity and immunological status. Cryobiology 2024; 114:104862. [PMID: 38360086 DOI: 10.1016/j.cryobiol.2024.104862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/06/2024] [Accepted: 02/12/2024] [Indexed: 02/17/2024]
Abstract
The objective of this work was to determine a relationship between a frequency of DNA fragmentation, a level of antioxidant activity and a preservation of histological structure depending on initial status of fragments of seminiferous tubules of testes (FSTT) of rats at the stages of cryopreservation. FSTT of animals of different ages (immature, mature), as well as animals with changed immunological status (adjuvant arthritis) were cryopreserved. Slow uncontrolled freezing was used in a cryomedium of fibrin gel with 0.7 M glycerol. The results showed that viability, TAS, γGGT and G6PD activities had the highest values in the group of intact immature animals both in fresh FSTT and after exposure to cryomedium or cryopreservation, while the indexes of DNA fragmentation and ROS content had the lowest values. It was found that an increase in the DNA fragmentation rate occurred in parallel with a decrease in the values of antioxidant activity and membrane integrity. The spermatogenenic epithelium after cryopreservation differed between the groups in a relative number of cells with pathologically changed nuclei and the frequency of exfoliation of epithelial cells into the tubule cavity namely, there was a tendency to an increase in the damaging effects in the series, "Immature → Sexually mature → Autoimmune arthritis". The obtained data can be taken into account in the development of low-temperature preservation protocols using cryotechnologies, which will ensure the maintenance of the morphological and functional characteristics of FSTT depending on sexual maturity and immunological status.
Collapse
Affiliation(s)
- Nataliia Volkova
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, str. Pereyaslavska, 23, Kharkiv, 61016, Ukraine.
| | - Mariia Yukhta
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, str. Pereyaslavska, 23, Kharkiv, 61016, Ukraine
| | - Anatoliy Goltsev
- Institute for Problems of Cryobiology and Cryomedicine, National Academy of Sciences of Ukraine, str. Pereyaslavska, 23, Kharkiv, 61016, Ukraine
| |
Collapse
|
12
|
Duffin K, Neuhaus N, Andersen CY, Barraud-Lange V, Braye A, Eguizabal C, Feraille A, Ginsberg JP, Gook D, Goossens E, Jahnukainen K, Jayasinghe Y, Keros V, Kliesch S, Lane S, Mulder CL, Orwig KE, van Pelt AMM, Poirot C, Rimmer MP, Rives N, Sadri-Ardekani H, Safrai M, Schlatt S, Stukenborg JB, van de Wetering MD, Wyns C, Mitchell RT. A 20-year overview of fertility preservation in boys: new insights gained through a comprehensive international survey. Hum Reprod Open 2024; 2024:hoae010. [PMID: 38449521 PMCID: PMC10914450 DOI: 10.1093/hropen/hoae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/02/2024] [Indexed: 03/08/2024] Open
Abstract
STUDY QUESTION Twenty years after the inception of the first fertility preservation programme for pre-pubertal boys, what are the current international practices with regard to cryopreservation of immature testicular tissue? SUMMARY ANSWER Worldwide, testicular tissue has been cryopreserved from over 3000 boys under the age of 18 years for a variety of malignant and non-malignant indications; there is variability in practices related to eligibility, clinical assessment, storage, and funding. WHAT IS KNOWN ALREADY For male patients receiving gonadotoxic treatment prior to puberty, testicular tissue cryopreservation may provide a method of fertility preservation. While this technique remains experimental, an increasing number of centres worldwide are cryopreserving immature testicular tissue and are approaching clinical application of methods to use this stored tissue to restore fertility. As such, standards for quality assurance and clinical care in preserving immature testicular tissue should be established. STUDY DESIGN SIZE DURATION A detailed survey was sent to 17 centres within the recently established ORCHID-NET consortium, which offer testicular tissue cryopreservation to patients under the age of 18 years. The study encompassed 60 questions and remained open from 1 July to 1 November 2022. PARTICIPANTS/MATERIALS SETTING METHODS Of the 17 invited centres, 16 completed the survey, with representation from Europe, Australia, and the USA. Collectively, these centres have cryopreserved testicular tissue from patients under the age of 18 years. Data are presented using descriptive analysis. MAIN RESULTS AND THE ROLE OF CHANCE Since the establishment of the first formal fertility preservation programme for pre-pubertal males in 2002, these 16 centres have cryopreserved tissue from 3118 patients under the age of 18 years, with both malignant (60.4%) and non-malignant (39.6%) diagnoses. All centres perform unilateral biopsies, while 6/16 sometimes perform bilateral biopsies. When cryopreserving tissue, 9/16 centres preserve fragments sized ≤5 mm3 with the remainder preserving fragments sized 6-20 mm3. Dimethylsulphoxide is commonly used as a cryoprotectant, with medium supplements varying across centres. There are variations in funding source, storage duration, and follow-up practice. Research, with consent, is conducted on stored tissue in 13/16 centres. LIMITATIONS REASONS FOR CAUTION While this is a multi-national study, it will not encompass every centre worldwide that is cryopreserving testicular tissue from males under 18 years of age. As such, it is likely that the actual number of patients is even higher than we report. Whilst the study is likely to reflect global practice overall, it will not provide a complete picture of practices in every centre. WIDER IMPLICATIONS OF THE FINDINGS Given the research advances, it is reasonable to suggest that cryopreserved immature testicular tissue will in the future be used clinically to restore fertility. The growing number of patients undergoing this procedure necessitates collaboration between centres to better harmonize clinical and research protocols evaluating tissue function and clinical outcomes in these patients. STUDY FUNDING/COMPETING INTERESTS K.D. is supported by a CRUK grant (C157/A25193). R.T.M. is supported by an UK Research and Innovation (UKRI) Future Leaders Fellowship (MR/S017151/1). The MRC Centre for Reproductive Health at the University of Edinburgh is supported by MRC (MR/N022556/1). C.L.M. is funded by Kika86 and ZonMW TAS 116003002. A.M.M.v.P. is supported by ZonMW TAS 116003002. E.G. was supported by the Research Program of the Research Foundation-Flanders (G.0109.18N), Kom op tegen Kanker, the Strategic Research Program (VUB_SRP89), and the Scientific Fund Willy Gepts. J.-B.S. is supported by the Swedish Childhood Cancer Foundation (TJ2020-0026). The work of NORDFERTIL is supported by the Swedish Childhood Cancer Foundation (PR2019-0123; PR2022-0115), the Swedish Research Council (2018-03094; 2021-02107), and the Birgitta and Carl-Axel Rydbeck's Research Grant for Paediatric Research (2020-00348; 2021-00073; 2022-00317; 2023-00353). C.E is supported by the Health Department of the Basque Government (Grants 2019111068 and 2022111067) and Inocente Inocente Foundation (FII22/001). M.P.R. is funded by a Medical Research Council Centre for Reproductive Health Grant No: MR/N022556/1. A.F. and N.R. received support from a French national research grant PHRC No. 2008/071/HP obtained by the French Institute of Cancer and the French Healthcare Organization. K.E.O. is funded by the University of Pittsburgh Medical Center and the US National Institutes of Health HD100197. V.B-L is supported by the French National Institute of Cancer (Grant Seq21-026). Y.J. is supported by the Royal Children's Hospital Foundation and a Medical Research Future Fund MRFAR000308. E.G., N.N., S.S., C.L.M., A.M.M.v.P., C.E., R.T.M., K.D., M.P.R. are members of COST Action CA20119 (ANDRONET) supported by COST (European Cooperation in Science and Technology). The Danish Child Cancer Foundation is also thanked for financial support (C.Y.A.). The authors declare no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Kathleen Duffin
- Department of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Nina Neuhaus
- Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, University Hospital of Copenhagen & Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Virginie Barraud-Lange
- Department of Reproductive Biology CECOS, AP-HP Centre—University of Paris Cité, Cochin Hospital, Paris, France
- AYA Unit, Fertility Preservation Consultation, Haematology Department, AP-HP Nord, University of Paris Cité, Saint-Louis Hospital, Paris, France
| | - Aude Braye
- Department of Genetics, Reproduction and Development (GRAD), Biology of the Testis (BITE), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Cristina Eguizabal
- Cell Therapy, Stem Cells and Tissues Group, Basque Center for Blood Transfusion and Human Tissues, Bizkaia, Spain
- Biocruces Bizkaia Health Research Institute, Bizkaia, Spain
| | - Aurélie Feraille
- NorDIC, Team “Adrenal and Gonadal Pathophysiology”, Biology of Reproduction-CECOS Laboratory, Rouen University Hospital, Université de Rouen Normandie, Rouen, France
| | - Jill P Ginsberg
- Division of Oncology, Children's Hospital of Philadelphia, Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Debra Gook
- Reproductive Services/Melbourne IVF, The Royal Women’s Hospital, Parkville, VIC, Australia
- Department of Obstetrics and Gynaecology, Royal Women’s Hospital, University of Melbourne, Parkville, VIC, Australia
| | - Ellen Goossens
- Department of Genetics, Reproduction and Development (GRAD), Biology of the Testis (BITE), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Kirsi Jahnukainen
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, NORDFERTIL Research Lab Stockholm, Karolinska Institutet and Karolinska University Hospital, Solna, Sweden
- Division of Haematology-Oncology and Stem Cell Transplantation, New Children’s Hospital, Pediatric Research Center, Department of Pediatrics, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Yasmin Jayasinghe
- Department of Obstetrics and Gynaecology, Royal Women’s Hospital, University of Melbourne, Parkville, VIC, Australia
- Oncofertility Program, Royal Children’s Hospital, Melbourne, VIC, Australia
| | - Victoria Keros
- Division of Gynecology and Reproduction, Department of Reproductive Medicine, Karolinska University Hospital, Stockholm, Sweden
- Division of Urology, Department of Clinical Science, Intervention and Technology—CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Sabine Kliesch
- Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - Sheila Lane
- Department of Paediatric Oncology and Haematology, Children’s Hospital Oxford, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Callista L Mulder
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ans M M van Pelt
- Reproductive Biology Laboratory, Center for Reproductive Medicine, Amsterdam UMC Location University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development Research Institute, Amsterdam, the Netherlands
| | - Catherine Poirot
- Fertility Preservation Consultation, Haematology Department, AYA Unit, Saint Louis Hospital, AP-HP Médecine Sorbonne Université, Paris, France
- Department of Reproductive Biology, Cochin Hospital, Paris, France
| | - Michael P Rimmer
- MRC Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Nathalie Rives
- NorDIC, Team “Adrenal and Gonadal Pathophysiology”, Biology of Reproduction-CECOS Laboratory, Rouen University Hospital, Université de Rouen Normandie, Rouen, France
| | - Hooman Sadri-Ardekani
- Department of Urology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Myriam Safrai
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Sackler Faculty of Medicine, Department of Obstetrics and Gynecology, Chaim Sheba Medical Center (Tel Hashomer), Tel Aviv University, Tel Aviv, Israel
| | - Stefan Schlatt
- Centre of Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - Jan-Bernd Stukenborg
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, NORDFERTIL Research Lab Stockholm, Karolinska Institutet and Karolinska University Hospital, Solna, Sweden
| | | | - Christine Wyns
- Department of Gynecology and Andrology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, Institute of Regeneration and Repair, University of Edinburgh, Edinburgh, UK
- Royal Hospital for Children and Young People, Edinburgh, UK
| |
Collapse
|
13
|
Song Y, Zhang X, Desmarais JA, Nagano M. Postnatal development of mouse spermatogonial stem cells as determined by immunophenotype, regenerative capacity, and long-term culture-initiating ability: a model for practical applications. Sci Rep 2024; 14:2299. [PMID: 38280889 PMCID: PMC10821885 DOI: 10.1038/s41598-024-52824-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 01/24/2024] [Indexed: 01/29/2024] Open
Abstract
Spermatogonial stem cells (SSCs) are the foundation of life-long spermatogenesis. While SSC research has advanced greatly over the past two decades, characterization of SSCs during postnatal development has not been well documented. Using the mouse as a model, in this study, we defined the immunophenotypic profiles of testis cells during the course of postnatal development using multi-parameter flow cytometry with up to five cell-surface antigens. We found that the profiles progress over time in a manner specific to developmental stages. We then isolated multiple cell fractions at different developmental stages using fluorescent-activated cell sorting (FACS) and identified specific cell populations with prominent capacities to regenerate spermatogenesis upon transplantation and to initiate long-term SSC culture. The data indicated that the cell fraction with the highest level of regeneration capacity exhibited the most prominent potential to initiate SSC culture, regardless of age. Interestingly, refinement of cell fractionation using GFRA1 and KIT did not lead to further enrichment of regenerative and culture-initiating stem cells, suggesting that when a high degree of SSC enrichment is achieved, standard markers of SSC self-renewal or commitment may lose their effectiveness to distinguish cells at the stem cell state from committed progenitors. This study provides a significant information resource for future studies and practical applications of mammalian SSCs.
Collapse
Affiliation(s)
- Youngmin Song
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada
| | - Xiangfan Zhang
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada
| | - Joëlle A Desmarais
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada
- JEFO Nutrition Inc, 5020 Avenue Jefo, Saint-Hyachinthe, Quebec, J2R 2E7, Canada
| | - Makoto Nagano
- Department of Obstetrics and Gynecology, McGill University, and the Child Health and Human Development Program, The Research Institute of the McGill University Health Centre, 1001 Decarie Boulevard, Rm# EM0.2212, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
14
|
De Windt S, Kourta D, Kanbar M, Wyns C. Optimized Recovery of Immature Germ Cells after Prepubertal Testicular Tissue Digestion and Multi-Step Differential Plating: A Step towards Fertility Restoration with Cancer-Cell-Contaminated Tissue. Int J Mol Sci 2023; 25:521. [PMID: 38203691 PMCID: PMC10779385 DOI: 10.3390/ijms25010521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Undifferentiated germ cells, including the spermatogonial stem cell subpopulation required for fertility restoration using human immature testicular tissue (ITT), are difficult to recover as they do not easily adhere to plastics. Due to the scarcity of human ITT for research, we used neonatal porcine ITT. Strategies for maximizing germ cell recovery, including a comparison of two enzymatic digestion protocols (P1 and P2) of ITT fragment sizes (4 mm3 and 8 mm3) and multi-step differential plating were explored. Cellular viability and yield, as well as numbers and proportions of DDX4+ germ cells, were assessed before incubating the cell suspensions overnight on uncoated plastics. Adherent cells were processed for immunocytochemistry (ICC) and floating cells were further incubated for three days on Poly-D-Lysine-coated plastics. Germ cell yield and cell types using ICC for SOX9, DDX4, ACTA2 and CYP19A1 were assessed at each step of the multi-step differential plating. Directly after digestion, cell suspensions contained >92% viable cells and 4.51% DDX4+ germ cells. Pooled results for fragment sizes revealed that the majority of DDX4+ cells adhere to uncoated plastics (P1; 82.36% vs. P2; 58.24%). Further incubation on Poly-D-Lysine-coated plastics increased germ cell recovery (4.80 ± 11.32 vs. 1.90 ± 2.07 DDX4+ germ cells/mm2, respectively for P1 and P2). The total proportion of DDX4+ germ cells after the complete multi-step differential plating was 3.12%. These results highlight a reduced proportion and number of germ cells lost when compared to data reported with other methods, suggesting that multi-step differential plating should be considered for optimization of immature germ cell recovery. While Poly-D-Lysine-coating increased the proportions of recovered germ cells by 16.18% (P1) and 28.98% (P2), future studies should now focus on less cell stress-inducing enzymatic digestion protocols to maximize the chances of fertility restoration with low amounts of cryo-banked human ITT.
Collapse
Affiliation(s)
- Sven De Windt
- Laboratoire d’andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (S.D.W.); (D.K.); (M.K.)
| | - Dhoha Kourta
- Laboratoire d’andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (S.D.W.); (D.K.); (M.K.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Marc Kanbar
- Laboratoire d’andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (S.D.W.); (D.K.); (M.K.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Christine Wyns
- Laboratoire d’andrologie, Pôle de Recherche en Physiologie de la Reproduction, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200 Brussels, Belgium; (S.D.W.); (D.K.); (M.K.)
- Department of Gynecology-Andrology, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
15
|
Dias Nunes J, Demeestere I, Devos M. BRCA Mutations and Fertility Preservation. Int J Mol Sci 2023; 25:204. [PMID: 38203374 PMCID: PMC10778779 DOI: 10.3390/ijms25010204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/18/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Hereditary cancers mostly affect the adolescent and young adult population (AYA) at reproductive age. Mutations in BReast CAncer (BRCA) genes are responsible for the majority of cases of hereditary breast and ovarian cancer. BRCA1 and BRCA2 act as tumor suppressor genes as they are key regulators of DNA repair through homologous recombination. Evidence of the accumulation of DNA double-strand break has been reported in aging oocytes, while BRCA expression decreases, leading to the hypothesis that BRCA mutation may impact fertility. Moreover, patients exposed to anticancer treatments are at higher risk of fertility-related issues, and BRCA mutations could exacerbate the treatment-induced depletion of the ovarian reserve. In this review, we summarized the functions of both genes and reported the current knowledge on the impact of BRCA mutations on ovarian ageing, premature ovarian insufficiency, female fertility preservation strategies and insights about male infertility. Altogether, this review provides relevant up-to-date information on the impact of BRCA1/2 mutations on fertility. Notably, BRCA-mutated patients should be adequately counselled for fertility preservation strategies, considering their higher sensitivity to chemotherapy gonadotoxic effects.
Collapse
Affiliation(s)
- Joana Dias Nunes
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (J.D.N.); (M.D.)
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (J.D.N.); (M.D.)
- Fertility Clinic, HUB-Erasme Hospital, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Melody Devos
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium; (J.D.N.); (M.D.)
| |
Collapse
|
16
|
Aponte PM, Gutierrez-Reinoso MA, Garcia-Herreros M. Bridging the Gap: Animal Models in Next-Generation Reproductive Technologies for Male Fertility Preservation. Life (Basel) 2023; 14:17. [PMID: 38276265 PMCID: PMC10820126 DOI: 10.3390/life14010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/12/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
This review aims to explore advanced reproductive technologies for male fertility preservation, underscoring the essential role that animal models have played in shaping these techniques through historical contexts and into modern applications. Rising infertility concerns have become more prevalent in human populations recently. The surge in male fertility issues has prompted advanced reproductive technologies, with animal models playing a pivotal role in their evolution. Historically, animal models have aided our understanding in the field, from early reproductive basic research to developing techniques like artificial insemination, multiple ovulation, and in vitro fertilization. The contemporary landscape of male fertility preservation encompasses techniques such as sperm cryopreservation, testicular sperm extraction, and intracytoplasmic sperm injection, among others. The relevance of animal models will undoubtedly bridge the gap between traditional methods and revolutionary next-generation reproductive techniques, fortifying our collective efforts in enhancing male fertility preservation strategies. While we possess extensive knowledge about spermatogenesis and its regulation, largely thanks to insights from animal models that paved the way for human infertility treatments, a pressing need remains to further understand specific infertility issues unique to humans. The primary aim of this review is to provide a comprehensive analysis of how animal models have influenced the development and refinement of advanced reproductive technologies for male fertility preservation, and to assess their future potential in bridging the gap between current practices and cutting-edge fertility techniques, particularly in addressing unique human male factor infertility.
Collapse
Affiliation(s)
- Pedro M. Aponte
- Colegio de Ciencias Biológicas y Ambientales (COCIBA), Universidad San Francisco de Quito (USFQ), Quito 170901, Ecuador
- Instituto de Investigaciones en Biomedicina “One-Health”, Universidad San Francisco de Quito (USFQ), Campus Cumbayá, Quito 170901, Ecuador
| | - Miguel A. Gutierrez-Reinoso
- Facultad de Ciencias Agropecuarias y Recursos Naturales, Carrera de Medicina Veterinaria, Universidad Técnica de Cotopaxi (UTC), Latacunga 050150, Ecuador;
- Laboratorio de Biotecnología Animal, Departamento de Ciencia Animal, Facultad de Ciencias Veterinarias, Universidad de Concepción (UdeC), Chillán 3780000, Chile
| | - Manuel Garcia-Herreros
- Instituto Nacional de Investigação Agrária e Veterinária (INIAV), 2005-048 Santarém, Portugal
| |
Collapse
|
17
|
Nam CS, Campbell KJ, Acquati C, Bole R, Adler A, Collins DJ, Collins E, Samplaski M, Anderson-Bialis J, Andino JJ, Asafu-Adjei D, Gaskins AJ, Bortoletto P, Vij SC, Orwig KE, Lundy SD. Deafening Silence of Male Infertility. Urology 2023; 182:111-124. [PMID: 37778476 DOI: 10.1016/j.urology.2023.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/07/2023] [Accepted: 09/23/2023] [Indexed: 10/03/2023]
Abstract
Think about 6 loved ones of reproductive age in your life. Now imagine that 1 of these 6 individuals is suffering from infertility. Perhaps they feel alone and isolated, unable to discuss their heartbreak with their closest friends, family, and support network. Suffering in silence. In this editorial, we discuss the infertility journey through the lens of the patients, the providers, and the scientists who struggle with infertility each and every day. Our goal is to open a dialogue surrounding infertility, with an emphasis on dismantling the longstanding societal barriers to acknowledging male infertility as a disease. Through education, communication, compassion, and advocacy, together we can all begin to break the deafening silence of male infertility.
Collapse
Affiliation(s)
- Catherine S Nam
- Department of Urology, University of Michigan, Ann Arbor, MI
| | | | - Chiara Acquati
- Graduate College of Social Work, University of Houston, Houston, TX; Department of Clinical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, TX; Department of Health Disparities Research, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Raevti Bole
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH
| | - Ava Adler
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - David J Collins
- Department of Urology, University of Southern California, Los Angeles, CA
| | - Erica Collins
- Department of Urology, University of Southern California, Los Angeles, CA
| | - Mary Samplaski
- Department of Urology, University of Southern California, Los Angeles, CA
| | | | - Juan J Andino
- Department of Urology, University of California Los Angeles, Los Angeles, CA
| | - Denise Asafu-Adjei
- Department of Urology, Department of Parkinson School of Health Sciences and Public Health, Loyola University Chicago Stritch School of Medicine, Chicago, IL
| | | | - Pietro Bortoletto
- Boston IVF, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Sarah C Vij
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH
| | - Kyle E Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Scott D Lundy
- Glickman Urological and Kidney Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH.
| |
Collapse
|
18
|
Lara NLM, Goldsmith T, Rodriguez-Villamil P, Ongaratto F, Solin S, Webster D, Ganbaatar U, Hodgson S, Corbière SMAS, Bondareva A, Carlson DF, Dobrinski I. DAZL Knockout Pigs as Recipients for Spermatogonial Stem Cell Transplantation. Cells 2023; 12:2582. [PMID: 37947660 PMCID: PMC10649044 DOI: 10.3390/cells12212582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/28/2023] [Accepted: 10/31/2023] [Indexed: 11/12/2023] Open
Abstract
Spermatogonial stem cell (SSC) transplantation into the testis of a germ cell (GC)-depleted surrogate allows transmission of donor genotype via donor-derived sperm produced by the recipient. Transplantation of gene-edited SSCs provides an approach to propagate gene-edited large animal models. DAZL is a conserved RNA-binding protein important for GC development, and DAZL knockout (KO) causes defects in GC commitment and differentiation. We characterized DAZL-KO pigs as SSC transplantation recipients. While there were GCs in 1-week-old (wko) KO, complete GC depletion was observed by 10 wko. Donor GCs were transplanted into 18 DAZL-KO recipients at 10-13 wko. At sexual maturity, semen and testes were evaluated for transplantation efficiency and spermatogenesis. Approximately 22% of recipient seminiferous tubules contained GCs, including elongated spermatids and proliferating spermatogonia. The ejaculate of 89% of recipients contained sperm, exclusively from donor origin. However, sperm concentration was lower than the wild-type range. Testicular protein expression and serum hormonal levels were comparable between DAZL-KO and wild-type. Intratesticular testosterone and Leydig cell volume were increased, and Leydig cell number decreased in transplanted DAZL-KO testis compared to wild-type. In summary, DAZL-KO pigs support donor-derived spermatogenesis following SSC transplantation, but low spermatogenic efficiency currently limits their use for the production of offspring.
Collapse
Affiliation(s)
- Nathalia L. M. Lara
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N4N1, Canada; (N.L.M.L.); (A.B.)
| | - Taylor Goldsmith
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | | | - Felipe Ongaratto
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | - Staci Solin
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | - Dennis Webster
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | - Uyanga Ganbaatar
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | - Shane Hodgson
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | | | - Alla Bondareva
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N4N1, Canada; (N.L.M.L.); (A.B.)
| | - Daniel F. Carlson
- Recombinetics, Inc., St. Paul, MN 55121, USA; (T.G.); (D.W.); (S.H.); (S.M.A.S.C.); (D.F.C.)
| | - Ina Dobrinski
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N4N1, Canada; (N.L.M.L.); (A.B.)
| |
Collapse
|
19
|
Younis N, Caldeira-Brant AL, Chu T, Abdalla S, Orwig KE. Human immature testicular tissue organ culture: a step towards fertility preservation and restoration. Front Endocrinol (Lausanne) 2023; 14:1242263. [PMID: 37701899 PMCID: PMC10494240 DOI: 10.3389/fendo.2023.1242263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/07/2023] [Indexed: 09/14/2023] Open
Abstract
Background Cryopreservation of immature testicular tissue (ITT) is currently the only option to preserve fertility of prepubertal patients. Autologous transplantation of ITT may not be safe or appropriate for all patients. Therefore, methods to mature ITT ex vivo are needed. Objectives Aim to investigate the feasibility of inducing in vitro spermatogenesis from ITT cryopreserved for pediatric patients prior to initiation of gonadotoxic therapy. Materials and methods Cryopreserved-thawed ITT from prepubertal and peripubertal patients were cultured for 7, 16, and 32 days in medium with no hormones or supplemented with 5 IU/L FSH, 1 IU/L hCG, or 5IU/L FSH+1 IU/L hCG. Samples were evaluated histologically to assess tissue integrity, and immunofluorescence staining was performed to identify VASA (DDX4)+ germ cells, UCHL1+ spermatogonia, SYCP3+ spermatocytes, CREM+ spermatids, SOX9+ Sertoli cells. Proliferation (KI67) and apoptosis (CASPASE3) of germ cells and Sertoli cells were also analyzed. Sertoli and Leydig cell maturation was evaluated by AR and INSL3 expression as well as expression of the blood testis barrier protein, CLAUDIN11, and testosterone secretion in the culture medium. Results Integrity of seminiferous tubules, VASA+ germ cells and SOX9+ Sertoli cells were maintained up to 32 days. The number of VASA+ germ cells was consistently higher in the peripubertal groups. UCHL1+ undifferentiated spermatogonia and SOX9+ Sertoli cell proliferation was confirmed in most samples. SYCP3+ primary spermatocytes began to appear by day 16 in both age groups. Sertoli cell maturation was demonstrated by AR expression but the expression of CLAUDIN11 was disorganized. Presence of mature and functional Leydig cells was verified by INSL3 expression and secretion of testosterone. Gonadotropin treatments did not consistently impact the number or proliferation of germ cells or somatic cells, but FSH was necessary to increase testosterone secretion over time in prepubertal samples. Conclusion ITT were maintained in organotypic culture for up to 32 days and spermatogonia differentiated to produce primary spermatocytes in both pre- and peripubertal age groups. However, complete spermatogenesis was not observed in either group.
Collapse
Affiliation(s)
- Nagham Younis
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Biological Sciences, School of Science, University of Jordan, Amman, Jordan
| | - Andre L. Caldeira-Brant
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Tianjiao Chu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Shtaywy Abdalla
- Department of Biological Sciences, School of Science, University of Jordan, Amman, Jordan
| | - Kyle E. Orwig
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
20
|
Close A, Burns K, Bjornard K, Webb M, Chavez J, Chow EJ, Meacham L. Fertility preservation in pediatric leukemia and lymphoma: A report from the Children's Oncology Group. Pediatr Blood Cancer 2023; 70:e30407. [PMID: 37194406 PMCID: PMC10525075 DOI: 10.1002/pbc.30407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/18/2023]
Abstract
Certain chemotherapy agents, radiation, and surgery can all negatively impact future fertility. Consults regarding treatment-related risk for infertility and gonadal late effects of these agents should occur at the time of diagnosis as well as during survivorship. Counseling on fertility risk has traditionally varied significantly across providers and institutions. We aim to provide a guide to standardize the assignment of gonadotoxic risk, which can be used in counseling patients both at the time of diagnosis and in survivorship. Gonadotoxic therapies were abstracted from 26 frontline Children's Oncology Group (COG) phase III protocols for leukemia/lymphoma, in use from 2000-2022. A stratification system based on gonadotoxic therapies, sex, and pubertal status was used to assign treatments into minimal, significant, and high level of increased risk for gonadal dysfunction/infertility. Risk levels were assigned to protocols and different treatment arms to aid oncologists and survivor care providers in counseling patients regarding treatment-related gonadotoxicity. Males were most commonly at high risk, with at least one high-risk arm in 14/26 protocols (54%), followed by pubertal females (23% of protocols) and prepubertal females (15% of protocols). All patients who received direct gonadal radiation or hematopoietic stem cell transplant (HSCT) were considered at high risk. Partnering with patients and their oncology/survivorship team is imperative for effective fertility counseling both prior to and post treatment, and this comprehensive guide can be used as a tool to standardize and improve reproductive health counseling in patients undergoing COG-based leukemia/lymphoma care.
Collapse
Affiliation(s)
- Allison Close
- Helen DeVos Children’s Hospital, Division of Hematology/Oncology, Michigan State University College of Human Medicine, Grand Rapids, MI, USA
| | - Karen Burns
- Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Kari Bjornard
- Department of Pediatrics, Division of Hematology/Oncology, Indiana University School of Medicine, Indianapolis, IN, USA; Riley Hospital for Children at Indiana University Health, Indianapolis, IN, USA
| | - Martine Webb
- University of California, Los Angeles, Division of Internal Medicine, Los Angeles, CA, USA
| | - Josuah Chavez
- Helen DeVos Children’s Hospital, Division of Hematology/Oncology, Grand Rapids, MI, USA
| | - Eric J. Chow
- Fred Hutchinson Cancer Center, Seattle Children’s Hospital, Seattle, WA, USA
| | - Lillian Meacham
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Pediatric Hematology/Oncology/BMT, Emory University Atlanta, GA USA
| |
Collapse
|
21
|
Whiley PAF, Nathaniel B, Stanton PG, Hobbs RM, Loveland KL. Spermatogonial fate in mice with increased activin A bioactivity and testicular somatic cell tumours. Front Cell Dev Biol 2023; 11:1237273. [PMID: 37564373 PMCID: PMC10409995 DOI: 10.3389/fcell.2023.1237273] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/13/2023] [Indexed: 08/12/2023] Open
Abstract
Adult male fertility depends on spermatogonial stem cells (SSCs) which undergo either self-renewal or differentiation in response to microenvironmental signals. Activin A acts on Sertoli and Leydig cells to regulate key aspects of testis development and function throughout life, including steroid production. Recognising that activin A levels are elevated in many pathophysiological conditions, this study investigates effects of this growth factor on the niche that determines spermatogonial fate. Although activin A can promote differentiation of isolated spermatogonia in vitro, its impacts on SSC and spermatogonial function in vivo are unknown. To assess this, we examined testes of Inha KO mice, which feature elevated activin A levels and bioactivity, and develop gonadal stromal cell tumours as adults. The GFRA1+ SSC-enriched population was more abundant and proliferative in Inha KO compared to wildtype controls, suggesting that chronic elevation of activin A promotes a niche which supports SSC self-renewal. Intriguingly, clusters of GFRA1+/EOMES+/LIN28A- cells, resembling a primitive SSC subset, were frequently observed in tubules adjacent to tumour regions. Transcriptional analyses of Inha KO tumours, tubules adjacent to tumours, and tubules distant from tumour regions revealed disrupted gene expression in each KO group increased in parallel with tumour proximity. Modest transcriptional changes were documented in Inha KO tubules with complete spermatogenesis. Importantly, tumours displaying upregulation of activin responsive genes were also enriched for factors that promote SSC self-renewal, including Gdnf, Igf1, and Fgf2, indicating the tumours generate a supportive microenvironment for SSCs. Tumour cells featured some characteristics of adult Sertoli cells but lacked consistent SOX9 expression and exhibited an enhanced steroidogenic phenotype, which could arise from maintenance or acquisition of a fetal cell identity or acquisition of another somatic phenotype. Tumour regions were also heavily infiltrated with endothelial, peritubular myoid and immune cells, which may contribute to adjacent SSC support. Our data show for the first time that chronically elevated activin A affects SSC fate in vivo. The discovery that testis stromal tumours in the Inha KO mouse create a microenvironment that supports SSC self-renewal but not differentiation offers a strategy for identifying pathways that improve spermatogonial propagation in vitro.
Collapse
Affiliation(s)
- Penny A. F. Whiley
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Benedict Nathaniel
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Peter G. Stanton
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Robin M. Hobbs
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Kate L. Loveland
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Sciences, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
22
|
Sciorio R, Manna C, Fauque P, Rinaudo P. Can Cryopreservation in Assisted Reproductive Technology (ART) Induce Epigenetic Changes to Gametes and Embryos? J Clin Med 2023; 12:4444. [PMID: 37445479 DOI: 10.3390/jcm12134444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 06/05/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Since the birth of Louise Brown in 1978, more than nine million children have been conceived using assisted reproductive technologies (ARTs). While the great majority of children are healthy, there are concerns about the potential epigenetic consequences of gametes and embryo manipulation. In fact, during the preimplantation period, major waves of epigenetic reprogramming occur. Epigenetic reprogramming is susceptible to environmental changes induced by ovarian stimulation, in-vitro fertilization, and embryo culture, as well as cryopreservation procedures. This review summarizes the evidence relating to oocytes and embryo cryopreservation and potential epigenetic regulation. Overall, it appears that the stress induced by vitrification, including osmotic shock, temperature and pH changes, and toxicity of cryoprotectants, might induce epigenetic and transcriptomic changes in oocytes and embryos. It is currently unclear if these changes will have potential consequences for the health of future offspring.
Collapse
Affiliation(s)
- Romualdo Sciorio
- Edinburgh Assisted Conception Programme, Royal Infirmary of Edinburgh, Edinburgh EH16 4SA, UK
| | - Claudio Manna
- Biofertility IVF and Infertility Center, 00198 Rome, Italy
| | - Patricia Fauque
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Development (GAD) INSERM UMR1231, F-21000 Dijon, France
- CHU Dijon Bourgogne, Laboratoire de Biologie de la Reproduction-CECOS, F-21000 Dijon, France
| | - Paolo Rinaudo
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, CA 92037, USA
| |
Collapse
|
23
|
Antonouli S, Di Nisio V, Messini C, Daponte A, Rajender S, Anifandis G. A comprehensive review and update on human fertility cryopreservation methods and tools. Front Vet Sci 2023; 10:1151254. [PMID: 37143497 PMCID: PMC10151698 DOI: 10.3389/fvets.2023.1151254] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023] Open
Abstract
The broad conceptualization of fertility preservation and restoration has become already a major concern in the modern western world since a large number of individuals often face it in the everyday life. Driven by different health conditions and/or social reasons, a variety of patients currently rely on routinely and non-routinely applied assisted reproductive technologies, and mostly on the possibility to cryopreserve gametes and/or gonadal tissues for expanding their reproductive lifespan. This review embraces the data present in human-focused literature regarding the up-to-date methodologies and tools contemporarily applied in IVF laboratories' clinical setting of the oocyte, sperm, and embryo cryopreservation and explores the latest news and issues related to the optimization of methods used in ovarian and testicular tissue cryopreservation.
Collapse
Affiliation(s)
- Sevastiani Antonouli
- Department of Clinical Chemistry, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Valentina Di Nisio
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Christina Messini
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| | - Alexandros Daponte
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| | - Singh Rajender
- Division of Endocrinology, Central Drug Research Institute, Lucknow, India
| | - George Anifandis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larisa, Greece
| |
Collapse
|
24
|
Marques R, Oura MJ, Dionísio R, Rodrigues C, Lorenzo J. Oncofertility in pediatric patients: current perspectives. Contemp Oncol (Pozn) 2022; 26:165-173. [PMID: 36381665 PMCID: PMC9641635 DOI: 10.5114/wo.2022.120362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/14/2022] [Indexed: 10/27/2024] Open
Abstract
Cancer treatments may affect fertility in different ways. Oncofertility, i.e. the study of interactions between cancer, anti-cancer therapy, fertility, and reproductive health, is an emerging field that addresses cancer patients' concerns regarding their future reproductive ability. As the number of cancer survivors increases, fertility preservation is becoming an important quality of life issue for many survivors of childhood cancer. There is a wide array of fertility preservation options according to gender and pubertal status, and shared decisions must take place at the time of diagnosis. Even though there might be several barriers that can negatively affect this process, the presence of a dedicated fertility preservation team may help overcome them. In this article, the authors aim to characterize what oncofertility is, the effects of cancer and its treatments on the fertility potential of pediatric patients and also on their mental health. Another goal is to expose the dif- ferent fertility preservation therapeutic options and potential barriers.
Collapse
Affiliation(s)
- Rafael Marques
- Centro Hospitalar Entre o Douro e Vouga, Santa Maria da Feira, Portugal
| | | | | | | | - Joana Lorenzo
- Centro Hospitalar Entre o Douro e Vouga, Santa Maria da Feira, Portugal
| |
Collapse
|
25
|
Holt WV, Comizzoli P. Conservation Biology and Reproduction in a Time of Developmental Plasticity. Biomolecules 2022; 12:1297. [PMID: 36139136 PMCID: PMC9496186 DOI: 10.3390/biom12091297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/24/2022] Open
Abstract
The objective of this review is to ask whether, and how, principles in conservation biology may need to be revisited in light of new knowledge about the power of epigenetics to alter developmental pathways. Importantly, conservation breeding programmes, used widely by zoological parks and aquariums, may appear in some cases to reduce fitness by decreasing animals' abilities to cope when confronted with the 'wild side' of their natural habitats. Would less comfortable captive conditions lead to the selection of individuals that, despite being adapted to life in a captive environment, be better able to thrive if relocated to a more natural environment? While threatened populations may benefit from advanced reproductive technologies, these may actually induce undesirable epigenetic changes. Thus, there may be inherent risks to the health and welfare of offspring (as is suspected in humans). Advanced breeding technologies, especially those that aim to regenerate the rarest species using stem cell reprogramming and artificial gametes, may also lead to unwanted epigenetic modifications. Current knowledge is still incomplete, and therefore ethical decisions about novel breeding methods remain controversial and difficult to resolve.
Collapse
Affiliation(s)
- William V. Holt
- Department of Oncology & Metabolism, The Medical School Beech Hill Road, Sheffield S10 2RX, UK
| | - Pierre Comizzoli
- Smithsonian’s National Zoo and Conservation Biology Institute, Washington, DC 20008, USA
| |
Collapse
|