1
|
Chung N, Yang C, Yang H, Shin J, Song CY, Min H, Kim JH, Lee K, Lee JR. Local delivery of platelet-derived factors mitigates ischemia and preserves ovarian function through angiogenic modulation: A personalized regenerative strategy for fertility preservation. Biomaterials 2025; 313:122768. [PMID: 39232332 DOI: 10.1016/j.biomaterials.2024.122768] [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: 06/19/2024] [Revised: 08/11/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
As the most prominent and ideal modality in female fertility preservation, ovarian tissue cryopreservation, and transplantation often confront the challenge of ischemic damage and follicular loss from avascular transplantation. To surmount this impediment, we engineered a novel platelet-derived factors-encapsulated fibrin hydrogel (PFH), a paradigmatic biomaterial. PFH encapsulates autologous platelet-derived factors, utilizing the physiological blood coagulation cascade for precise local delivery of bioactive molecules. In our study, PFH markedly bolstered the success of avascular ovarian tissue transplantation. Notably, the quantity and quality of follicles were preserved with improved neovascularization, accompanied by decreased DNA damage, increased ovulation, and superior embryonic development rates under a Low-concentration Platelet-rich plasma-derived factors encapsulated fibrin hydrogel (L-PFH) regimen. At a stabilized point of tissue engraftment, gene expression analysis mirrored normal ovarian tissue profiles, underscoring the effectiveness of L-PFH in mitigating the initial ischemic insult. This autologous blood-derived biomaterial, inspired by nature, capitalizes on the blood coagulation cascade, and combines biodegradability, biocompatibility, safety, and cost-effectiveness. The adjustable properties of this biomaterial, even in injectable form, extend its potential applications into the broader realm of personalized regenerative medicine. PFH emerges as a promising strategy to counter ischemic damage in tissue transplantation, signifying a broader therapeutic prospect. (197 words).
Collapse
Affiliation(s)
- Nanum Chung
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Chungmo Yang
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea; Program in Nanoscience and Technology, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea
| | - Heeseon Yang
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Jungwoo Shin
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Chae Young Song
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Hyewon Min
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Ji Hyang Kim
- Department of Obstetrics and Gynecology, Fertility Center of CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, 13496, Republic of Korea.
| | - Kangwon Lee
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826, Republic of Korea.
| | - Jung Ryeol Lee
- Department of Translational Medicines, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
2
|
León-Félix CM, Ouni E, Herinckx G, Vertommen D, Amorim CA, Lucci CM. Decellularized extracellular matrix from bovine ovarian tissue maintains the protein composition of the native matrisome. J Proteomics 2025; 311:105347. [PMID: 39521401 DOI: 10.1016/j.jprot.2024.105347] [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: 08/27/2024] [Revised: 10/15/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Recent approaches of regenerative reproductive medicine investigate the decellularized extracellular matrix to develop a transplantable engineered ovary (TEO). However, a full proteomic analysis is not usually performed after the decellularization process to evaluate the preservation of the extracellular matrix (ECM). In this study, the ECM of the bovine ovarian cortex was analyzed before and after decellularization using mass spectrometry and bioinformatics. A total of 155 matrisome proteins were identified in the native ECM of the bovine ovarian cortex, with 145 matrisome proteins detected in the decellularized ECM. After decellularization, only 10 matrisome proteins were lost, and notably, none belonged to the category of reproductive biological processes. Histology and histochemistry analyses were employed to assess the general morphology of both native and decellularized ECM, allowing for the identification of the most abundant ECM proteins. Moreover, our study highlighted collagen type VI alpha 3 and heparan sulfate proteoglycan 2 as the most abundant components in the bovine ovarian ECM, mirroring the composition observed in the human ovary. These findings enhance our understanding of the composition of both native and decellularized ECM, with the potential implications for the development of a TEO. SIGNIFICANCE: The significance of the present study lies on the possibility of advancing towards developing a bioengineered ovary, which is the ultimate strategy to regain fertility in women. The results demonstrate that the decellularized extracellular matrix of the bovine ovary maintains the protein composition of the native matrisome, using a recently described decellularization protocol. The decellularized matrix may serve as scaffolding for seeding ovarian stromal cells and follicles to create a bioengineered ovary, and as closer its composition is to the native matrix the better. Also, comparing the bovine ovarian matrisome, which was described for the first time here, with the human ovarian matrisome, we could see a great similarity, suggesting that the bovine ovary decellularized matrix may serve as a model for developing a human bioengineered ovary.
Collapse
Affiliation(s)
- Cecibel M León-Félix
- Institute of Biological Sciences, Department of Physiology, University of Brasilia, Brasilia 70910-900, Brazil
| | - Emna Ouni
- Tumor Cell Dynamics Unit, Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif 94800, France
| | - Gaëtan Herinckx
- PHOS Unit and MASSPROT Platform de Duve Institute, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Didier Vertommen
- PHOS Unit and MASSPROT Platform de Duve Institute, Université Catholique de Louvain, Brussels 1200, Belgium
| | - Christiani A Amorim
- Pôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels 1200, Belgium.
| | - Carolina M Lucci
- Institute of Biological Sciences, Department of Physiology, University of Brasilia, Brasilia 70910-900, Brazil.
| |
Collapse
|
3
|
Tsilionis V, Moustakli E, Dafopoulos S, Zikopoulos A, Sotiriou S, Zachariou A, Dafopoulos K. Reproductive Health in Women with Major β-Thalassemia: Evaluating Ovarian Reserve and Endocrine Complications. Metabolites 2024; 14:717. [PMID: 39728498 DOI: 10.3390/metabo14120717] [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: 11/18/2024] [Revised: 12/12/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
Thalassemia is an autosomal recessive hereditary chronic hemolytic anemia characterized by a partial or complete deficiency in the synthesis of alpha- or beta-globin chains, which are essential components of adult hemoglobin. Mutations in the globin genes lead to the production of unstable globin chains that precipitate within cells, causing hemolysis. This shortens the lifespan of mature red blood cells (RBCs) and results in the premature destruction of RBC precursors in the bone marrow. Regular red blood cell transfusions are the standard treatment for thalassemia. However, these transfusions can lead to increased iron overload, which can impair vital systems such as the liver, heart, ovaries, and endocrine system. Focusing on female reproductive endocrinology, recurrent blood transfusions can cause iron accumulation in the pituitary and hypothalamus, leading to hypogonadotropic hypogonadism (HH), the most common endocrinopathy in these patients, affecting 40-91% of women. Recurrent transfusions and the resulting iron overload can also lead to oxidative stress and ovarian damage in patients with beta-thalassemia major (BTM). Despite advancements in iron chelation therapy, hypothalamic-pituitary damage associated with HH contributes to subfertility and sexual dysfunction, often with little to no recovery. In women exposed to gonadotoxic drugs, particularly those with BTM, anti-Mullerian hormone (AMH)-a marker of ovarian reserve-is frequently used to assess ovarian damage. This review aims to explore the pathophysiology of β-thalassemia and its major clinical manifestations, with a focus on endocrine complications and their impact on ovarian reserve. It also investigates how metabolomics can provide insights into the disease's metabolic alterations and inform current and emerging therapeutic strategies to mitigate complications and optimize patient outcomes, potentially leading to more effective and personalized treatments.
Collapse
Affiliation(s)
- Vasileios Tsilionis
- Department of Obstetrics and Gynecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece
| | - Efthalia Moustakli
- Laboratory of Medical Genetics, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Stefanos Dafopoulos
- Department of Health Sciences, European University Cyprus, Nicosia 2404, Cyprus
| | - Athanasios Zikopoulos
- Obstetrics and Gynecology, Royal Devon and Exeter Hospital Barrack Rd., Exeter EX2 5DW, UK
| | - Sotirios Sotiriou
- Department of Embryology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece
| | - Athanasios Zachariou
- Department of Urology, School of Medicine, Ioannina University, 45110 Ioannina, Greece
| | - Konstantinos Dafopoulos
- IVF Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece
| |
Collapse
|
4
|
Yang J, Li W, Zhang Z, Xu Z, Zhu W, Wang J, Wang W. Development and Applications of Organoids in Gynecological Diseases. Stem Cell Rev Rep 2024:10.1007/s12015-024-10833-0. [PMID: 39666266 DOI: 10.1007/s12015-024-10833-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Organoids are rapidly self-organizing 3D in vitro cultures derived from pluripotent stem cells (PSCs) or adult stem cells (ASCs) that possess disease-like characteristics with high success rates. Due to their ability to retain tissue structure, biological phenotypes, and genetic information, they have been utilized as a novel in vitro model for disease research. In recent years, scientists have established self-organizing 3D organoids for human endometrium, fallopian tubes, ovaries, and cervix by culturing stem cells with cytokines in 3D scaffolds. The integration of organoids with animal models, organ-on-a-chip systems, and 3D printing technologies offers a novel preclinical model for exploring disease mechanisms and developing treatments. This review elaborate on the recent research progress of stem cells-formed organoids in the field of gynecology from the aspects of constructing gynecological disease organoids, drug screening and new drug development, simulation modeling, allogeneic transplantation, regenerative medicine and personalized treatment."
Collapse
Affiliation(s)
- Jian Yang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenwen Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zihan Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhonglei Xu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenjing Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Anhui Women and Children's Medical Center, Hefei, Anhui, China
| | - Wenyan Wang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
5
|
Cathelain A, Keller L, Collinet P, Kerbage Y, d'Orazio E, Piver P, Pigny P, Decanter C. Folliculogenesis resumption after ovarian cortex transplantation: what is the earliest hormonal indicator? Arch Gynecol Obstet 2024; 310:3209-3218. [PMID: 39613984 DOI: 10.1007/s00404-024-07810-w] [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: 03/04/2024] [Accepted: 10/23/2024] [Indexed: 12/01/2024]
Abstract
INTRODUCTION Ovarian tissue cryopreservation (OTC) is recommended by scientific societies for women undergoing highly gonadotoxic cancer treatments. Following transplantation, the restoration of ovarian function is typically characterised by the resumption of spontaneous menstruation. Yet, a few studies have looked at the longitudinal hormonal variations following transplantation. This study aims to investigate the fluctuation of gonadotropins and granulosa/theca cells secretions during the interval between ovarian transplantation and the recovery of menstrual function in two young women with no residual ovarian activity. METHOD We selected two patients diagnosed with Hodgkin's lymphoma, initially referred for OTC at the ages of 19 and 15, respectively, and who had both undergone two consecutive stem cell transplants due to recurrent disease episodes. Both patients presented with premature ovarian failure and returned at ages 29 and 26, respectively, for ovarian cortex transplantation. Hormonal secretions and menstrual function were closely monitored both prior and in the months following the ovarian transplantation. RESULTS Menstruation resumed at 7 and 5 months post-transplantation, respectively. FSH and LH levels significantly decreased as early as 1 and 3 months before the first menstruation. As for ovarian hormonal secretion, AMH, measured with an ultra-sensitive assay ("pico AMH"), and Inhibin B were the first to increase, starting 1 month before the resumption of menstruation. Subsequently, AMH levels consistently remained very low throughout the follow-up, as did androgens, which showed a slight increase after the graft but remained at postmenopausal levels. CONCLUSION Pico AMH, measured by an ultra-sensitive assay, Inhibin B and estradiol are the first ovarian hormones to be secreted following an ovarian graft, with levels rising 1 month prior the return of menstruation. However, the earliest hormonal indicators of graft success are the significant drops in FSH and LH levels, accompanied by a rise in estradiol levels, which occur 1-3 months before menstruation resumes.
Collapse
Affiliation(s)
- Alice Cathelain
- Department of Assisted Reproductive Technologies and Fertility Preservation, Jeanne de Flandre Hospital, CHU Lille, 59000, Lille, France.
- Department of Gynaecological Surgery, CHU Lille, 59000, Lille, France.
| | - Laura Keller
- Institut de Biologie de la Reproduction-Spermiologie-Centre d'étude et de Conservation des Oeufs et du Sperme Humain (CECOS), Hôpital Jeanne de Flandre, CHU Lille, Lille, France
| | - Pierre Collinet
- Department of Gynaecological Surgery, CHU Lille, 59000, Lille, France
- Univ. Lille, Faculty of Medicine, Lille, France
| | - Yohan Kerbage
- Department of Gynaecological Surgery, CHU Lille, 59000, Lille, France
- Univ. Lille, Faculty of Medicine, Lille, France
| | - Emmanuelle d'Orazio
- Department of Assisted Reproductive Technologies and Fertility Preservation, Jeanne de Flandre Hospital, CHU Lille, 59000, Lille, France
| | - Pascal Piver
- Department of Gynecology and Obstetrics, Mother and Child Hospital, University Hospital Center of Limoges, 87000, Limoges, France
| | - Pascal Pigny
- Service de Biochimie et Hormonologie, Centre de Biologie Pathologie, CHU Lille, Lille, France
| | - Christine Decanter
- Department of Assisted Reproductive Technologies and Fertility Preservation, Jeanne de Flandre Hospital, CHU Lille, 59000, Lille, France
| |
Collapse
|
6
|
Stewart S, White A, Ou W, Liu W, Nagashima J, Songsasen N, He X. Controlled Ice Nucleation With a Sand-PDMS Film Device Enhances Cryopreservation of Mouse Preantral Ovarian Follicles. J Med Device 2024; 18:041007. [PMID: 39465055 PMCID: PMC11500804 DOI: 10.1115/1.4066445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/28/2024] [Indexed: 10/29/2024] Open
Abstract
Ovarian follicle cryopreservation is a promising strategy for fertility preservation; however, cryopreservation protocols have room for improvement to maximize post-thaw follicle viability and quality. Current slow-freezing protocols use either manual ice-seeding in combination with expensive programmable-rate freezers or other clinically incompatible ice initiators to control the ice-seeding temperature in the extracellular solution, a critical parameter that impacts post-cryopreservation cell/tissue quality. Previously, sand has been shown to be an excellent, biocompatible ice initiator, and its use in cryopreservation of human induced pluripotent stem cells enables high cell viability and quality after cryopreservation. This study applies sand as an ice initiator to cryopreserve multicellular microtissue, preantral ovarian follicles, using a simple slow-freezing protocol in the mouse model. Ovarian follicles cryopreserved using the sand partially embedded in polydimethylsiloxane (PDMS) film to seed ice in the extracellular solution exhibit healthy morphology, high viability, and the ability to grow similarly to fresh follicles in culture post-thaw. This sand-based cryopreservation strategy can facilitate convenient ovarian follicle cryopreservation using simple equipment, and this study further demonstrates the translatability of this strategy to not only single cells but also multicellular tissues.
Collapse
Affiliation(s)
- Samantha Stewart
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Alisa White
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Wenquan Ou
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Wei Liu
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
| | - Jennifer Nagashima
- Center for Species Survival, Smithsonian National Zoo and Conservation Biology Institute, 1500 Remount Road, Front Royal, VA 22630
| | - Nucharin Songsasen
- Center for Species Survival, Smithsonian National Zoo and Conservation Biology Institute, 1500 Remount Road, Front Royal, VA 22630
| | - Xiaoming He
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
7
|
Ruan X. Standardization for ovarian tissue cryopreservation and transplantation in China. Gynecol Endocrinol 2024; 40:2431223. [PMID: 39616622 DOI: 10.1080/09513590.2024.2431223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/17/2024] [Accepted: 11/13/2024] [Indexed: 12/18/2024] Open
Abstract
Ovarian tissue cryopreservation and transplantation is one of the most advanced and promising fertility preservation methods. Prior to any procedure that may lead to a toxic ovarian injury (such as chemotherapy or radiotherapy), a portion of the ovary is removed and cryopreserved. At an appropriate time, after toxic therapy is concluded, the cryopreserved ovarian tissue is then thawed and transplanted back to the patient when conditions permit. This technique can not only preserve female fertility but also restore ovarian endocrine function. However, there is no standardization for ovarian tissue cryopreservation and transplantation in China. In order to promote the standardized development of ovarian tissue cryopreservation technology in the whole country, it is urgent to establish the standard of this technology.
Collapse
Affiliation(s)
- Xiangyan Ruan
- Department of Gynecological Endocrinology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University. Beijing Maternal and Child Health Care Hospital, Beijing, China
- Department of Women's Health, University Women's Hospital and Research Centre for Women's Health, University Hospitals of Tuebingen, Tuebingen, Germany
| |
Collapse
|
8
|
Li N, Du X, Zhao Y, Zeng Q, Han C, Xiong D, He L, Zhang G, Liu W. Exploring stem cell technology: Pioneering new pathways for female fertility preservation and restoration. Reprod Biol 2024; 24:100958. [PMID: 39393314 DOI: 10.1016/j.repbio.2024.100958] [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: 05/03/2024] [Revised: 09/24/2024] [Accepted: 09/28/2024] [Indexed: 10/13/2024]
Abstract
The fertility of women is crucial for the well-being of individuals and families. However, various factors such as chemotherapy, lifestyle changes, among others, may lead to a decline in female fertility, thus emphasizing the significance of preserving and restoring fertility. Stem cells, with their unique capacity for self-renewal and pluripotent differentiation, have made significant strides in areas such as ovarian tissue cryopreservation, in vitro culture of frozen-thawed ovarian tissue, and construction of ovarian-like organs. This review aims to summarize the latest findings in these fields, highlighting the pivotal role, mechanisms, and future prospects of stem cell technology in preserving and restoring female fertility. Additionally, the importance of interdisciplinary collaboration is underscored, as personalized stem cell therapy regimens tailored through interdisciplinary cooperation between reproductive medicine and stem cell fields hold promise in providing reliable solutions for the preservation and restoration of female fertility.
Collapse
Affiliation(s)
- Ningjing Li
- School of Medicine and life sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xinrong Du
- School of Medicine and life sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuhong Zhao
- College of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, China
| | - Qin Zeng
- Sichuan Provincial Woman's and Children's Hospital / The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China
| | - Changli Han
- School of Medicine and life sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dongsheng Xiong
- Sichuan Provincial Woman's and Children's Hospital / The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China
| | - Libing He
- Sichuan Provincial Woman's and Children's Hospital / The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China
| | - Guohui Zhang
- Sichuan Provincial Woman's and Children's Hospital / The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China.
| | - Weixin Liu
- Sichuan Provincial Woman's and Children's Hospital / The Affiliated Women's and Children's Hospital of Chengdu Medical College, Chengdu 610045, China.
| |
Collapse
|
9
|
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
|
10
|
Vieira ARS, de Sousa FC, Barros CHSC, Santana MJ, Alves BG, Teixeira DÍA. Color Doppler Ultrasonographic Examination of Ovarian Grafts in Goats. Vet Sci 2024; 11:580. [PMID: 39591354 PMCID: PMC11598874 DOI: 10.3390/vetsci11110580] [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: 08/20/2024] [Revised: 10/17/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024] Open
Abstract
The aim of this study was to evaluate the effectiveness of color Doppler ultrasonography for examination of the blood flow areas in superficial grafts after 7 or 15 days of heterotopic allotransplantation, comparing an in-ear subcutaneous area (IE) with an in-neck cervical intramuscular area (IN) in goats. To monitor the local blood flow in the graft areas, color Doppler signals were assessed daily until days seven and fifteen, when the left and right implants, respectively, were removed. The areas of blood flow around the transplanted ovarian fragments were significantly larger (p < 0.05) in the IE area compared to the IN area after 7 (IE: 4.70 ± 0.33A vs. IN: 3.67 ± 0.33B) and 15 (IE: 5.27 ± 0.21A vs. IN: 4.66 ± 0.22B) days of transplantation. A positive and significant correlation was observed between the area of blood flow and the day of assessment after 7 (IE: r = 0.43; p < 0.05) and 15 (IE: r = 0.52; p = 0.001; IN: r = 0.42; p = 0.001) days of transplantation. In conclusion, color Doppler ultrasonography can be used for real-time assessment of local blood perfusion in ovarian grafts, making it possible to identify alterations in the blood flow area in the period following a transplant procedure.
Collapse
Affiliation(s)
- Antonio Renilson Sousa Vieira
- Laboratory of Diagnostic Imaging Applied to Animal Reproduction, State University of Ceará—UECE, Av. Silas Munguba, 1700, Campus Itaperi, Fortaleza 60714-903, CE, Brazil
- INTA University Center—UNINTA, Rua Lucimar, 637, Dom Expedito, Campus Sobral, Sobral 62050-140, CE, Brazil
| | - Francisco Carlos de Sousa
- Federal Institute of Education, Science and Technology of Ceará—IFCE, R. Carlos Antônio Sáles, s/n—Floresta, Campus Umirim, Limoeiro do Norte 62660-000, CE, Brazil
| | | | - Maria Janiele Santana
- Laboratory of Diagnostic Imaging Applied to Animal Reproduction, State University of Ceará—UECE, Av. Silas Munguba, 1700, Campus Itaperi, Fortaleza 60714-903, CE, Brazil
| | | | - Dárcio Ítalo Alves Teixeira
- Laboratory of Diagnostic Imaging Applied to Animal Reproduction, State University of Ceará—UECE, Av. Silas Munguba, 1700, Campus Itaperi, Fortaleza 60714-903, CE, Brazil
| |
Collapse
|
11
|
Elsherbiny NM, Abdel-Maksoud MS, Prabahar K, Mohammedsaleh ZM, Badr OAM, Dessouky AA, Salem HA, Refadah OA, Farid AS, Shamaa AA, Ebrahim N. MSCs-derived EVs protect against chemotherapy-induced ovarian toxicity: role of PI3K/AKT/mTOR axis. J Ovarian Res 2024; 17:222. [PMID: 39529187 PMCID: PMC11552115 DOI: 10.1186/s13048-024-01545-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Chemotherapy detrimentally impacts fertility via depletion of follicular reserves in the ovaries leading to ovarian failure (OF) and development of estrogen deficiency-related complications. The currently proposed options to preserve fertility such as Oocyte or ovarian cortex cryopreservation are faced with many technical obstacles that limit their effective implementation. Therefore, developing new modalities to protect ovarian function remains a pending target. Exosomes are nano-sized cell-derived extracellular vesicles (EVs) with documented efficacy in the field of regenerative medicine. The current study sought to determine the potential beneficial effects of mesenchymal stem cells (MSCs)-derived EVs in experimentally induced OF. Female albino rats were randomly allocated to four groups: control, OF group, OF + MSCs-EVs group, OF + Rapamycin (mTOR inhibitor) group, and OF + Quercetin (PI3K/AKT inhibitor) group. Follicular development was assessed via histopathological and immunohistochemical examination, and ovarian function was evaluated by hormonal assay. PI3K/Akt/mTOR signaling pathway as a key modulator of ovarian follicular activation was also assessed. MSCs-EVs administration to OF rats resulted in restored serum hormonal levels, preserved primordial follicles and oocytes, suppressed ovarian PI3K/AKT axis and downstream effectors (mTOR and FOXO3), modulated miRNA that target this axis, decreased expression of ovarian apoptotic markers (BAX, BCl2) and increased expression of proliferation marker Ki67. The present study validated the effectiveness of MSCs-EVs therapy in preventing ovarian insufficiency induced by chemotherapy. Concomitant MSCs-EVs treatment during chemotherapy could significantly preserve ovarian function and fertility by suppressing the PI3K/Akt axis, preventing follicular overactivation, maintaining normal ovarian cellular proliferation, and inhibiting granulosa cell apoptosis.
Collapse
Affiliation(s)
- Nehal M Elsherbiny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.
| | - Mohamed S Abdel-Maksoud
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Kousalya Prabahar
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Zuhair M Mohammedsaleh
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, 71491, Saudi Arabia
| | - Omnia A M Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Benha, Egypt
| | - Arigue A Dessouky
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Hoda A Salem
- Department of Pharmacy Practice, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Omnia A Refadah
- PharmD Program, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
| | - Ayman Samir Farid
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Toukh, Qalyubia, 13736, Egypt
| | - Ashraf A Shamaa
- Surgery, Anesthesiology and Radiology Department, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | - Nesrine Ebrahim
- Department of Medical Histology and Cell Biology Faculty of Medicine, Benha University, Benha, Egypt.
- Stem Cell Unit, Faculty of Medicine, Benha University, Benha, Egypt.
- Faculty of Medicine, Benha National University, Al Obour City, Egypt.
- Cell and Tissue Engineering, School of Pharmacy and Bioengineering, Keele University, Keele, UK.
| |
Collapse
|
12
|
Dunlop CE, Anderson RA. Clinical dilemmas in ovarian tissue cryopreservation. Fertil Steril 2024; 122:559-564. [PMID: 38825305 DOI: 10.1016/j.fertnstert.2024.05.167] [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: 05/05/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/04/2024]
Abstract
Ovarian tissue cryopreservation (OTC) is increasingly offered globally as a fertility preservation strategy for both postpubertal women and prepubertal girls, with subsequent reimplantation of cryopreserved ovarian cortex resulting in a rapidly growing number of live births. There remains very limited evidence of efficacy from tissue stored when the patient was prepubertal or from conditions affecting the ovary directly, e.g., Turner syndrome. Although OTC is becoming a more established practice, several clinical dilemmas remain from a practical and ethical standpoint. This review discusses the challenges regarding optimal patient selection for the procedure, the use of OTC in patients with a poor prognosis, the potential of reimplantation of tissue contaminated with malignant cells, and the role of OTC in those with an intrinsic ovarian disorder.
Collapse
Affiliation(s)
- Cheryl E Dunlop
- Obstetrics & Gynaecology Department, Edinburgh Royal Infirmary, Edinburgh, United Kingdom.
| | - Richard A Anderson
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
13
|
Paik H, Hee Hong Y, Eun Kim T, Gyeong Jeong H, Ki Kim S, Ryeol Lee J. Factors associated with successful intraoperative oocyte retrieval for fertility preservation during open pelvic surgery for gynecologic indications. Eur J Obstet Gynecol Reprod Biol 2024; 301:43-48. [PMID: 39088939 DOI: 10.1016/j.ejogrb.2024.07.055] [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: 11/02/2023] [Revised: 05/09/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
OBJECTIVE The study investigated factors associated with successful intra-operative oocyte retrieval for fertility preservation during transabdominal gynecologic surgery. STUDY DESIGN A total of 29 patients who underwent intraoperative oocyte retrieval during staging surgery at a single academic hospital from May 2014 to August 2022 were enrolled in this study, and their outcomes were analyzed. RESULTS Among 29 patients who underwent intra-operative oocyte retrieval during staging surgery, oocytes were obtained in 24 patients, representing 82.8 % of the retrieval rate (24/29), and two patients returned to use cryopreserved oocytes (6.9 %). Among 24 women who succeeded in obtaining oocytes, 20 patients succeeded in oocyte cryopreservation, and two patients proceeded to embryo cryopreservation. The cryopreservation rate was 91.7 % (22/24). All patients with failed oocyte retrieval (n = 5) and cryopreservation (n = 7) were diagnosed with malignancy. AMH of those with successful cryopreservation oocytes was higher than those without cryopreservation (4.10 ng/mL vs. 1.18 ng/mL, p = 0.003). A higher portion of the unstimulated cycle was observed in those with failed cryopreservation (8.3 % vs. 40.0 %, p = 0.01). No complications were noted. CONCLUSION For women planning to undergo open pelvic surgery, intra-operative oocyte retrieval is a feasible option. High serum AMH and ovarian stimulation before surgery may predict successful oocyte cryopreservation.
Collapse
Affiliation(s)
- Haerin Paik
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yeon Hee Hong
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Tae Eun Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hye Gyeong Jeong
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Obstetrics and Gynecology, Korea University Anam Hospital, Seoul, Republic of Korea
| | - Seul Ki Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jung Ryeol Lee
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Gotschel F, Sonigo C, Becquart C, Sellami I, Mayeur A, Grynberg M. New Insights on In Vitro Maturation of Oocytes for Fertility Preservation. Int J Mol Sci 2024; 25:10605. [PMID: 39408934 PMCID: PMC11477201 DOI: 10.3390/ijms251910605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
In the last decade, the evolution of oncofertility has sparked a resurgence of interest in in vitro maturation (IVM) due to its suitability in certain oncological scenarios where controlled ovarian hyperstimulation may not be feasible. The retrieval of immature cumulus-oocyte complexes from small antral follicles, regardless of the menstrual cycle phase, presents a swift opportunity to vitrify mature oocytes or embryos post-IVM in urgent situations or when stimulation is not advisable. Harvesting immature cumulus-oocyte complexes and immature oocytes can be achieved transvaginally or directly in the laboratory from extracorporeal ovarian tissue. Although IVM has transitioned from an experimental status due to safety validations, it relies on the intricate process of oocyte maturation. Despite successful live births resulting from IVM in fertility preservation contexts, the comparatively lower developmental competence of in vitro matured oocytes highlights the necessity to enhance IVM culture systems. Recent advancements in IVM systems hold promise in bolstering oocyte competence post-IVM, thereby narrowing the gap between IVM and outcomes from ovarian stimulation. Additionally, for optimizing the chances of conception in cancer survivors, the combination of IVM and ovarian tissue cryopreservation stands as the favored choice when ovarian stimulation is unfeasible.
Collapse
Affiliation(s)
- Flavie Gotschel
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
| | - Charlotte Sonigo
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
- Inserm, Physiologie et Physiopathologie Endocrinienne, Université Paris-Saclay, 94276 Le Kremlin-Bicêtre, France
| | - Celeste Becquart
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
| | - Ines Sellami
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
| | - Anne Mayeur
- Histology-Embryology-Cytogenetic Laboratory, Université Paris-Saclay, Assistance Publique Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France;
| | - Michael Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Université Paris-Saclay, Assistance Publique-Hôpitaux de Paris, Antoine Beclère Hospital, 92140 Clamart, France; (F.G.); (C.B.); (I.S.)
- Department of Reproductive Medicine and Fertility Preservation, Hôpitaux Universitaires Paris-Seine-Saint-Denis, Assistance Publique-Hôpitaux de Paris, Jean Verdier Hospital, 93143 Bondy, France
- Unité Inserm U1133, Université Paris-Diderot, 75013 Paris, France
| |
Collapse
|
15
|
Gadek LM, Joswiak C, Laronda MM. Thawing fertility: a view of ovarian tissue cryopreservation processes and review of ovarian transplant research. Fertil Steril 2024; 122:574-585. [PMID: 38992745 DOI: 10.1016/j.fertnstert.2024.07.005] [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: 05/21/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024]
Abstract
Individuals with a disease or treatment that increases their risk of premature gonadal insufficiency may opt to undergo fertility preservation. Those who are postpubertal can often cryopreserve gametes, sperm, or eggs to expand their biologic family using assisted reproductive technologies. Ovarian tissue cryopreservation (OTC) and testicular tissue cryopreservation may be an option for individuals who are unable to use standard fertility preservation techniques. The development of OTC was critical for many patients, including prepubertal children with ovaries that do not yet produce eggs, adolescents who make few good-quality eggs, and adult women with ovaries who cannot undergo ovarian stimulation. The only option to restore fertility and hormone production after OTC is through ovarian tissue transplantation (OTT). Ovarian tissue cryopreservation and OTT have been successful for some patients. Although OTC is no longer considered experimental by the American Society for Reproductive Medicine, the process is far from standardized. Significant research needs to be done, especially at the point of OTT, to improve the success and longevity of ovarian tissue function. This article lists the main steps from surgical procurement of the ovarian tissue to transplantation and restoration of function. Our pediatric hospital program has had to decide which options in procurement, processing, cryopreservation, and warming will be used in our clinical laboratory. The options and limitations within the research and analyses are briefly discussed. Literature focusing on techniques to improve OTT effectiveness and longevity was reviewed. Ovarian tissue transplantation studies that performed xenograft experiments after pretreatment of the tissue graft by a ligand or drug, treatment of the host, or encapsulation of the ovarian tissue were identified. The intended effects of the treatments include increasing vascularization, reducing apoptosis, and directing activation or suppression of primordial follicles. Robust research in this area must continue with rigorous analyses to make strides in improving fertility preservation and restoration options for patients.
Collapse
Affiliation(s)
- Lauren M Gadek
- Department of Surgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Chicago, Illinois; Division of Endocrinology, Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Claire Joswiak
- Department of Surgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Chicago, Illinois; Division of Endocrinology, Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Monica M Laronda
- Department of Surgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Stanley Manne Children's Research Institute, Chicago, Illinois; Division of Endocrinology, Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois.
| |
Collapse
|
16
|
Zupo R, Castellana F, Nawrot TS, Lampignano L, Bortone I, Murgia F, Campobasso G, Gruszecka Kosowska A, Giannico OV, Sardone R. Air pollutants and ovarian reserve: a systematic review of the evidence. Front Public Health 2024; 12:1425876. [PMID: 39376999 PMCID: PMC11457886 DOI: 10.3389/fpubh.2024.1425876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024] Open
Abstract
Background Growing evidence indicates an association between ambient air pollution and decreased human reproductive potential. This study aims to systematically review the association between air pollutants and female ovarian reserve. Methods The literature was searched in six electronic databases through June 2024. Screening the 136 articles retrieved for inclusion criteria resulted in the selection of 15 human observational studies that evaluated the effect of environmental pollutants on ovarian reserve markers. The study protocol was registered on the International Prospective Register of Systematic Reviews (PROSPERO, registration code: CRD42023474218). Results The study design of the selected studies was found to be cross-sectional (2 of 15), retrospective cohort (10 of 15), prospective cohort (2 of 15), and case-control (1 of 15). The study population was distributed as follows: Asians (53%, eight studies), Americans (33%, five studies), and Europeans (14%, two studies). The main findings showed a higher body of evidence for the environmental pollutants PM2.5, PM10, and NO2, while a low body of evidence for PM1, O3, SO2, and a very low body of evidence for benzene, formaldehyde, and benzo(a)pyrene, yet consistently showing significant inverse association data. The overall methodological quality of the selected studies was rated moderated across the 14 domains of the National Institutes of Health (NIH) toolkit. Conclusion The data suggest that increased exposure to air pollutants seems to be associated with reduced ovarian reserve, with the most substantial evidence for pollutants such as PM2.5, PM10, and NO2. However, more evidence is needed to draw conclusions about causality.
Collapse
Affiliation(s)
- Roberta Zupo
- Department of Interdisciplinary Medicine (DIM), University of Bari Aldo Moro, Piazza Giulio Cesare, Bari, Italy
| | - Fabio Castellana
- Department of Interdisciplinary Medicine (DIM), University of Bari Aldo Moro, Piazza Giulio Cesare, Bari, Italy
| | - Tim S. Nawrot
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Department of Public Health and Primary Care, Environment and Health Unit, Leuven University, Leuven, Belgium
| | | | - Ilaria Bortone
- Department of Translational Biomedicine and Neuroscience "DiBraiN", University of Bari "Aldo Moro", Bari, Italy
| | - Ferdinando Murgia
- Department of Obstetrics and Gynecology, "Miulli" General Hospital, Bari, Italy
| | | | - Agnieskza Gruszecka Kosowska
- Department of Environmental Protection, Faculty of Geology, Geophysics and Environmental Protection, AGH University of Krakow, Al. Mickiewicza, Krakow, Poland
| | | | - Rodolfo Sardone
- Unit of Statistics and Epidemiology, Local Health Authority of Taranto, Taranto, Italy
- Department of Eye and Vision Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
17
|
Gowkielewicz M, Lipka A, Zdanowski W, Waśniewski T, Majewska M, Carlberg C. Anti-Müllerian hormone: biology and role in endocrinology and cancers. Front Endocrinol (Lausanne) 2024; 15:1468364. [PMID: 39351532 PMCID: PMC11439669 DOI: 10.3389/fendo.2024.1468364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Anti-Müllerian hormone (AMH) is a peptide belonging to the transforming growth factor beta superfamily and acts exclusively through its receptor type 2 (AMHR2). From the 8th week of pregnancy, AMH is produced by Sertoli cells, and from the 23rd week of gestation, it is produced by granulosa cells of the ovary. AMH plays a critical role in regulating gonadotropin secretion, ovarian tissue responsiveness to pituitary hormones, and the pathogenesis of polycystic ovarian syndrome. It inhibits the transition from primordial to primary follicles and is considered the best marker of ovarian reserve. Therefore, measuring AMH concentration of the hormone is valuable in managing assisted reproductive technologies. AMH was initially discovered through its role in the degeneration of Müllerian ducts in male fetuses. However, due to its ability to inhibit the cell cycle and induce apoptosis, it has also garnered interest in oncology. For example, antibodies targeting AMHR2 are being investigated for their potential in diagnosing and treating various cancers. Additionally, AMH is present in motor neurons and functions as a protective and growth factor. Consequently, it is involved in learning and memory processes and may support the treatment of Alzheimer's disease. This review aims to provide a comprehensive overview of the biology of AMH and its role in both endocrinology and oncology.
Collapse
Affiliation(s)
- Marek Gowkielewicz
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Aleksandra Lipka
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Wojciech Zdanowski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Tomasz Waśniewski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marta Majewska
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
18
|
Andersen CY, Donnez J, Ernst E, Gook D, Pellicer A, Von Wolff M, Suzuki N, Roux C, Dolmans MM. Ovarian tissue cryopreservation in breast cancer patients: glass half empty or glass half full? Reprod Biomed Online 2024; 50:104442. [PMID: 39561658 DOI: 10.1016/j.rbmo.2024.104442] [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: 07/02/2024] [Revised: 08/16/2024] [Accepted: 08/31/2024] [Indexed: 11/21/2024]
Abstract
This is a commentary to a paper recently published in RBMOnline by Macklon and De Vos, in which they argue for a discontinuation of ovarian tissue freezing for fertility preservation in women with breast cancer. Instead, they suggest the use of oocyte vitrification following ovarian stimulation as the preferred method of fertility preservation. This commentary presents nine separate arguments that should be considered in the context of ovarian tissue freezing and fertility preservation in girls and women. Collectively, the authors support ovarian tissue freezing going forward and suggest continuing this procedure for fertility preservation in women with breast cancer. Ovarian tissue freezing represents several advantages for patients and provides them with more options following treatment compared with oocyte vitrification.
Collapse
Affiliation(s)
- Claus Y Andersen
- The Fertility Clinic, Copenhagen University Hospital Herlev, Herlev, Denmark; Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jacques Donnez
- Université Catholique de Louvain and Society for Research into Infertility (SRI), Brussels, Belgium
| | - Erik Ernst
- University Clinic for Fertility, Regional Hospital Horsens, Horsens, Denmark
| | - Debbie Gook
- Reproductive Services, The Royal Women's Hospital, and Department of Obstetrics and Gynecology, University of Melbourne, Parkville, Victoria, Australia
| | - Antonio Pellicer
- IVIRMA-Global, IVI Rome, Valencia University School of Medicine, Valencia, Spain
| | - Michael Von Wolff
- Division of Gynecological Endocrinology and Reproductive Medicine, University Women's Hospital, University of Bern, Inselspital Bern, Switzerland
| | - Nao Suzuki
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Christophe Roux
- Assisted Reproductive Technologies Department, INSERM CIC 1431, University Hospital, Besançon, France
| | - Marie-Madeleine Dolmans
- Gynaecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
19
|
Li Y, Zhang J, Han W, Liu B, Zhai M, Li N, Wang Z, Zhao J. Multifunctional Laser-Induced Graphene-Based Microfluidic Chip for High-Performance Oocyte Cryopreservation with Low Concentration of Cryoprotectants. Adv Healthc Mater 2024; 13:e2400981. [PMID: 38885030 DOI: 10.1002/adhm.202400981] [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: 03/16/2024] [Revised: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Oocyte cryopreservation is essential in the field of assisted reproduction, but due to the large size and poor environmental tolerance of oocytes, cell freezing technology needs further improvement. Here, a Y-shaped microfluidic chip based on 3D graphene is ingeniously devised by combining laser-induced graphene (LIG) technology and fiber etching technology. The prepared LIG/PDMS microfluidic chip can effectively suppress ice crystal size and delay ice crystal freezing time by adjusting surface hydrophobicity. In addition, LIG endows the microfluidic chip with an outstanding photothermal effect, which allows to sharply increase its surface temperature from 25 to 71.8 °C with 10 s of low-power 808 nm laser irradiation (0.4 W cm-2). Notably, the LIG/PDMS microfluidic chip not only replaces the traditional cryopreservation carriers, but also effectively reduces the dosage of cryoprotectants (CPAs) needed in mouse oocyte cryopreservation. Even when the concentration of CPAs is cut in half (final concentration of 7.5% ethylene glycol (EG) and 7.5% dimethyl sulfoxide (DMSO)), the survival rate of oocytes is still as high as 92.4%, significantly higher than the control group's 85.8%. Therefore, this work provides a novel design strategy to construct multifunctional microfluidic chips for high-performance oocytes cryopreservation.
Collapse
Affiliation(s)
- Yifang Li
- School of Mechatronics and Vehicle Engineering, Chongqing Jiaotong University, Chongqing, 400074, China
- Institute of Solid-State Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
- Key Laboratory of Photovoltaic and Energy Conservation Materials, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jixiang Zhang
- School of Mechatronics and Vehicle Engineering, Chongqing Jiaotong University, Chongqing, 400074, China
- Institute of Solid-State Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
- Key Laboratory of Photovoltaic and Energy Conservation Materials, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Wei Han
- School of Mechatronics and Vehicle Engineering, Chongqing Jiaotong University, Chongqing, 400074, China
- Institute of Solid-State Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
- Key Laboratory of Photovoltaic and Energy Conservation Materials, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Bianhua Liu
- Institute of Solid-State Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
- Key Laboratory of Photovoltaic and Energy Conservation Materials, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Mengjie Zhai
- School of Mechatronics and Vehicle Engineering, Chongqing Jiaotong University, Chongqing, 400074, China
- Institute of Solid-State Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
- Key Laboratory of Photovoltaic and Energy Conservation Materials, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Nian Li
- Institute of Solid-State Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
- Key Laboratory of Photovoltaic and Energy Conservation Materials, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Zhenyang Wang
- Institute of Solid-State Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
- Key Laboratory of Photovoltaic and Energy Conservation Materials, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Jun Zhao
- Institute of Solid-State Physics, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
- Key Laboratory of Photovoltaic and Energy Conservation Materials, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| |
Collapse
|
20
|
Raimondo D, Raffone A, Neola D, Genovese F, Travaglino A, Aguzzi A, De Gobbi V, Virgilio A, Di Santo S, Vicenti R, Magnani V, Guida M, Pippucci T, Seracchioli R. Molecular Factors Predicting Ovarian Chemotoxicity in Fertile Women: A Systematic Review. Cancers (Basel) 2024; 16:2793. [PMID: 39199566 PMCID: PMC11352339 DOI: 10.3390/cancers16162793] [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: 06/27/2024] [Revised: 07/28/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
Background: Recent advances in cancer diagnosis and treatment have significantly improved survival rates among women of reproductive age facing cancer. However, the potential iatrogenic loss of fertility caused by chemotherapeutic agents underscores the need to understand and predict chemotherapy-induced ovarian damage. This study addresses this gap by systematically reviewing the literature to investigate genetic markers associated with chemotherapy-induced ovarian failure (CIOF). Objective: The primary objective is to identify genetic markers linked to CIOF, contributing to a comprehensive understanding of the factors influencing fertility preservation in female cancer survivors. Methods: A systematic review was conducted using PubMed, EMBASE, Web of Science, Scopus, and OVID electronic databases from inception through December 2023. Studies were included if they featured genomic assessments of genes or polymorphisms related to CIOF in women with histologically confirmed tumors. Exclusion criteria comprised in vitro and animal studies, reviews, and pilot studies. The resulting four human-based studies were scrutinized for insights into genetic influences on CIOF. Results: Of the 5179 articles initially identified, four studies met the inclusion criteria, focusing on alkylating agents, particularly cyclophosphamide, and anthracyclines. Su et al. explored CYP3A41B variants, revealing modified associations with CIOF based on age. Charo et al. investigated GSTA1 and CYP2C19 polymorphisms, emphasizing the need to consider age and tamoxifen therapy in assessing associations. Oktay et al. delved into the impact of BRCA mutations on anti-Müllerian hormone (AMH) levels post-chemotherapy, supported by in vitro assays. Van der Perk et al. focused on childhood cancer survivors and revealed significant associations of CYP3A43 and CYP2B6*2 SNPs with AMH levels. Conclusions: This systematic review analyzes evidence regarding genetic markers influencing CIOF, emphasizing the complex interplay of age, specific genetic variants, and chemotherapy regimens. The findings underscore the need for a personalized approach in assessing CIOF risk, integrating genetic markers with traditional ovarian reserve testing. The implications of this study extend to potential advancements in fertility preservation strategies, offering clinicians a comprehensive baseline assessment for tailored interventions based on each patient's unique genetic profile. Further research is essential to validate these findings and establish a robust framework for integrating genetic markers into clinical practice.
Collapse
Affiliation(s)
- Diego Raimondo
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
| | - Antonio Raffone
- Department of Woman, Child, and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Daniele Neola
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (F.G.); (M.G.)
| | - Federica Genovese
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (F.G.); (M.G.)
| | - Antonio Travaglino
- Unit of Pathology, Department of Medicine and Technological Innovation, University of Insubria, 21100 Varese, Italy;
| | - Alberto Aguzzi
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Valeria De Gobbi
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Agnese Virgilio
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Sara Di Santo
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Rossella Vicenti
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Valentina Magnani
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Maurizio Guida
- Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (F.G.); (M.G.)
| | - Tommaso Pippucci
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Renato Seracchioli
- Division of Gynecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy (A.A.); (V.D.G.); (S.D.S.); (R.V.); (V.M.); (R.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| |
Collapse
|
21
|
Di Nisio V, Daponte N, Messini C, Anifandis G, Antonouli S. Oncofertility and Fertility Preservation for Women with Gynecological Malignancies: Where Do We Stand Today? Biomolecules 2024; 14:943. [PMID: 39199331 PMCID: PMC11353009 DOI: 10.3390/biom14080943] [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: 05/19/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 09/01/2024] Open
Abstract
Oncofertility is a growing medical and research field that includes two main areas: oncology and reproductive medicine. Nowadays, the percentage of patients surviving cancer has exponentially increased, leading to the need for intervention for fertility preservation in both men and women. Specifically, gynecological malignancies in women pose an additional layer of complexity due to the reproductive organs being affected. In the present review, we report fertility preservation options with a cancer- and stage-specific focus. We explore the drawbacks and the necessity for planning fertility preservation applications during emergency statuses (i.e., the COVID-19 pandemic) and comment on the importance of repro-counseling for multifaceted patients during their oncological and reproductive journey.
Collapse
Affiliation(s)
- Valentina Di Nisio
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, Huddinge, 14186 Stockholm, Sweden;
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, 14186 Stockholm, Sweden
| | - Nikoletta Daponte
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| | - Christina Messini
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| | - George Anifandis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| | - Sevastiani Antonouli
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece; (N.D.); (C.M.); (G.A.)
| |
Collapse
|
22
|
Li D, Zhao YJ, Wang Q, Chu MW, Xie JK, Zhang CL. Fertility preservation in hematological cancer patients. Clin Transl Oncol 2024; 26:1836-1843. [PMID: 38575837 DOI: 10.1007/s12094-024-03419-2] [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] [Received: 09/20/2023] [Accepted: 02/23/2024] [Indexed: 04/06/2024]
Abstract
Among adolescents and young adults, hematological malignancies are the most common malignancies. Although the survival rate of hematological malignancies in young patients has been dramatically improved, due to the continuous improvement and development of tumor diagnosis and treatment options, cytotoxic therapies can significantly reduce a patient's reproductive capacity and cause irreversible infertility. The most two established solutions are embryo cryopreservation and oocyte cryopreservation which can be considered in single female. Sperm or testicular tissue cryopreservation in adult male are feasible approaches that must be considered before gonadotoxic therapy. A comprehensive consultation with reproductive specialists when once diagnosed is a significantly issue which would help those survivors who want to have children. In this article, we review germ cell toxicity, which happens during the treatment of hematological malignancies, and aims to propose safety, efficacy fertility preservation methods in younger patients with hematological malignancies.
Collapse
Affiliation(s)
- Dan Li
- Reproductive Medicine Center, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yi-Jun Zhao
- Reproductive Medicine Center, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Qian Wang
- Reproductive Medicine Center, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Man-Wei Chu
- Reproductive Medicine Center, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Juan-Ke Xie
- Reproductive Medicine Center, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Cui-Lian Zhang
- Reproductive Medicine Center, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
| |
Collapse
|
23
|
Ligon JA, Cupit-Link MC, Yu C, Levine J, Foley T, Rotz S, Sharma A, Gomez-Lobo V, Shah NN. Pediatric Cancer Immunotherapy and Potential for Impact on Fertility: A Need for Evidence-Based Guidance. Transplant Cell Ther 2024; 30:737-749. [PMID: 38866240 DOI: 10.1016/j.jtct.2024.06.006] [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: 02/27/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024]
Abstract
The use of immunotherapies for the treatment of cancer in children, adolescents, and young adults has become common. As the use of immunotherapy has expanded, including in earlier lines of therapy, it has become evident that several aspects of how these immunotherapies impact longer-term outcomes among survivors are understudied. Traditional cancer therapies like alkylating and platin agents carry the greatest risk of infertility, but little is known about the impact of novel immunotherapies on fertility. This topic is of great interest to patients, patient advocates, and clinicians. In this article, we review immunotherapeutic agents used to treat childhood and young adult cancers and discuss potential mechanisms by which they may impact fertility based on the known interplay between the immune system and reproductive organs. We highlight the relative paucity of high-quality literature examining these late effects. We discuss interventions to optimize fertility preservation (FP) for our patients. Conducting longitudinal, collaborative, and prospective research on the fertility outcomes of pediatric and young adult patients with cancer who receive immunotherapy is critical to learn how to effectively counsel our patients on long-term fertility outcomes and indications for FP procedures. Collection of patient-level data will be necessary to draft evidence-based guidelines on which providers can make therapy recommendations.
Collapse
Affiliation(s)
- John A Ligon
- Department of Pediatrics, Division of Hematology Oncology, University of Florida, Gainesville, Florida; University of Florida Health Cancer Center, Gainesville, Florida.
| | | | - Christine Yu
- Department of Pediatric Medicine, St Jude Children's Research Hospital, Memphis, Tennessee
| | - Jennifer Levine
- Center for Cancer and Blood Disorders, Division of Oncology, Children's National Hospital, Washington District of Columbia
| | - Toni Foley
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Seth Rotz
- Division of Pediatric Hematology and Oncology and Blood and Marrow Transplantation, Cleveland Clinic, Cleveland, Ohio
| | - Akshay Sharma
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Veronica Gomez-Lobo
- Pediatric and Adolescent Gynecology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland; Pediatric and Adolescent Gynecology, Children's National Hospital, Washington, District of Columbia
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
24
|
McElhinney KL, Rowell EE, Laronda MM. Encapsulation of Bovine Primordial Follicles in Rigid Alginate Does Not Affect Growth Dynamics. Bioengineering (Basel) 2024; 11:734. [PMID: 39061816 PMCID: PMC11273846 DOI: 10.3390/bioengineering11070734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/02/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
The only fertility preservation and subsequent restoration option for many patients facing gonadotoxic treatments is ovarian tissue cryopreservation and transplantation. While this process is successful for some, there is significant room for improvement to extend the life of the transplant and to make it safe for patients that may have metastatic disease within their ovarian tissue. We need a deeper understanding of how the physical properties of the ovarian microenvironment may affect folliculogenesis to engineer an environment that supports isolated follicles and maintains primordial follicle quiescence. Bovine ovaries were used here as a monovulatory model of folliculogenesis to examine the effects of primordial follicle activation and growth under different physical conditions. We found that there were no differences in activation, growth or survival when primordial follicles were cultured in isolation or in situ (remaining in the tissue) under two significantly differently rigid alginate gels. To determine if the extra rigid environment did not affect activation in isolated follicles due to an immediate activation event, we used 5-ethynyl-2'-deoxyuridine (EdU) to track follicle activation during the isolation process. We identified EdU incorporation in granulosa cells after primordial follicles were isolated from the surrounding extracellular matrix (ECM). These findings support that isolation of primordial follicles from the ECM is an activating event and that the differentially rigid environments assessed here had no effect on follicle growth. Further work is needed to suppress activation in primordial follicles to maintain the ovarian reserve and extend the life of an ovarian tissue transplant.
Collapse
Affiliation(s)
- Kathryn L. McElhinney
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (K.L.M.)
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Erin E. Rowell
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (K.L.M.)
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Monica M. Laronda
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (K.L.M.)
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
25
|
Yazdani A, Amir J, Christianson MS. Ovarian tissue cryopreservation for fertility preservation: with innovation must come the development of guidelines and best practices. Fertil Steril 2024; 122:83-84. [PMID: 38723933 DOI: 10.1016/j.fertnstert.2024.04.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 06/21/2024]
Affiliation(s)
- Ariella Yazdani
- Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Jawaria Amir
- Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| | - Mindy S Christianson
- Reproductive Endocrinology and Infertility, Obstetrics and Gynecology and Women's Health Institute, Cleveland Clinic Foundation, Cleveland, Ohio
| |
Collapse
|
26
|
Sänger N, John J, Einenkel R, Schallmoser A. First report on successful delivery after retransplantation of vitrified, rapid warmed ovarian tissue in Europe. Reprod Biomed Online 2024; 49:103940. [PMID: 38744030 DOI: 10.1016/j.rbmo.2024.103940] [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: 12/21/2023] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 05/16/2024]
Abstract
RESEARCH QUESTION Cryopreservation of ovarian tissue is one feasible option to preserve female fertility prior to cancer treatment. The slow freezing protocol represents the current standard approach, while vitrification has been suggested as a promising alternative. This paper reports the follow-up and first successful delivery after retransplantation of vitrified, rapid warmed ovarian tissue in Europe. DESIGN After the patient received a diagnosis of breast cancer, ovarian tissue was removed laparoscopically and sent via overnight transportation to University Hospital Bonn for vitrification on site. The patient was treated with chemotherapy, leading to ovarian failure. After 2 years, retransplantation of the vitrified, rapid warmed tissue was conducted on site. RESULTS Two months after grafting, the patient reported regular menstrual cycles. After 1 further month a clinical pregnancy occurred, which ended in a spontaneous abortion at the 8th week of pregnancy. Six months after grafting, another naturally conceived pregnancy was determined, resulting in the birth of a healthy boy 14 months after retransplantation of the ovarian tissue. CONCLUSIONS Complementing the successful deliveries reported by the groups of Suzuki (Japan) and Silber (USA) regarding vitrified tissue, the current results confirm the high potential of this cryopreservation method in a clinical routine setting as an alternative approach to the widespread slow freezing method.
Collapse
Affiliation(s)
- Nicole Sänger
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany.
| | - Julia John
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany
| | - Rebekka Einenkel
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany
| | - Andreas Schallmoser
- Department of Gynecological Endocrinology and Reproductive Medicine, University Hospital of Bonn, Germany
| |
Collapse
|
27
|
Varghese J, Khan MHA, Sukumar S. First Childbirth after Fertility Preservation and Gender Reassigning Surgery from Indian Transgender Community. J Hum Reprod Sci 2024; 17:216-220. [PMID: 39544677 PMCID: PMC11559351 DOI: 10.4103/jhrs.jhrs_49_24] [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: 03/26/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 11/17/2024] Open
Abstract
Fertility preservation (FP) is critical for those members of the transgender community who desire biological offspring in the future. As gender-affirming hormone therapy and gender-affirming surgeries (GASs) negatively affect future fertility potential, awareness amongst care providers and the transgender population regarding FP and family-building options is the need of the hour. As the first case of childbirth after transgender FP and GAS in India, this will be a beacon of hope for transgender family-building options in India.
Collapse
Affiliation(s)
- Jisha Varghese
- Department of Reproductive Medicine, Renai Centre for Fertility and Reproductive Health, Renai Medicity, Kochi, Kerala, India
| | - M H Azif Khan
- Department of Reproductive Medicine, Renai Centre for Fertility and Reproductive Health, Renai Medicity, Kochi, Kerala, India
| | - Suja Sukumar
- Department of Endocrinology, Renai Medicity, Kochi, Kerala, India
| |
Collapse
|
28
|
Raimondo D, Raspollini A, Bertoldo L, Ferla S, Vicenti R, Magnani V, Raffone A, Seracchioli R. Laparoscopic medulla-sparing ovarian tissue biopsy for cryopreservation: step-by-step surgical technique. Fertil Steril 2024; 122:184-186. [PMID: 38492928 DOI: 10.1016/j.fertnstert.2024.03.011] [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: 11/16/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
OBJECTIVE To describe a laparoscopic technique for ovarian tissue biopsy (OTB) for fertility preservation. In the last years, the demand for fertility preservation has grown because of the increasing survival rates among patients with cancer and the rising awareness of the importance of quality of life after gonadotoxic therapy. Among fertility-sparing approaches, ovarian tissue cryopreservation is a valid strategy to preserve ovarian endocrine and reproductive function in prepubertal and postpubertal women who will undergo gonadotoxic cancer treatments. Currently, there is no universal consensus regarding ovarian tissue retrieval technique for fertility preservation. DESIGN Step-by-step description of the surgical technique with narrated video footage. SETTING Academic tertiary hospital. PATIENT(S) Patients with a high risk of premature ovarian insufficiency, usually due to gonadotoxic treatments, who undergo OTB for fertility preservation were included in the study. In this video, we present the clinical case of a 28-year-old patient affected by Hodgkin lymphoma who underwent laparoscopy for OTB before chemotherapy. INTERVENTION(S) After exposing the chosen ovary, an incision at the tubal pole of the ovary is made with scissors. Through section and dissection, a large cortical biopsy of the ovary is performed without removing and avoiding any damage to the medulla. At the end of the procedure, hemostasis was achieved with selective coagulation using bipolar coagulation. MAIN OUTCOME MEASURE(S) Step by step educational video. RESULT(S) The post-operative course was uneventful and the patient was discharge 24 hours after surgery. CONCLUSION(S) Standardization of a step-by-step laparoscopic technique can provide an effective method to optimize ovarian tissue removal while minimizing tissue injury. Medulla-sparing ovarian biopsy allows retrieval of only the cortical part of the ovary, maximizing the number of primordial follicles obtained without damaging the vascular supply of the ovary contained within the medulla. Primordial follicles are resistant to cryoinjury owing to their relatively inactive metabolism, and they are usually found at approximately 0.8 mm below the surface of the cortex. This technique could also reduce the back-table processing time of the ovarian tissue before cryopreservation. One disadvantage could be the difficulty of the technique compared to an oophorectomy because it requires a skilled surgeon that can easily find the cleavage plane between the medulla and the cortex, even in patients submitted to previous chemoradiotherapy or during gonadotropin-releasing hormone analogue therapy.
Collapse
Affiliation(s)
- Diego Raimondo
- Division of Gynaecology and Human Reproduction Physiopathology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Azienda Ospedaliero - Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Arianna Raspollini
- Division of Gynaecology and Human Reproduction Physiopathology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Azienda Ospedaliero - Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy.
| | - Linda Bertoldo
- Division of Gynaecology and Human Reproduction Physiopathology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Azienda Ospedaliero - Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Stefano Ferla
- Division of Gynaecology and Human Reproduction Physiopathology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Azienda Ospedaliero - Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Rossella Vicenti
- Division of Gynaecology and Human Reproduction Physiopathology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Azienda Ospedaliero - Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Valentina Magnani
- Division of Gynaecology and Human Reproduction Physiopathology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Azienda Ospedaliero - Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Antonio Raffone
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; Gynecology and Obstetrics Unit, Department of Neuroscience, Reproductive Sciences and Dentistry, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Renato Seracchioli
- Division of Gynaecology and Human Reproduction Physiopathology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Azienda Ospedaliero - Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| |
Collapse
|
29
|
Chevillon F, Rebotier M, Dhédin N, Bruno B, Cacciatore C, Charbonier A, Joseph L, Le Bourgeois A, Talouarn M, Magro L, Barraud Lange V. [Fertility preservation and hematopoietic stem cell transplantation (SFGM-TC)]. Bull Cancer 2024:S0007-4551(24)00185-1. [PMID: 38918137 DOI: 10.1016/j.bulcan.2024.04.009] [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: 02/12/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 06/27/2024]
Abstract
Conditioning regimen prior to hematopoietic stem cell transplantation have an impact on patient fertility through the use of gonadal irradiation and/or bifunctional alkylating agents. Their impact on fertility depends mainly on the dose used and, in women, on age at the time of treatment. All patients should benefit before treatment from a consultation informing them of the potential impact on fertility and of fertility preservation techniques. In the absence of contraindications, the major toxicity of myeloablative conditioning regimen justifies fertility preservation. There are few data concerning fertility after reduced-intensity conditioning. Despite lower theoretical gonadotoxicity, we also recommend fertility preservation, if possible before transplantation. The fertility preservation techniques used depend on the patient's age, pathology and conditioning. In the event of subsequent use of harvested gonadal tissue in the context of acute leukemia or aggressive lymphoma, it is advisable to assess the risk of reintroduction of tumor cells. Finally, it is recommended to assess gonadal function after transplant, especially after reduced conditioning. If there is persistent residual gonadal function, post-treatment fertility preservation should be discuss.
Collapse
Affiliation(s)
- Florian Chevillon
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France.
| | - Marine Rebotier
- Service oncogynécologie, centre Leon-Berard et IHOPe, 28, promenade Léa et Napoléon Bullukian, 69008 Lyon, France
| | - Nathalie Dhédin
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - Bénédicte Bruno
- Service hématologie pédiatrique, hôpital Jeanne-de-Flandre, avenue Eugène-Avinée, 59037 Lille, France
| | - Carlotta Cacciatore
- Service de médecine interne, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | | | - Laure Joseph
- Service hématologie, département de biothérapie, hôpital Necker-enfants malades, AP-HP, 149, rue de Sèvres, 75015 Paris, France
| | - Amandine Le Bourgeois
- Service d'hématologie, CHU de Nantes, 1, place Alexis-Ricordeau, 44000 Nantes, France
| | - Marie Talouarn
- Service d'hématologie, hôpital Saint-Antoine, AP-HP, 184, rue du faubourg Saint-Antoine, 75012 Paris, France
| | - Leonardo Magro
- Service d'hématologie, CHU de Lille, 2, avenue Oscar-Lambret, 59000 Lille, France
| | - Virginie Barraud Lange
- Service hématologie adolescents jeunes adultes, hôpital Saint-Louis, AP-HP, 1, avenue Claude-Vellefaux, 75010 Paris, France; Service biologie de la reproduction, hôpital Cochin Port Royal, AP-HP, 123, boulevard de Port Royal, 75014 Paris, France
| |
Collapse
|
30
|
Haering C, Coyne K, Daunov K, Anim S, Christianson MS, Flyckt R. Ovarian Tissue Cryopreservation for Fertility Preservation in Patients with Hemoglobin Disorders: A Comprehensive Review. J Clin Med 2024; 13:3631. [PMID: 38999197 PMCID: PMC11242023 DOI: 10.3390/jcm13133631] [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: 05/17/2024] [Revised: 06/11/2024] [Accepted: 06/17/2024] [Indexed: 07/14/2024] Open
Abstract
Hemoglobin diseases like sickle cell disease (SCD) and β-thalassemia (BT) present fertility challenges for affected patients. SCD and BT result from abnormal hemoglobin production or structure and pose numerous health concerns. Despite medical advancements improving the quality of life or even providing cures, SCD and BT pose unique fertility concerns for women. Young women with these disorders already contend with reduced ovarian reserve and a narrower fertile window, a situation that is compounded by the gonadotoxic effects of treatments like medications, transfusions, stem cell transplants, and gene therapy. While crucial for disease control, these interventions may lead to reproductive health issues, increasing infertility and early menopause risks. Ovarian tissue cryopreservation (OTC) offers potential for future motherhood to women with hemoglobin disorders facing infertility related to curative treatments. OTC involves surgically removing, preparing, and freezing ovarian tissue containing primordial follicles capable of producing mature oocytes, offering advantages over oocyte cryopreservation alone. However, the application of OTC for patients with hemoglobin disorders presents unique challenges, including special health risks, financial barriers, and access to care. This comprehensive literature review delves into the current state of ovarian tissue cryopreservation for fertility preservation in patients with hemoglobin disorders. Empowering patients with informed reproductive choices in the context of their hemoglobin disorders stands as the ultimate goal.
Collapse
Affiliation(s)
- Catherine Haering
- Division of Reproductive Endocrinology and Infertility, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| | - Kathryn Coyne
- Department of Reproductive Endocrinology and Infertility, University Hospitals Ahuja, Beachwood, OH 44122, USA
| | - Katherine Daunov
- Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Samuel Anim
- Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Mindy S. Christianson
- Department of Reproductive Endocrinology and Infertility, Cleveland Clinic Foundation Beachwood Family Health Center, Beachwood, OH 44122, USA
| | - Rebecca Flyckt
- Department of Reproductive Endocrinology and Infertility, University Hospitals Ahuja, Beachwood, OH 44122, USA
| |
Collapse
|
31
|
McElhinney KL, Kennedy T, Rowell EE, Laronda MM. A dozen years of ovarian tissue cryopreservation at a pediatric hospital: tracking program and patient metrics while adapting to increasing needs. F S Rep 2024; 5:197-204. [PMID: 38983744 PMCID: PMC11228781 DOI: 10.1016/j.xfre.2024.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 07/11/2024] Open
Abstract
Objective To review the program and patient metrics for ovarian tissue cryopreservation (OTC) within a comprehensive pediatric fertility preservation program in its first 12 years of development. Design Retrospective review. Setting A tertiary children's hospital in a large urban center between March 2011 and February 2023. Patients Pediatric patients who underwent OTC. Interventions Unilateral oophorectomy for OTC. Main Outcome Measures Patient demographics and clinical course information were collected for analysis. Results A total of 184 patients underwent OTC in the first 12 years. One hundred fifteen patients were prepubertal at the time of OTC, and 69 were postpubertal. In total, 128 patients (69.6%) received part of their planned therapy before OTC. Starting in 2018, 104 participants (92.0%) donated tissue to research, 99 participants (87.6%) donated blood, and 102 (90.2%) donated media to research. There was a decrease in the median age of patients who underwent OTC from 16.4-6.6 years and an overall increase in the proportion of patients per year that were prepubertal. Forty-eight (26.0%) patients who underwent OTC were outside referrals and traveled from as far as Seattle, Washington. Conclusion During the first 12 years of this program, oncofertility research increased, annual tissue cryopreservation cases increased, and the median age of those who underwent OTC decreased. The program was adapted to build a stand-alone gonadal tissue processing suite and specialized in prepubertal ovarian tissue processing. The program will continue to adapt to patient needs in the upcoming decades because restoration technologies advance through research supported by this and collaborating programs.
Collapse
Affiliation(s)
- Kathryn L McElhinney
- Department of Surgery, Division of Pediatric Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Fertility & Hormone Preservation & Restoration Program, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Tara Kennedy
- Department of Surgery, Division of Pediatric Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Fertility & Hormone Preservation & Restoration Program, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Erin E Rowell
- Department of Surgery, Division of Pediatric Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Fertility & Hormone Preservation & Restoration Program, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Monica M Laronda
- Department of Surgery, Division of Pediatric Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Fertility & Hormone Preservation & Restoration Program, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
32
|
Foster KL, Lee DJ, Witchel SF, Gordon CM. Ovarian Insufficiency and Fertility Preservation During and After Childhood Cancer Treatment. J Adolesc Young Adult Oncol 2024; 13:377-388. [PMID: 38265460 DOI: 10.1089/jayao.2023.0111] [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: 01/25/2024] Open
Abstract
Premature ovarian insufficiency (POI) is one of many potential long-term consequences of childhood cancer treatment in females. Causes of POI in this patient population can include chemotherapy, especially alkylating agents, and radiation therapy. Rarely, ovarian tumors lead to ovarian dysfunction. POI can manifest as delayed pubertal development, irregular menses or amenorrhea, and infertility. This diagnosis often negatively impacts emotional health due to the implications of impaired ovarian function after already enduring treatment for a primary malignancy. The emerging adult may be challenged by the impact on energy level, quality of life, and fertility potential. POI can also lead to low bone density and compromised skeletal strength. This review discusses the health consequences of POI in childhood cancer survivors (CCS). We also explore the role of fertility preservation for CCS, including ovarian tissue cryopreservation and other available options. Lastly, knowledge gaps are identified that will drive a future research agenda.
Collapse
Affiliation(s)
- Kayla L Foster
- Texas Children's Cancer and Hematology Center, Department of Pediatrics, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, USA
| | - Danielle J Lee
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Selma F Witchel
- Division of Pediatric Endocrinology, Department of Pediatrics, UPMC Children's Hospital, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Catherine M Gordon
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
33
|
Nishimura T, Takebe T. Synthetic human gonadal tissues for toxicology. Reprod Toxicol 2024; 126:108598. [PMID: 38657700 DOI: 10.1016/j.reprotox.2024.108598] [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: 11/17/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
The process of mammalian reproduction involves the development of fertile germ cells in the testis and ovary, supported by the surrounders. Fertilization leads to embryo development and ultimately the birth of offspring inheriting parental genome information. Any disruption in this process can result in disorders such as infertility and cancer. Chemical toxicity affecting the reproductive system and embryogenesis can impact birth rates, overall health, and fertility, highlighting the need for animal toxicity studies during drug development. However, the translation of animal data to human health remains challenging due to interspecies differences. In vitro culture systems offer a promising solution to bridge this gap, allowing the study of mammalian cells in an environment that mimics the physiology of the human body. Current advances on in vitro culture systems, such as organoids, enable the development of biomaterials that recapitulate the physiological state of reproductive organs. Application of these technologies to human gonadal cells would provide effective tools for drug screening and toxicity testing, and these models would be a powerful tool to study reproductive biology and pathology. This review focuses on the 2D/3D culture systems of human primary testicular and ovarian cells, highlighting the novel approaches for in vitro study of human reproductive toxicology, specifically in the context of testis and ovary.
Collapse
Affiliation(s)
- Toshiya Nishimura
- WPI Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka 565-0871, Japan.
| | - Takanori Takebe
- WPI Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Osaka 565-0871, Japan; Division of Stem Cell and Organoid Medicine, Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Division of Gastroenterology, Hepatology and Nutrition, Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Institute of Research, Tokyo Medical and Dental University (TMDU), Tokyo 113-8510, Japan; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA; Communication Design Center, Advanced Medical Research Center, Yokohama City University, Yokohama 236-0004, Japan.
| |
Collapse
|
34
|
McDowell HB, McElhinney KL, Tsui EL, Laronda MM. Generation of Tailored Extracellular Matrix Hydrogels for the Study of In Vitro Folliculogenesis in Response to Matrisome-Dependent Biochemical Cues. Bioengineering (Basel) 2024; 11:543. [PMID: 38927779 PMCID: PMC11200611 DOI: 10.3390/bioengineering11060543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/21/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
While ovarian tissue cryopreservation (OTC) is an important fertility preservation option, it has its limitations. Improving OTC and ovarian tissue transplantation (OTT) must include extending the function of reimplanted tissue by reducing the extensive activation of primordial follicles (PMFs) and eliminating the risk of reimplanting malignant cells. To develop a more effective OTT, we must understand the effects of the ovarian microenvironment on folliculogenesis. Here, we describe a method for producing decellularized extracellular matrix (dECM) hydrogels that reflect the protein composition of the ovary. These ovarian dECM hydrogels were engineered to assess the effects of ECM on in vitro follicle growth, and we developed a novel method for selectively removing proteins of interest from dECM hydrogels. Finally, we validated the depletion of these proteins and successfully cultured murine follicles encapsulated in the compartment-specific ovarian dECM hydrogels and these same hydrogels depleted of EMILIN1. These are the first, optically clear, tailored tissue-specific hydrogels that support follicle survival and growth comparable to the "gold standard" alginate hydrogels. Furthermore, depleted hydrogels can serve as a novel tool for many tissue types to evaluate the impact of specific ECM proteins on cellular and molecular behavior.
Collapse
Affiliation(s)
- Hannah B. McDowell
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.B.M.)
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kathryn L. McElhinney
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.B.M.)
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Elizabeth L. Tsui
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.B.M.)
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Monica M. Laronda
- Stanley Manne Children’s Research Institute, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA; (H.B.M.)
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
35
|
Barral Y, Borrás A, Carrillo P, Agustí I, Peralta S, Casals G, Fàbregues F, Guimerà M, Corral JM, Calafell JM, Redondo M, Glickman A, Rovira M, Adamo B, Ricart E, Trullàs DM. Current status of fertility preservation in a Spanish tertiary public hospital: multidisciplinary approach and experience in over 1500 patients. Clin Transl Oncol 2024; 26:1129-1138. [PMID: 37872422 DOI: 10.1007/s12094-023-03330-2] [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: 07/24/2023] [Accepted: 10/03/2023] [Indexed: 10/25/2023]
Abstract
PURPOSE Currently, 15% of gynaecological and 9% of haematological malignancies are diagnosed before the age of 40. The increased survival rates of cancer patients who are candidates for gonadotoxic treatments, the delay in childbearing to older ages, and the optimization of in vitro fertilisation techniques have all contributed to an increased interest in fertility preservation (FP) treatments. This study reviews the experience of the Fertility Preservation Programme (FPP) of a tertiary public hospital with a multidisciplinary approach. METHODS This retrospective study included all the available (FP) treatments, performed in patients of childbearing age between 2006 and 2022. RESULTS 1556 patients were referred to the FPP: 332 oocyte vitrification cycles, 115 ovarian cortex cryopreservation with 11 orthotopic autotransplantations, 175 gonadotropin-releasing hormone (GnRH) agonist treatments, 109 fertility-sparing treatments for gynaecological cancer, and 576 sperm cryopreservation were performed. Malignancy was the main indication for FP (the main indications being breast cancer in women and haematological malignancies in men), although non-oncological pathologies, such as endometriosis and autoimmune diseases, have increased in recent years. Currently, the most widely used FP technique is oocyte vitrification, the increase of which has been associated with a decrease in the use of cortex CP and GnRH agonists. CONCLUSIONS The increase in FP treatment reflects the implementation of reproductive counselling in oncology programmes. A multidisciplinary approach in a tertiary public hospital allows individualised FP treatment for each patient. In recent years, there has been a change in trend with the introduction of new indications for FP and a change in techniques due to their optimisation.
Collapse
Affiliation(s)
- Yasmina Barral
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Aina Borrás
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Pilar Carrillo
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Inés Agustí
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Sara Peralta
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Gemma Casals
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Francesc Fàbregues
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Marta Guimerà
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Juan Manuel Corral
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Josep Maria Calafell
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Miriam Redondo
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain
| | - Ariel Glickman
- Gynecologic Oncology Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Montserrat Rovira
- Department of Hematology, Clinical Institute of Hematological and Oncological Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Bárbara Adamo
- Department of Medical Oncology, Hospital Clínic, Clinical Institute of Hematological and Oncological Diseases, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Elena Ricart
- Gastroenterology Department, Clinical Institute of Digestive and Metabolic Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Dolors Manau Trullàs
- Assisted Reproduction Unit, Clinical Institute of Gynecology, Obstetrics and Neonatology, Hospital Clinic de Barcelona, C/Villarroel 170, 08036, Barcelona, Spain.
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
36
|
Charpentier P, Cavalieri M, Desmoulins I, Coutant C. [Live birth rates after breast cancer among women who desired a child]. Bull Cancer 2024; 111:463-472. [PMID: 38580527 DOI: 10.1016/j.bulcan.2024.02.005] [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: 10/28/2023] [Revised: 01/15/2024] [Accepted: 02/01/2024] [Indexed: 04/07/2024]
Abstract
INTRODUCTION In France, the breast cancer is the most common cancer among women under the age of 40. From 38 to 70% of women have not fulfilled their parental plans at the time of diagnosis. The gonadotoxicity of the treatments and the follicular physiological decline linked to age can become an obstacle to this project. METHODS Among the patients, 386 were treated for breast cancer at the Centre Georges-François-Leclerc in Dijon between January 2011 and December 2018 were identified. 192 patients aged under 39 met the inclusion criteria. We excluded metastatic cancers, cancer in situ and pregnant patients at diagnosis. A total of 124 patients agreed to participate in the study. The included patients filled out a self-questionnaire. Data were collected from the patient's electronic medical. The primary endpoint of this study was the live birth rate. RESULT Among women who desired a child after breast cancer, the overall rate of live births was 36.2 % (21/58). Most achieved pregnancies were spontaneous (90.5 %). No factor was significantly associated with the absence of obtaining birth. Fertility was preserved by oocyte cryopreservation in 13.8 % of patients (17/124). The median time to conception in patients who received chemotherapy was 8 months [1.0-60.0] vs 2 months [1.0-7.0] in women who did not receive chemotherapy. DISCUSSION The non-negligible proportion of live births following spontaneous pregnancy after breast cancer allows us to be reassuring for patients. However, the emergence of new chemotherapy protocols whose consequences on long-term gonadotoxicity are still not well known requires further studies and prompts the promotion of fertility preservation as a precautionary measure.
Collapse
Affiliation(s)
- Pauline Charpentier
- Service de gynécologie-obstétrique et médecine de la reproduction, CHU François-Mitterrand, Cote d'Or, Dijon, France.
| | - Mathilde Cavalieri
- Service de gynécologie-obstétrique et médecine de la reproduction, CHU François-Mitterrand, Cote d'Or, Dijon, France
| | - Isabelle Desmoulins
- Centre Francois-Leclerc, 1, rue du Professeur-Marion, Cote d'Or, 21000 France
| | - Charles Coutant
- Centre Francois-Leclerc, 1, rue du Professeur-Marion, Cote d'Or, 21000 France
| |
Collapse
|
37
|
Silber SJ, Goldsmith S, Castleman L, Hayashi K. In Vitro Maturation, In Vitro Oogenesis, and Ovarian Longevity. Reprod Sci 2024; 31:1234-1245. [PMID: 38160209 PMCID: PMC11090930 DOI: 10.1007/s43032-023-01427-1] [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: 06/05/2023] [Accepted: 12/11/2023] [Indexed: 01/03/2024]
Abstract
This paper will review a remarkable new approach to in vitro maturation "IVM" of oocytes from ovarian tissue, based on our results with in vitro oogenesis from somatic cells. As an aside benefit we also have derived a better understanding of ovarian longevity from ovary transplant. We have found that primordial follicle recruitment is triggered by tissue pressure gradients. Increased pressure holds the follicle in meiotic arrest and prevents recruitment. Therefore recruitment occurs first in the least dense inner tissue of the cortico-medullary junction. Many oocytes can be obtained from human ovarian tissue and mature to metaphase 2 in vitro with no need for ovarian stimulation. Ovarian stimulation may only be necessary for removing the oocyte from the ovary, but this can also be accomplished by simple dissection at the time of ovary tissue cryopreservation. By using surgical dissection of the removed ovary, rather than a needle stick, we can obtain many oocytes from very small follicles not visible with ultrasound. A clearer understanding of ovarian function has come from in vitro oogenesis experiments, and that explains why IVM has now become so simple and robust. Tissue pressure (and just a few "core genes" in the mouse) direct primordial follicle recruitment and development to mature oocyte, and therefore also control ovarian longevity. There are three distinct phases to oocyte development both in vitro and in vivo: in vitro differentiation "IVD" which is not gonadotropin sensitive (the longest phase), in vitro gonadotropin sensitivity "IVG" which is the phase of gonadotropin stimulation to prepare for meiotic competence, and IVM to metaphase II. On any given day 35% of GVs in ovarian tissue have already undergone "IVD" and "IVG" in vivo, and therefore are ready for IVM.
Collapse
Affiliation(s)
- Sherman J Silber
- Infertility Center of St. Louis at St. Luke's Hospital, St. Louis, MO, 63017, USA.
| | - Sierra Goldsmith
- Infertility Center of St. Louis at St. Luke's Hospital, St. Louis, MO, 63017, USA.
| | - Leilani Castleman
- Infertility Center of St. Louis at St. Luke's Hospital, St. Louis, MO, 63017, USA
| | - Katsuhiko Hayashi
- Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| |
Collapse
|
38
|
Li J, Fan H, Liu W, Zhang J, Xiao Y, Peng Y, Yang W, Liu W, He Y, Qin L, Ma X, Li J. Mesenchymal stem cells promote ovarian reconstruction in mice. Stem Cell Res Ther 2024; 15:115. [PMID: 38650029 PMCID: PMC11036642 DOI: 10.1186/s13287-024-03718-z] [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] [Accepted: 04/07/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Studies have shown that chemotherapy and radiotherapy can cause premature ovarian failure and loss of fertility in female cancer patients. Ovarian cortex cryopreservation is a good choice to preserve female fertility before cancer treatment. Following the remission of the disease, the thawed ovarian tissue can be transplanted back and restore fertility of the patient. However, there is a risk to reintroduce cancer cells in the body and leads to the recurrence of cancer. Given the low success rate of current in vitro culture techniques for obtaining mature oocytes from primordial follicles, an artificial ovary with primordial follicles may be a good way to solve this problem. METHODS In the study, we established an artificial ovary model based on the participation of mesenchymal stem cells (MSCs) to evaluate the effect of MSCs on follicular development and oocyte maturation. P2.5 mouse ovaries were digested into single cell suspensions and mixed with bone marrow derived mesenchymal stem cells (BM-MSCs) at a 1:1 ratio. The reconstituted ovarian model was then generated by using phytohemagglutinin. The phenotype and mechanism studies were explored by follicle counting, immunohistochemistry, immunofluorescence, in vitro maturation (IVM), in vitro fertilization (IVF), real-time quantitative polymerase chain reaction (RT-PCR), and Terminal-deoxynucleotidyl transferase mediated nick end labeling(TUNEL) assay. RESULTS Our study found that the addition of BM-MSCs to the reconstituted ovary can enhance the survival of oocytes and promote the growth and development of follicles. After transplanting the reconstituted ovaries under kidney capsules of the recipient mice, we observed normal folliculogenesis and oocyte maturation. Interestingly, we found that BM-MSCs did not contribute to the formation of follicles in ovarian aggregation, nor did they undergo proliferation during follicle growth. Instead, the cells were found to be located around growing follicles in the reconstituted ovary. When theca cells were labeled with CYP17a1, we found some overlapped staining with green fluorescent protein(GFP)-labeled BM-MSCs. The results suggest that BM-MSCs may participate in directing the differentiation of theca layer in the reconstituted ovary. CONCLUSIONS The presence of BM-MSCs in the artificial ovary was found to promote the survival of ovarian cells, as well as facilitate follicle formation and development. Since the cells didn't proliferate in the reconstituted ovary, this discovery suggests a potential new and safe method for the application of MSCs in clinical fertility preservation by enhancing the success rate of cryo-thawed ovarian tissues after transplantation.
Collapse
Affiliation(s)
- Jiazhao Li
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
- Scientific Research Department, Wannan Medical College, 241002, Wuhu, China
| | - Haonan Fan
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
| | - Wei Liu
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
| | - Jing Zhang
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
| | - Yue Xiao
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhejiang University School of Medicine, 310003, Hangzhou, China
| | - Yue Peng
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
- Pathology Department, Nanjing Kingmed Medical Laboratory Co.,Ltd., 210032, Nanjing, China
| | - Weijie Yang
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Zhejiang University School of Medicine, 310016, Hangzhou, China
| | - Wenwen Liu
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
- Women's Hospital of Nanjing Medical University (Nanjing Maternity and Child Health Care Hospital), 21003, Nanjing, China
| | - Yuanlin He
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China
| | - Lianju Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center of Clinical Reproductive Medicine, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.
| | - Xiang Ma
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center of Clinical Reproductive Medicine, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, 210029, Nanjing, China.
- Prenatal Diagnosis Department, First Affiliated Hospital, Nanjing Medical University, 210029, Nanjing, China.
| | - Jing Li
- State Key Laboratory of Reproductive Medicine and Offspring health, Nanjing Medical University, 210029, Nanjing, China.
| |
Collapse
|
39
|
Tsui EL, McDowell HB, Laronda MM. Restoring Ovarian Fertility and Hormone Function: Recent Advancements, Ongoing Efforts and Future Applications. J Endocr Soc 2024; 8:bvae073. [PMID: 38698870 PMCID: PMC11065362 DOI: 10.1210/jendso/bvae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Indexed: 05/05/2024] Open
Abstract
The last 20 years have seen substantial improvements in fertility and hormone preservation and restoration technologies for a growing number of cancer survivors. However, further advancements are required to fill the gaps for those who cannot use current technologies or to improve the efficacy and longevity of current fertility and hormone restoration technologies. Ovarian tissue cryopreservation (OTC) followed by ovarian tissue transplantation (OTT) offers those unable to undergo ovarian stimulation for egg retrieval and cryopreservation an option that restores both fertility and hormone function. However, those with metastatic disease in their ovaries are unable to transplant this tissue. Therefore, new technologies to produce good-quality eggs and restore long-term cyclic ovarian function are being investigated and developed to expand options for a variety of patients. This mini-review describes current and near future technologies including in vitro maturation, in vitro follicle growth and maturation, bioprosthetic ovaries, and stem cell applications in fertility restoration research by their proximity to clinical application.
Collapse
Affiliation(s)
- Elizabeth L Tsui
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Hannah B McDowell
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| | - Monica M Laronda
- Department of Pediatrics, Division of Endocrinology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
40
|
Kocsuta V, Shah S, Lawson AK, Pavone ME. Do Fertility Preservation Outcomes in Patients Diagnosed with Lymphoma Differ Based on Cancer Stage? J Adolesc Young Adult Oncol 2024; 13:300-306. [PMID: 36809174 DOI: 10.1089/jayao.2022.0069] [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: 02/23/2023] Open
Abstract
Purpose: This study aims to investigate whether oocyte stimulation outcomes in fertility preservation (FP) vary in patients with different stages of lymphoma. Methods: This is a retrospective cohort study conducted at Northwestern Memorial Hospital (NMH). Between 2006 and 2017, 89 patients were identified with a diagnosis of lymphoma who contacted the FP navigator at NMH. Anti-müllerian hormone (AMH) levels and FP ovarian stimulation outcomes were collected for analysis. The data were analyzed using chi-squared and analysis of variance tests. A regression analysis was also done to adjust for potential confounding variables. Results: Of the 89 patients who contacted the FP navigator, there were 12 patients (13.5%) with stage 1 lymphoma, 43 patients (48.3%) with stage 2, 13 patients (14.6%) with stage 3, 13 patients (14.6%) with stage 4, and 8 patients (9.0%) where staging information was not available. Forty-five of the patients proceeded with ovarian stimulation before initiating cancer treatment. Patients who underwent ovarian stimulation had a mean AMH level of 2.62 and median peak estradiol levels of 1772.0 pg/mL. Median oocytes retrieved was 16.77, mature oocytes were 11.00 and median oocytes frozen after completing FP was 8.00. These measures were also stratified by stage of lymphoma. Conclusion: We found no significant difference in number of retrieved, mature or vitrified oocytes between different cancer stages. There was also no difference in AMH levels in the different cancer stage groups. This suggests that even in higher stages of lymphoma, many patients respond to ovarian stimulation techniques and have a successful stimulation cycle.
Collapse
Affiliation(s)
- Victoria Kocsuta
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Shriya Shah
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Angela K Lawson
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mary Ellen Pavone
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
41
|
Boumerdassi Y, Labrosse J, Hammami F, Dahoun M, Bouyer J, O'Neill L, Sarandi S, Peigné M, Cedrin I, Grynberg M, Sifer C. Impact of oxygen tension during in vitro maturation: a sibling-oocyte prospective double-blinded study. Fertil Steril 2024; 121:615-621. [PMID: 38103883 DOI: 10.1016/j.fertnstert.2023.12.014] [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: 03/31/2022] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
OBJECTIVE To determine whether oxygen (O2) tension (20% vs. 5%) has an impact on oocyte maturation rates and morphology during in vitro maturation (IVM). DESIGN A prospective, observational, monocentric, sibling-oocyte study. SETTING University Hospital. PATIENTS A total of 143 patients who underwent IVM for fertility preservation purposes from November 2016 to April 2021 were analyzed. Patients were included when ≥2 cumulus-oocyte complexes (COCs) were retrieved. The cohort of COCs obtained for each patient was randomly split into two groups: group 20% O2 and group 5% O2. INTERVENTION Cumulus-oocyte complexes were incubated for 48 hours either under 5% O2 or 20% O2. After 24 and 48 hours of culture, every oocyte was assessed for maturity and morphology, to estimate oocyte quality. Morphology was evaluated considering six parameters (shape, size, ooplasm, perivitelline space, zona pellucida, and polar body characteristics), giving a total oocyte score ranging from -6 to +6. MAIN OUTCOME MEASURES Maturation rates and total oocyte scores were compared using paired-sample analysis between group 20% O2 and group 5% O2. RESULTS Patient median age was 31.4 [28.1-35.2] years-old. The mean serum antimüllerian hormone levels and antral follicle count were 3.2 ± 2.3 ng/mL and 27.2 ± 16.0 follicles, respectively. A mean of 10.7 COCs per cycle were retrieved, leading to 6.1 ± 2.4 metaphase II oocytes vitrified (total maturation rate = 57.3%; 991 metaphase II oocytes/1,728 COCs). A total of 864 COCs were included in each group. Oocyte maturation rates were not different between the two groups (group 20% O2: 56.82% vs. group 5% O2: 57.87%, respectively). Regarding oocyte morphology, the mean total oocyte score was significantly higher in group 5% O2 compared with group 20% O2 (3.44 ± 1.26 vs. 3.16 ± 1.32, P=.014). CONCLUSION As culture under low O2 tension (5% O2) improves oocyte morphology IVM, our results suggest that culture under hypoxia should be standardized. Additional studies are warranted to assess the impact of O2 tension on oocyte maturation and the benefit of IVM under low O2 tension for embryo culture after utilization of frozen material.
Collapse
Affiliation(s)
- Yasmine Boumerdassi
- Department of Reproductive Biology, Hôpital Jean Verdier, Bondy, France; Université Sorbonne Paris Nord, Villetaneuse, France
| | - Julie Labrosse
- Department of Reproductive Medicine and Fertility Preservation, Hôpital Jean Verdier, Bondy, France
| | - Fatma Hammami
- Department of Reproductive Biology, Hôpital Jean Verdier, Bondy, France
| | - Mehdi Dahoun
- Department of Reproductive Biology, Hôpital Jean Verdier, Bondy, France
| | - Jean Bouyer
- Centre for Research in Epidemiology and Population Health, Université Paris Saclay, France
| | - Louis O'Neill
- Department of Reproductive Biology, Hôpital Jean Verdier, Bondy, France
| | - Solmaz Sarandi
- Department of Reproductive Biology, Hôpital Jean Verdier, Bondy, France
| | - Maeliss Peigné
- Department of Reproductive Medicine and Fertility Preservation, Hôpital Jean Verdier, Bondy, France
| | - Isabelle Cedrin
- Department of Reproductive Medicine and Fertility Preservation, Hôpital Jean Verdier, Bondy, France
| | - Michael Grynberg
- Department of Reproductive Medicine and Fertility Preservation, Hôpital Jean Verdier, Bondy, France; Department of Reproductive Medicine and Fertility Preservation, Hôpital Antoine Béclère, Clamart, France; Université Paris Saclay, Le Kremlin Bicêtre, France; Université Paris-Diderot, Paris, France
| | - Christophe Sifer
- Department of Reproductive Biology, Hôpital Jean Verdier, Bondy, France; Équipe RHuMA, UMR-BREED, UFR Simone Veil Santé, Montigny le Bretonneux, France.
| |
Collapse
|
42
|
Ying H, Shi L, Zhang S. Research progress on mechanism of follicle injury after ovarian tissue transplantation and protective strategies. Zhejiang Da Xue Xue Bao Yi Xue Ban 2024; 53:321-330. [PMID: 38562041 PMCID: PMC11348700 DOI: 10.3724/zdxbyxb-2023-0566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Ovarian tissue cryopreservation and transplantation is the only way to preserve fertility for female cancer patients in prepubertal ages and those who cannot delay radiotherapy or chemotherapy. However, the success rate of cryopreservation and transplantation of ovarian tissue is still low at present due to the risk of ischemia and hypoxia of the grafted tissues. Abnormal activation of primordial follicles and ischemia-reperfusion injury after blood supply recovery also cause massive loss of follicles in grafted ovarian tissues. Various studies have explored the use of different drugs to reduce the damage of follicles during freezing and transplantation as well as to extend the duration of endocrine and reproductive function in patients with ovarian transplantation. For example, melatonin, N-acetylcysteine, erythropoietin or other antioxidants have been used to reduce oxidative stress; mesenchymal stem cells derived from different tissues, basic fibroblast growth factor, vascular endothelial growth factor, angiopoietin 2 and gonadotropin have been used to promote revascularization; anti-Müllerian hormone and rapamycin have been used to reduce abnormal activation of primordial follicles. This article reviews the research progress on the main mechanisms of follicle loss after ovarian tissue transplantation, including hypoxia, ischemia-reperfusion injury and associated cell death, and abnormal activation of follicles. The methods for reducing follicle loss in grafted ovarian tissues are further explored to provide a reference for improving the efficiency of ovarian tissue cryopreservation and transplantation.
Collapse
Affiliation(s)
- Hanqi Ying
- Reproductive Medicine Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
| | - Libing Shi
- Reproductive Medicine Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
| | - Songying Zhang
- Reproductive Medicine Center, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou 310016, China.
| |
Collapse
|
43
|
Ruan X, Xu C, Huang H, Xu B, Du J, Cheng J, Jin F, Gu M, Kong W, Yin C, Wu Y, Tian Q, Cao Y, Wu R, Xu L, Jin J, Li Y, Dai Y, Ju R, Ma F, Wang G, Wei W, Huang X, Qin M, Lin Y, Sun Y, Liu R, Zhang W, Li X, Zou L, Hao M, Ye X, Wang F, Wang Y, Hu Z, Huang Y, Zhu T, Yang C, Wang J, Yang X, Ni R, Wang L, Luo G, Min A, Zhang S, Li P, Cheng L, Li L, Jin Q, Shi D, Li Y, Ren F, Cheng Y, Niu J, Tian Y, Mueck AO. Practice guideline on ovarian tissue cryopreservation and transplantation in the prevention and treatment of iatrogenic premature ovarian insufficiency. Maturitas 2024; 182:107922. [PMID: 38325136 DOI: 10.1016/j.maturitas.2024.107922] [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: 09/28/2023] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 02/09/2024]
Abstract
Premature ovarian insufficiency (POI) refers to the decline of ovarian function before the age of 40. POI causes a reduction in or loss of female fertility, accompanied by different degrees of menopausal symptoms, which increases the risk of chronic diseases related to early menopause and seriously affects patients' quality of life and health. It is conservatively estimated that at least one million prepubertal girls and women of reproductive age in China are at risk of iatrogenic POI caused by radiotherapy and chemotherapy every year. With the development of medical technology and the breakthrough of scientific and technological advances, preventing and treating iatrogenic POI have become possible. International and national guidelines consider cryopreserved ovarian tissue transplantation to be the most promising method of preserving the ovarian function and fertility of prepubertal girls and women of reproductive age who cannot delay radiotherapy and chemotherapy. In order to guide the clinical application of ovarian tissue cryopreservation and transplantation technology in China, the Guideline Working Group finally included 14 scientific questions and 18 recommendations through a questionnaire survey, field investigation, and consultation of a large number of Chinese and English literature databases in order to provide a reference for colleagues in clinical practice.
Collapse
Affiliation(s)
- Xiangyan Ruan
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China; Department for Women's Health, University Women's Hospital and Research Center for Women's Health, University of Tuebingen, Tuebingen, Germany.
| | - Che Xu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China; Fuxing Hospital, Capital Medical University, Beijing, China
| | - Hefeng Huang
- Ministry of Education Key Laboratory of Reproductive Genetics, Shool of Medicine, Zhejiang University, Hangzhou, China
| | - Binghe Xu
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Juan Du
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Jiaojiao Cheng
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fengyu Jin
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Muqing Gu
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Weimin Kong
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Chenghong Yin
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yurui Wu
- Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Qinjie Tian
- Peking Union Medical College Hospital, Peking Union Medical College/Chinese Academy of Medical Sciences, Beijing, China
| | - Yunxia Cao
- The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Ruifang Wu
- Peking University Shenzhen Hospital, Shenzhen, China
| | - Liangzhi Xu
- West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jing Jin
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yanglu Li
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yinmei Dai
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Rui Ju
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Fei Ma
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gang Wang
- Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Wei Wei
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | | | - Maoquan Qin
- National Center for Children's Health, Hematology Center, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yuan Lin
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fujian, China
| | - Yuan Sun
- Beijing Jingdu Children's Hospital, Beijing, China
| | - Rong Liu
- Children's Hospital, Capital Institute of Pediatrics, Beijing, China
| | - Wei Zhang
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaodong Li
- The First Hospital of Hebei Medical University, Hebei, China
| | - Lin Zou
- Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Min Hao
- Second Hospital of Shanxi Medical University, Shanxi, China
| | - Xiyang Ye
- Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Fuling Wang
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yue Wang
- Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhuoying Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanhong Huang
- Xi'an International Medical Center Hospital, Xi'an, China
| | - Tianyuan Zhu
- Gansu Provincial Maternal and Child-care Hospital/Gansu Province Central Hospital, Lanzhou, China
| | - Caihong Yang
- The General Hospital of Ningxia Medical University, Ningxia, China
| | - Jinping Wang
- Zibo Maternal And Child Health Hospital, Zibo, China
| | - Xiaomin Yang
- Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Rong Ni
- The Central Hospital of Enshi Tu Jia and Miao Autonomous Prefecture, Enshi, China
| | - Liqun Wang
- Jiangxi Maternal and Child Health Hospital, Jiangxi, China
| | - Guangxia Luo
- The First People's Hospital of Huaihua (Hunan University of Medicine General Hospital), Huaihua, China
| | - Aiping Min
- People's Hospital of Leshan City, Leshan, China
| | - Siyou Zhang
- The First People's Hospital of Foshan, Foshan, China
| | - Peiling Li
- The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linghui Cheng
- The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Lianfang Li
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Huairou Maternal and Child Health Care Hospital, Huairou, China
| | - Quanfang Jin
- Shanghai Baoshan Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Dongmei Shi
- Maternal and Child Health Hospital of Yinchuan, Yinchuan, China
| | - Yan Li
- Luoyang Anhe Hospital, Luoyang, China
| | | | | | - Jumin Niu
- Shenyang Women's and Children's Hospital, Shenyang, China
| | - Ying Tian
- XiangXi Ninger Obstetrics and Gynecology Hospital, Xiangxi, China
| | - Alfred O Mueck
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China; Department for Women's Health, University Women's Hospital and Research Center for Women's Health, University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
44
|
Wang L, Liu HR, Wang T, Feng ML, Jiang ZY, Yang Q, Sun D, Song CR, Zhang XJ, Liang CG. C-phycocyanin improves the developmental potential of cryopreserved human oocytes by minimizing ROS production and cell apoptosis. PLoS One 2024; 19:e0300538. [PMID: 38558076 PMCID: PMC10984518 DOI: 10.1371/journal.pone.0300538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
PURPOSE The cryopreservation process damages oocytes and impairs development potential. As a potent antioxidant, C-phycocyanin (PC) regulates reproductive performance. However, its beneficial effects on vitrified human oocytes remain unknown. METHODS In this study, human GV-stage oocytes obtained from controlled ovarian hyperstimulation (COH) cycles were randomly allocated to three groups: fresh oocyte without freezing (F group), vitrification in medium supplemented with PC (P group), and vitrification in medium without PC as control group (C group). After warming, viable oocytes underwent in vitro maturation. RESULTS Our results showed that 3 μg/mL PC treatment increased the oocyte maturation rate after cryopreservation. We also found that PC treatment maintains the regular morphological features of oocytes. After PC treatment, confocal fluorescence staining showed a significant increase in the mitochondrial membrane potential of the vitrified oocytes, along with a notable decrease in intracellular reactive oxygen species and the early apoptosis rate. Finally, after in vitro maturation and parthenogenetic activation, vitrified oocytes had a higher potential for cleavage and blastocyst formation after PC treatment. CONCLUSION Our results suggest that PC improves the developmental potential of cryopreserved human GV-stage oocytes by attenuating oxidative stress and early apoptosis and increasing the mitochondrial membrane potential.
Collapse
Affiliation(s)
- Lu Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, People’s Republic of China
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Inner Mongolia Baogang Hospital, Baotou, Inner Mongolia, People’s Republic of China
| | - Hao-Ran Liu
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Teng Wang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Meng-Lei Feng
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Zhao-Yu Jiang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Qi Yang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Dui Sun
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Chun-Ru Song
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, People’s Republic of China
| | - Xiu-Juan Zhang
- Inner Mongolia Academy of Science and Technology, Hohhot, Inner Mongolia, People’s Republic of China
| | - Cheng-Guang Liang
- State Key Laboratory of Reproductive Regulation & Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia, People’s Republic of China
| |
Collapse
|
45
|
Vitale F, Dolmans MM. Comprehensive Review of In Vitro Human Follicle Development for Fertility Restoration: Recent Achievements, Current Challenges, and Future Optimization Strategies. J Clin Med 2024; 13:1791. [PMID: 38542015 PMCID: PMC10970962 DOI: 10.3390/jcm13061791] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 11/11/2024] Open
Abstract
Ovarian tissue cryopreservation (OTC) and subsequent transplantation (OTT) is a fertility preservation technique widely offered to prepubertal girls and young fertile women who need to undergo oncological treatment but are at a high risk of infertility. However, OTT is not considered safe in patients with certain diseases like leukemia, Burkitt's lymphoma, and ovarian cancer because of the associated risk of malignant cell reintroduction. In vitro follicle development has therefore emerged as a promising means of obtaining mature metaphase II (MII) oocytes from the primordial follicle (PMF) pool contained within cryopreserved ovarian tissue, without the need for transplantation. Despite its significant potential, this novel approach remains highly challenging, as it requires replication of the intricate process of intraovarian folliculogenesis. Recent advances in multi-step in vitro culture (IVC) systems, tailored to the specific needs of each follicle stage, have demonstrated the feasibility of generating mature oocytes (MII) from early-stage human follicles. While significant progress has been made, there is still room for improvement in terms of efficiency and productivity, and a long way to go before this IVC approach can be implemented in a clinical setting. This comprehensive review outlines the most significant improvements in recent years, current limitations, and future optimization strategies.
Collapse
Affiliation(s)
- Francisco Vitale
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, 1200 Brussels, Belgium;
| | - Marie-Madeleine Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, 1200 Brussels, Belgium;
- Gynecology Department, Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| |
Collapse
|
46
|
Fabbri R, Vicenti R, Magnani V, Paradisi R, De Meis L, Raimondo D, Arena A, Venturoli S, Raffone A, Raspollini A, Seracchioli R. Ovarian tissue transplantation: 10 years of experience at the Bologna University. Front Endocrinol (Lausanne) 2024; 15:1332673. [PMID: 38516411 PMCID: PMC10954843 DOI: 10.3389/fendo.2024.1332673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 01/30/2024] [Indexed: 03/23/2024] Open
Abstract
Objective The efficiency of ovarian tissue transplantation (OTT) was established in terms of ovarian function recovery (95% of cases), number of live births (over 200 worldwide to date) and induction of puberty. Unfortunately, the lack of international registries and the fact that many centers have not yet reported their outcomes, lead to poor knowledge of the exact fertility data. The aim of the study is to describe our experience with OTT to restore ovarian function and fertility. Methods This study was designed as a single-center, observational, retrospective, cohort study that includes women who underwent OTT between December 2012 and June 2023 at our center. After approval by the oncologist/hematologist, a small fragment of ovarian tissue was thawed and analyzed to detect the presence of micrometastases before OTT. Thawed ovarian tissue was grafted laparoscopically at multiple sites, including the remaining ovary and pelvic side wall (orthotopic transplantation) and/or abdominal wall (heterotopic transplantation). After OTT, ovarian function was monitored by hormonal assay, ultrasound and color Doppler at approximately 4-week intervals. Results Between December 2012 and June 2023, 30 women performed OTT. Prior to OTT, immunohistochemical and molecular analyses revealed no micrometastases in all thawed ovarian tissue samples. In our series of 30 women, 20 of women were on premature ovarian insufficiency (POI), and the remaining ten cases still had oligomenorrhea and difficulty getting pregnant. Among the women with POI before OTT and at least 6 months follow-up, recovery of endocrine function was observed in all but one woman who underwent orthotopic transplantation (13 of 14 cases), in one out of two women who underwent both orthotopic and heterotopic transplantation (1 of 2 cases) and in all women who underwent heterotopic transplantation (4 of 4 cases). Women who underwent OTT to enhance fertility had no alterations in menstrual cycle and hormonal levels. In total, ten pregnancies were obtained in 25 women, resulting in four live births, two ongoing pregnancies and four spontaneous abortions. Conclusion Our data can help patients and physicians in their discussions and decisions about the need and possibilities of preserving fertility.
Collapse
Affiliation(s)
- Raffaella Fabbri
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Rossella Vicenti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Valentina Magnani
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Roberto Paradisi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Lucia De Meis
- Division of Gynaecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Diego Raimondo
- Division of Gynaecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alessandro Arena
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Stefano Venturoli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Antonio Raffone
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Arianna Raspollini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Renato Seracchioli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Division of Gynaecology and Human Reproduction Physiopathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
47
|
Wakimoto K, Wakimoto Y, Matsuda I, Yoshihara S, Ukita Y, Fukui A, Hirota S, Shibahara H. A case of non-Hodgkin lymphoma of the upper gingiva with minimal residual disease detected in cryopreserved ovarian tissue: A case report. J Obstet Gynaecol Res 2024; 50:526-529. [PMID: 38062974 DOI: 10.1111/jog.15845] [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: 06/28/2023] [Accepted: 11/24/2023] [Indexed: 03/04/2024]
Abstract
Recently, more than 200 live births following ovarian tissue cryopreservation (OTC) and transplantation in cancer survivors have been reported worldwide. However, cancer survivors with minimal residual disease (MRD) in cryopreserved ovarian tissue are at the risk of relapse through the graft. Here, we report a rare case of a 19-year-old female patient with non-Hodgkin lymphoma who had MRD in the ovary harvested for OTC. The patient was diagnosed with aggressive B-cell lymphoma after gingival biopsy. The 18F-fluoro-2-deoxy-D-glucose positron emission tomography scan performed before OTC showed no viable lesions in either ovary. However, on histological evaluation, we detected infiltration of lymphoma cells in the ovary. Informed consent about MRD is required even if there is no evidence of MRD in the ovary before OTC. Patients whose cryopreserved ovaries have MRD may require the development of alternative assisted reproductive technologies such as in vitro growth or artificial ovary.
Collapse
Affiliation(s)
- Ken Wakimoto
- Department of Obstetrics and Gynecology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Yu Wakimoto
- Department of Obstetrics and Gynecology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Ikuo Matsuda
- Department of Surgical Pathology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Satoshi Yoshihara
- Department of Respiratory Medicine and Hematology, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Yuji Ukita
- Department of Obstetrics and Gynecology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
- Reproduction Ukita Clinic, Otsu, Shiga, Japan
| | - Atushi Fukui
- Department of Obstetrics and Gynecology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Seiichi Hirota
- Department of Surgical Pathology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| | - Hiroaki Shibahara
- Department of Obstetrics and Gynecology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo, Japan
| |
Collapse
|
48
|
León-Félix CM, Maranhão AQ, Amorim CA, Lucci CM. Optimizing Decellularization of Bovine Ovarian Tissue: Toward a Transplantable Artificial Ovary Scaffold with Minimized Residual Toxicity and Preserved Extracellular Matrix Morphology. Cells Tissues Organs 2024; 213:413-423. [PMID: 38359805 DOI: 10.1159/000537838] [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: 10/09/2023] [Accepted: 02/09/2024] [Indexed: 02/17/2024] Open
Abstract
INTRODUCTION The decellularized extracellular matrix (dECM) from ovarian tissue could be the best scaffold for the development of a transplantable artificial ovary. Typically, dECM from ovarian tissue has been obtained using sodium dodecyl sulfate (SDS), at a concentration of 1% for 24 h. However, SDS can leave residues in the tissue, which may be toxic to the seeded cells. This study aimed to obtain dECM from bovine ovarian tissue using SDS and NaOH at a minimum concentration in the shortest incubation time. METHODS The respective SDS and NaOH concentrations investigated were 1% and 0.2 m; 0.5% and 0.1 m; 0.1% and 0.02 m; and 0.05% and 0.01 m, with 24-, 12-, and 6-h incubation periods. After the incubation time, the tissue was washed in 50 mL of distilled water for 6 h. RESULTS Histological analysis confirmed decellularization and showed the conservation of collagen fibers in all samples following treatment. Furthermore, the lowest SDS and NaOH concentrations that showed no DNA remaining during electrophoresis analysis were 0.1% and 0.02 m when incubated for 24 and 12 h. DNA quantification resulted in <0.2 ng DNA/mg ovarian tissue using these protocols. Additionally, the coculture of dECM (obtained by 0.1% SDS and 0.02 m NaOH for 12 h) with ovarian cells showed that there was no toxic effect for the cells for up to 72 h. CONCLUSION The protocol involving 0.1% SDS and 0.02 m NaOH for 12-h incubation decellularizes bovine ovarian tissue, generating a dECM that preserves the native ECM morphology and is nontoxic to ovarian cells.
Collapse
Affiliation(s)
- Cecibel M León-Félix
- Department of Physiology, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil,
| | - Andrea Q Maranhão
- Department of Cellular Biology, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| | - Christiani A Amorim
- Department of Gynecology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Carolina M Lucci
- Department of Physiology, Institute of Biological Sciences, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
49
|
Rezaei-Tazangi F, Kooshesh L, Tayyebiazar A, Taghizabet N, Tavakoli A, Hassanpour A, Aliakbari F, Kharazinejad E, Sharifi AM. Effects of kisspeptin on the maturation of human ovarian primordial follicles in vitro. ZYGOTE 2024; 32:66-70. [PMID: 38099429 DOI: 10.1017/s0967199423000527] [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: 01/16/2024]
Abstract
At this time, with advances in medical science, many cancers and chronic diseases are treatable, but one of their side effects is infertility. Some women also want to delay pregnancy for personal reasons. There has been some evidence that kisspeptin activates broad signals by binding to its receptor, suggesting that the role of kisspeptin in direct control of ovarian function includes follicle growth and steroid production. In this study, the effect of kisspeptin on improving the quality and results for human ovarian follicles was investigated. A section of ovary was removed laparoscopically from women between 20 and 35 years of age (n = 12). Pieces were divided randomly into two groups, control and treatment (with 1 μM kisspeptin). Real-time PCR was performed for GDF9, BMP15 and mTOR gene expression assessments. Western blotting was carried out to measure AKT and FOXO3a protein expression. Data were analyzed using one-way analysis of variance (ANOVA) and Tukey's test; means were considered significantly different at a P-value < 0.05. During treatment with the kisspeptin group, maturity genes are expressed. Therefore, kisspeptin is an effective substance to improve the quality of the human ovarian medium as it increases the maturity of follicles.
Collapse
Affiliation(s)
- Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Leila Kooshesh
- Department of Genetics, Fars Academic Center for Education, Culture and Research, ACECR, Shiraz, Iran
| | - Ali Tayyebiazar
- Clinical Research Development Unit of Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, I.R. Iran
| | - Neda Taghizabet
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anahita Tavakoli
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| | - Ashraf Hassanpour
- Anatomical Department, Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Aliakbari
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ebrahim Kharazinejad
- Department of Anatomy, Faculty of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| | - Ali-Mohammad Sharifi
- Clinical Research Development Center, Shahid Modarres Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Pang L, Jiang M, Sui X, Dou Y, Yu W, Huxley R, Saldiva P, Hu J, Schikowski T, Krafft T, Gao P, Zhao Y, Zhao H, Zhao Q, Chen ZJ. Association of PM 2.5 mass and its components with ovarian reserve in a northern peninsular province, China: The critical exposure period and components. JOURNAL OF HAZARDOUS MATERIALS 2024; 462:132735. [PMID: 37832436 DOI: 10.1016/j.jhazmat.2023.132735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 09/21/2023] [Accepted: 10/05/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND A possible role of PM2.5 components on ovarian reserve has not been adequately unexplored. OBJECTIVE To evaluate the association between PM2.5 components and women' ovarian reserve over critical exposure periods in northern China, where the level of air pollution is among the nation's highest. METHODS We included 15,102 women with serum anti-Müllerian hormone (AMH) measurements from the Center for Reproductive Medicine of Shandong University during 2015-2019. Concentrations of PM2.5 and its five major components (0.1° × 0.1°), including sulfate, nitrate, ammonium, organic matter, and black carbon, were assigned to each residential address. Multivariable linear mixed effect models combined with constituent-residual models were performed to estimate the effect sizes of essential components over six short- to long-term exposure periods. RESULTS The strength of association was stronger during the process from primary to small antral follicle compared with other longer windows. For every interquartile range increase in PM2.5 mass was associated with - 8.7% (95%CI: -12.3%, -4.9%) change in AMH and the effect size was greatest for sulfate. Women with the lower level of attained education and those living inland were more susceptible compared with other population subgroups. CONCLUSION Exposure to specific components of air pollution during critical exposure windows is associated with a decline in ovarian reserve. These data add to the growing body of evidence that environmental factors have adverse effects on reproductive health, particularly for vulnerable population subgroups.
Collapse
Affiliation(s)
- Lihong Pang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong 250012, China; Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
| | - Mingdong Jiang
- Dezhou Center for Disease Control and Prevention, Dezhou, Shandong 253000, China
| | - Xinlei Sui
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong 250012, China; Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
| | - Yunde Dou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong 250012, China; Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
| | - Wenhao Yu
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Rachel Huxley
- Faculty of Health, Deakin University, Melbourne 3000, Australia
| | - Paulo Saldiva
- Department of Pathology, Faculty of Medicine, University of São Paulo, São Paulo 01000, Brazil
| | - Jingmei Hu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong 250012, China; Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
| | - Tamara Schikowski
- Department of Epidemiology, IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf 40225, Germany
| | - Thomas Krafft
- Department of Health, Ethics & Society, Care and Public Health Research Institute CAPHRI, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht 6211, the Netherlands
| | - Panjun Gao
- Department of Health, Ethics & Society, Care and Public Health Research Institute CAPHRI, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht 6211, the Netherlands
| | - Yueran Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong 250012, China; Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China.
| | - Han Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong 250012, China; Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China.
| | - Qi Zhao
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Faculty of Health, Deakin University, Melbourne 3000, Australia.
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Shandong University, Jinan, Shandong 250012, China; Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250012, China; Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong 250012, China; Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250012, China; Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China; Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China; Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai 200135, China; Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, Shandong 250012, China
| |
Collapse
|