1
|
Cacciottola L, Camboni A, Dolmans MM. Immune system regulation of physiological and pathological aspects of the ovarian follicle pool throughout the female reproductive lifespan. Hum Reprod 2024:deae254. [PMID: 39607771 DOI: 10.1093/humrep/deae254] [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: 05/08/2024] [Revised: 10/14/2024] [Indexed: 11/30/2024] Open
Abstract
The immune system plays a major role in ovarian physiology by regulating the ovarian follicle pool through complex signaling of different growth factors, cytokines, and chemokines. These may promote follicle activation and further growth but could also trigger follicle atresia and clearance of aging or damaged cells within the ovarian cortex. Moreover, extraglandular steroidogenesis potentially occurring in different immune cells like macrophages and natural killer cells might be another way of modulating follicle growth. Ovarian macrophages have recently been found to contain two different populations, namely resident macrophages and monocyte-derived cells, with potentially different roles. The immune system also plays a role in the development of pathological conditions, including premature ovarian insufficiency (POI). Indeed, autoimmune activation against various ovarian antigen targets results in lymphocytic oophoritis mainly targeting early growing follicles, but later leading to complete follicle pool depletion. Immune-mediated ovarian damage may also be caused by viral infection or be the consequence of iatrogenic damage. Certain novel cancer immunotherapies like checkpoint inhibitors have recently been shown to induce ovarian reserve damage in a murine model. Studies are needed to corroborate these findings and further investigate the potential of newly developed immunotherapies to treat POI. Technological advances such as single-cell analyses of less represented cell populations like immune cells inside the ovary are now contributing to valuable new information, which will hopefully lead to the development of new therapeutic strategies for women with fertility issues.
Collapse
Affiliation(s)
- L Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - A Camboni
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Anatomopathology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - M M Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Gynecology Department, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
2
|
Zhang X, Ge J, Wang Y, Chen M, Guo X, Zhu S, Wang H, Wang Q. Integrative Omics Reveals the Metabolic Patterns During Oocyte Growth. Mol Cell Proteomics 2024; 23:100862. [PMID: 39414232 PMCID: PMC11585809 DOI: 10.1016/j.mcpro.2024.100862] [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: 04/29/2024] [Revised: 08/01/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024] Open
Abstract
Well-controlled metabolism is associated with high-quality oocytes and optimal development of a healthy embryo. However, the metabolic framework that controls mammalian oocyte growth remains unknown. In the present study, we comprehensively depict the temporal metabolic dynamics of mouse oocytes during in vivo growth through the integrated analysis of metabolomics and proteomics. Many novel metabolic features are discovered during this process. Of note, glycolysis is enhanced, and oxidative phosphorylation capacity is reduced in the growing oocytes, presenting a Warburg-like metabolic program. For nucleotide biosynthesis, the salvage pathway is markedly activated during oocyte growth, whereas the de novo pathway is evidently suppressed. Fatty acid synthesis and channeling into phosphoinositides are specifically elevated in oocytes accompanying primordial follicle activation; nevertheless, fatty acid oxidation is reduced in these oocytes simultaneously. Our data establish the metabolic landscape during in vivo oocyte growth and serve as a broad resource for probing mammalian oocyte metabolism.
Collapse
Affiliation(s)
- Xiang Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Juan Ge
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Yue Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Minjian Chen
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xuejiang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China
| | - Shuai Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China.
| | - Hui Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, China.
| | - Qiang Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Changzhou Maternity and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Nanjing, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
3
|
Longobardi S, Klinger FG, Zheng W, Campitiello MR, D’Hooghe T, La Marca A. Gonadotropin Activity during Early Folliculogenesis and Implications for Polycystic Ovarian Syndrome and Premature Ovarian Insufficiency: A Narrative Review. Int J Mol Sci 2024; 25:7520. [PMID: 39062762 PMCID: PMC11277126 DOI: 10.3390/ijms25147520] [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: 06/07/2024] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Female fertility depends on the ovarian reserve of follicles, which is determined at birth. Primordial follicle development and oocyte maturation are regulated by multiple factors and pathways and classified into gonadotropin-independent and gonadotropin-dependent phases, according to the response to gonadotropins. Folliculogenesis has always been considered to be gonadotropin-dependent only from the antral stage, but evidence from the literature highlights the role of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) during early folliculogenesis with a potential role in the progression of the pool of primordial follicles. Hormonal and molecular pathway alterations during the very earliest stages of folliculogenesis may be the root cause of anovulation in polycystic ovary syndrome (PCOS) and in PCOS-like phenotypes related to antiepileptic treatment. Excessive induction of primordial follicle activation can also lead to premature ovarian insufficiency (POI), a condition characterized by menopause in women before 40 years of age. Future treatments aiming to suppress initial recruitment or prevent the growth of resting follicles could help in prolonging female fertility, especially in women with PCOS or POI. This review will briefly introduce the impact of gonadotropins on early folliculogenesis. We will discuss the influence of LH on ovarian reserve and its potential role in PCOS and POI infertility.
Collapse
Affiliation(s)
| | - Francesca Gioia Klinger
- Department of Histology and Embryology, University of Health Sciences, Saint Camillus International, Via di Sant’Alessandro 8, 00131 Rome, Italy
| | | | - Maria Rosaria Campitiello
- Department of Obstetrics and Gynecology and Physiopathology of Human Reproduction, ASL Salerno, 84124 Salerno, Italy
| | - Thomas D’Hooghe
- Merck KGaA, 64293 Darmstadt, Germany (T.D.)
- Department of Development and Regeneration, Biomedical Sciences Group, KU Leuven (University of Leuven), 3000 Leuven, Belgium
| | - Antonio La Marca
- Department of Maternal-Child and Adult Medical and Surgical Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| |
Collapse
|
4
|
Giaccari C, Antonouli S, Anifandis G, Cecconi S, Di Nisio V. An Update on Physiopathological Roles of Akt in the ReprodAKTive Mammalian Ovary. Life (Basel) 2024; 14:722. [PMID: 38929705 PMCID: PMC11204812 DOI: 10.3390/life14060722] [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: 04/20/2024] [Revised: 05/19/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
The phosphoinositide 3-kinase (PI3K)/Akt pathway is a key signaling cascade responsible for the regulation of cell survival, proliferation, and metabolism in the ovarian microenvironment. The optimal finetuning of this pathway is essential for physiological processes concerning oogenesis, folliculogenesis, oocyte maturation, and embryo development. The dysregulation of PI3K/Akt can impair molecular and structural mechanisms that will lead to follicle atresia, or the inability of embryos to reach later stages of development. Due to its pivotal role in the control of cell proliferation, apoptosis, and survival mechanisms, the dysregulation of this molecular pathway can trigger the onset of pathological conditions. Among these, we will focus on diseases that can harm female fertility, such as polycystic ovary syndrome and premature ovarian failure, or women's general health, such as ovarian cancer. In this review, we report the functions of the PI3K/Akt pathway in both its physiological and pathological roles, and we address the existing application of inhibitors and activators for the balancing of the molecular cascade in ovarian pathological environments.
Collapse
Affiliation(s)
- Carlo Giaccari
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Caserta, Italy;
| | - Sevastiani Antonouli
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41334 Larisa, Greece; (S.A.); (G.A.)
| | - George Anifandis
- Department of Obstetrics and Gynaecology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41334 Larisa, Greece; (S.A.); (G.A.)
| | - Sandra Cecconi
- Department of Life, Health, and Environmental Sciences, Università dell’Aquila, 67100 L’Aquila, Italy
| | - Valentina Di Nisio
- Department of Gynecology and Reproductive Medicine, Karolinska University Hospital, SE-14186 Stockholm, Sweden;
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, SE-14186 Stockholm, Sweden
| |
Collapse
|
5
|
Marco A, Gargallo M, Ciriza J, Shikanov A, Baquedano L, García Pérez-Llantada J, Malo C. Current Fertility Preservation Steps in Young Women Suffering from Cancer and Future Perspectives. Int J Mol Sci 2024; 25:4360. [PMID: 38673945 PMCID: PMC11050570 DOI: 10.3390/ijms25084360] [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: 03/07/2024] [Revised: 04/05/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Childhood cancer incidence, especially in high-income countries, has led to a focus on preserving fertility in this vulnerable population. The common treatments, such as radiation and certain chemotherapeutic agents, though effective, pose a risk to fertility. For adult women, established techniques like embryo and egg freezing are standard, requiring ovarian stimulation. However, for prepubescent girls, ovarian tissue freezing has become the primary option, eliminating the need for hormonal preparation. This review describes the beginning, evolution, and current situation of the fertility preservation options for this young population. A total of 75 studies were included, covering the steps in the current fertility preservation protocols: (i) ovarian tissue extraction, (ii) the freezing method, and (iii) thawing and transplantation. Cryopreservation and the subsequent transplantation of ovarian tissue have resulted in successful fertility restoration, with over 200 recorded live births, including cases involving ovarian tissue cryopreserved from prepubescent girls. Despite promising results, challenges persist, such as follicular loss during transplantation, which is attributed to ischemic and oxidative damage. Optimizing ovarian tissue-freezing processes and exploring alternatives to transplantation, like in vitro systems for follicles to establish maturation, are essential to mitigating associated risks. Further research is required in fertility preservation techniques to enhance clinical outcomes in the future. Ovarian tissue cryopreservation appears to be a method with specific benefits, indications, and risks, which can be an important tool in terms of preserving fertility in younger women.
Collapse
Affiliation(s)
- Alicia Marco
- Faculty of Medicine, University of Zaragoza, 50018 Zaragoza, Spain;
| | - Marta Gargallo
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; (M.G.); (J.C.)
| | - Jesús Ciriza
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; (M.G.); (J.C.)
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
| | - Ariella Shikanov
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA;
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA
| | - Laura Baquedano
- Department of Gynecology, University Hospital Miguel Servat, 50009 Zaragoza, Spain;
| | | | - Clara Malo
- Institute for Health Research Aragón (IIS Aragón), 50009 Zaragoza, Spain; (M.G.); (J.C.)
- Tissue Microenvironment (TME) Lab, Aragón Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
| |
Collapse
|
6
|
Ren P, Tong X, Li J, Jiang H, Liu S, Li X, Lai M, Yang W, Rong Y, Zhang Y, Jin J, Ma Y, Pan W, Fan HY, Zhang S, Zhang YL. CRL4 DCAF13 E3 ubiquitin ligase targets MeCP2 for degradation to prevent DNA hypermethylation and ensure normal transcription in growing oocytes. Cell Mol Life Sci 2024; 81:165. [PMID: 38578457 PMCID: PMC10997554 DOI: 10.1007/s00018-024-05185-4] [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: 01/29/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 04/06/2024]
Abstract
The DNA methylation is gradually acquired during oogenesis, a process sustained by successful follicle development. However, the functional roles of methyl-CpG-binding protein 2 (MeCP2), an epigenetic regulator displaying specifical binding with methylated DNA, remains unknown in oogenesis. In this study, we found MeCP2 protein was highly expressed in primordial and primary follicle, but was almost undetectable in secondary follicles. However, in aged ovary, MeCP2 protein is significantly increased in both oocyte and granulosa cells. Overexpression of MeCP2 in growing oocyte caused transcription dysregulation, DNA hypermethylation, and genome instability, ultimately leading to follicle growth arrest and apoptosis. MeCP2 is targeted by DCAF13, a substrate recognition adaptor of the Cullin 4-RING (CRL4) E3 ligase, and polyubiquitinated for degradation in both cells and oocytes. Dcaf13-null oocyte exhibited an accumulation of MeCP2 protein, and the partial rescue of follicle growth arrest induced by Dcaf13 deletion was observed following MeCP2 knockdown. The RNA-seq results revealed that large amounts of genes were regulated by the DCAF13-MeCP2 axis in growing oocytes. Our study demonstrated that CRL4DCAF13 E3 ubiquitin ligase targets MeCP2 for degradation to ensure normal DNA methylome and transcription in growing oocytes. Moreover, in aged ovarian follicles, deceased DCAF13 and DDB1 protein were observed, indicating a potential novel mechanism that regulates ovary aging.
Collapse
Affiliation(s)
- Peipei Ren
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Xiaomei Tong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Junjian Li
- College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Huifang Jiang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Siya Liu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Xiang Li
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Mengru Lai
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Weijie Yang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yan Rong
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yingyi Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Jiamin Jin
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Yerong Ma
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| | - Weiwei Pan
- Department of Cell Biology, College of Medicine, Jiaxing University, Jiaxing, 314001, China
| | - Heng-Yu Fan
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
- Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China.
| | - Yin-Li Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
7
|
Ellibishy F, Tarek M, Abd-Elsalam MM, Elgayar N, El Bakly W. Metformin improves d-galactose induced premature ovarian insufficiency through PI3K-Akt-FOXO3a pathway. Adv Med Sci 2024; 69:70-80. [PMID: 38387407 DOI: 10.1016/j.advms.2024.02.004] [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: 10/17/2023] [Revised: 12/23/2023] [Accepted: 02/12/2024] [Indexed: 02/24/2024]
Abstract
PURPOSE Metformin (MET), a first-line treatment for type 2 diabetes mellitus, restores ovarian function in women with polycystic ovary syndrome. MET has been shown to increase the rate of success for in vitro fertilization when utilized in assisted reproductive technologies. This study was designed to examine the impact of MET on ovarian function and fertility in a mouse model of galactose-induced premature ovarian insufficiency (POI). We further investigated the underlying mechanisms. MATERIALS AND METHODS Female mice were divided into 4 groups: saline, d-galactose, d-galactose + MET, and MET. Body weight, ovarian index, and fertility were assessed. The hormonal profile was done. Advanced glycation end products (AGEPs), receptor for advanced glycation end products (RAGE), phosphoinositide 3-kinase (PI3K), protein kinase B (Akt), forkhead box O3a (FOXO3a) expression were measured. Ovarian follicle counting and morphology were analyzed. Immunohistochemistry of cleaved caspase-3 expression was performed. RESULTS Our findings demonstrated that MET reversed irregularities in the estrus cycle, enhanced the ovarian index, and improved the abnormal levels of hormones and AGEs induced by d-galactose. Furthermore, the expression levels of PI3K, Akt, FOXO3a, and RAGE were upregulated with d-galactose. However, MET attenuated their expression levels. The primordial follicles ratio was improved, whereas atretic follicles and apoptotic-related cleaved caspase-3 expression were decreased in the d-galactose + MET group compared to the d-galactose group. CONCLUSION This study demonstrates that MET partially rescued ovarian dysfunction and apoptosis induced by d-galactose via a mechanism involving PI3K-Akt-FOXO3a pathway. Our finding proposed that MET may be a promising alternative treatment for POI.
Collapse
Affiliation(s)
- Fatima Ellibishy
- Clinical Pharmacology Department, Faculty of Medicine, Kafr Elsheikh University, Kafr Elsheikh, Egypt.
| | - Maha Tarek
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Marwa M Abd-Elsalam
- Histology & Cell Biology Department, Faculty of Medicine, Kafr Elsheikh University, Kafr Elsheikh, Egypt
| | - Nesreen Elgayar
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Wesam El Bakly
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
8
|
Barretta M, Cacciottola L, Hossay C, Donnez J, Dolmans MM. Impact of human ovarian tissue manipulation on follicles: evidence of a potential first wave of follicle activation during fertility preservation procedures. J Assist Reprod Genet 2023; 40:2769-2776. [PMID: 37713145 PMCID: PMC10656389 DOI: 10.1007/s10815-023-02930-9] [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: 03/20/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023] Open
Abstract
PURPOSE The aim of this study was to investigate the impact of processing human ovarian tissue on follicle activation dynamics. METHODS Fresh ovarian tissue was retrieved from 9 women undergoing laparoscopic surgery for benign conditions. Biopsies from each patient were divided into 3 fragments, the first of which was immediately fixed in the operating room (T0) and the second and third just after processing at 25 (T25) and (T90) 90 min. To evaluate follicle activation, markers of the PI3K and Hippo signaling pathways were immunolabeled at each time point, targeting phospho-Akt (p-Akt) by immunohistochemistry and yes-associated protein (YAP) cellular localization in the granulosa cell layer by immunofluorescence. RESULTS Four hundred forty primordial follicles were evaluated for p-Akt and 420 for YAP. Significantly stronger p-Akt expression was observed at T25 (23.01 ± 13.45%; p=0.04) and T90 (38.99 ± 25.21%; p<0.001) than at T0 (2.72 ± 3.35%). A significant nucleus-to-cytoplasm shift in YAP was detected at T25 (1.21 ± 0.25; p=0.015 compared to T0 (0.95 ± 0.09), while T90 (1.10 ± 0.16) values were similar to T25. CONCLUSION Our data prove that ovarian tissue manipulation significantly impacts follicle dynamics by stimulating the PI3K and Hippo signaling pathways involved in primordial follicle activation. Further experimental evidence must nevertheless be gathered to understand and gain control of follicle activation mechanisms in non-physiological conditions (like ovarian tissue manipulation), in order to optimize fertility preservation and restoration strategies.
Collapse
Affiliation(s)
- Marta Barretta
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte B1.52.02, 1200, Brussels, Belgium
- Department of Biomedical Science for Health, University of Milan, Milan, Italy
| | - Luciana Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte B1.52.02, 1200, Brussels, Belgium
| | - Camille Hossay
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte B1.52.02, 1200, Brussels, Belgium
| | - Jacques Donnez
- Université Catholique de Louvain, Brussels, Belgium
- Société de Recherche pour l'Infertilité, Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte B1.52.02, 1200, Brussels, Belgium.
- Gynecology Department, Cliniques Universitaires Saint-Luc, Avenue Mounier 52, bte B1.52.02, 1200, Brussels, Belgium.
| |
Collapse
|
9
|
Nguyen TTA, Demeestere I. A Journey to Reach the Ovary Using Next-Generation Technologies. Int J Mol Sci 2023; 24:16593. [PMID: 38068916 PMCID: PMC10705884 DOI: 10.3390/ijms242316593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
Although effective in terms of the chances of future live birth, the current methods for fertility preservation, such as oocyte, embryo, or ovarian tissue cryopreservation, cannot be offered to all cancer patients in all clinical contexts. Expanding options for fertility preservation is crucial to addressing the need to encompass all situations. One emerging strategy is pharmacoprotection, a non-invasive approach that has the potential to fill existing gaps in fertility preservation. In addition to the identification of the most effective therapeutic agents, the potential for off-target effects remains one of the main limitations of this strategy for clinical application, particularly when healthy ovarian tissue is targeted. This review focuses on the advances in pharmacoprotective approaches and the challenge of targeting the ovaries to deliver these agents. The unique properties of gold nanoparticles (AuNPs) make them an attractive candidate for this purpose. We discuss how AuNPs meet many of the requirements for an ideal drug delivery system, as well as the existing limitations that have hindered the progression of AuNP research into more clinical trials. Additionally, the review highlights microRNA (miRNA) therapy as a next-generation approach to address the issues of fertility preservation and discusses the obstacles that currently impede its clinical availability.
Collapse
Affiliation(s)
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium;
| |
Collapse
|
10
|
Lundin K, Bentzen JG, Bozdag G, Ebner T, Harper J, Le Clef N, Moffett A, Norcross S, Polyzos NP, Rautakallio-Hokkanen S, Sfontouris I, Sermon K, Vermeulen N, Pinborg A. Good practice recommendations on add-ons in reproductive medicine†. Hum Reprod 2023; 38:2062-2104. [PMID: 37747409 PMCID: PMC10628516 DOI: 10.1093/humrep/dead184] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Indexed: 09/26/2023] Open
Abstract
STUDY QUESTION Which add-ons are safe and effective to be used in ART treatment? SUMMARY ANSWER Forty-two recommendations were formulated on the use of add-ons in the diagnosis of fertility problems, the IVF laboratory and clinical management of IVF treatment. WHAT IS KNOWN ALREADY The innovative nature of ART combined with the extremely high motivation of the patients has opened the door to the wide application of what has become known as 'add-ons' in reproductive medicine. These supplementary options are available to patients in addition to standard fertility procedures, typically incurring an additional cost. A diverse array of supplementary options is made available, encompassing tests, drugs, equipment, complementary or alternative therapies, laboratory procedures, and surgical interventions. These options share the common aim of stating to enhance pregnancy or live birth rates, mitigate the risk of miscarriage, or expedite the time to achieving pregnancy. STUDY DESIGN, SIZE, DURATION ESHRE aimed to develop clinically relevant and evidence-based recommendations focusing on the safety and efficacy of add-ons currently used in fertility procedures in order to improve the quality of care for patients with infertility. PARTICIPANTS/MATERIALS, SETTING, METHODS ESHRE appointed a European multidisciplinary working group consisting of practising clinicians, embryologists, and researchers who have demonstrated leadership and expertise in the care and research of infertility. Patient representatives were included in the working group. To ensure that the guidelines are evidence-based, the literature identified from a systematic search was reviewed and critically appraised. In the absence of any clear scientific evidence, recommendations were based on the professional experience and consensus of the working group. The guidelines are thus based on the best available evidence and expert agreement. Prior to publication, the guidelines were reviewed by 46 independent international reviewers. A total of 272 comments were received and incorporated where relevant. MAIN RESULTS AND THE ROLE OF CHANCE The multidisciplinary working group formulated 42 recommendations in three sections; diagnosis and diagnostic tests, laboratory tests and interventions, and clinical management. LIMITATIONS, REASONS FOR CAUTION Of the 42 recommendations, none could be based on high-quality evidence and only four could be based on moderate-quality evidence, implicating that 95% of the recommendations are supported only by low-quality randomized controlled trials, observational data, professional experience, or consensus of the development group. WIDER IMPLICATIONS OF THE FINDINGS These guidelines offer valuable direction for healthcare professionals who are responsible for the care of patients undergoing ART treatment for infertility. Their purpose is to promote safe and effective ART treatment, enabling patients to make informed decisions based on realistic expectations. The guidelines aim to ensure that patients are fully informed about the various treatment options available to them and the likelihood of any additional treatment or test to improve the chance of achieving a live birth. STUDY FUNDING/COMPETING INTEREST(S) All costs relating to the development process were covered from ESHRE funds. There was no external funding of the development process or manuscript production. K.L. reports speakers fees from Merck and was part of a research study by Vitrolife (unpaid). T.E. reports consulting fees from Gynemed, speakers fees from Gynemed and is part of the scientific advisory board of Hamilton Thorne. N.P.P. reports grants from Merck Serono, Ferring Pharmaceutical, Theramex, Gedeon Richter, Organon, Roche, IBSA and Besins Healthcare, speakers fees from Merck Serono, Ferring Pharmaceutical, Theramex, Gedeon Richter, Organon, Roche, IBSA and Besins Healthcare. S.R.H. declares being managing director of Fertility Europe, a not-for-profit organization receiving financial support from ESHRE. I.S. is a scientific advisor for and has stock options from Alife Health, is co-founder of IVFvision LTD (unpaid) and received speakers' fee from the 2023 ART Young Leader Prestige workshop in China. A.P. reports grants from Gedeon Richter, Ferring Pharmaceuticals and Merck A/S, consulting fees from Preglem, Novo Nordisk, Ferring Pharmaceuticals, Gedeon Richter, Cryos and Merck A/S, speakers fees from Gedeon Richter, Ferring Pharmaceuticals, Merck A/S, Theramex and Organon, travel fees from Gedeon Richter. The other authors disclosed no conflicts of interest. DISCLAIMER This Good Practice Recommendations (GPRs) document represents the views of ESHRE, which are the result of consensus between the relevant ESHRE stakeholders and are based on the scientific evidence available at the time of preparation.ESHRE GPRs should be used for information and educational purposes. They should not be interpreted as setting a standard of care or bedeemedinclusive of all proper methods of care, or be exclusive of other methods of care reasonably directed to obtaining the same results.Theydo not replace the need for application of clinical judgement to each individual presentation, or variations based on locality and facility type.Furthermore, ESHRE GPRs do not constitute or imply the endorsement, or favouring, of any of the included technologies by ESHRE.
Collapse
Affiliation(s)
| | - K Lundin
- Department Reproductive Medicine, Sahlgrenska University Hospital, Göteborg, Sweden
| | - J G Bentzen
- The Fertility Department, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
| | - G Bozdag
- Department Obstetrics and Gynecology, Koc University School of Medicine, Istanbul, Turkey
| | - T Ebner
- Department of Gynecology, Obstetrics, and Gynecological Endocrinology, Kepler University, MedCampus IV, Linz, Austria
| | - J Harper
- Institute for Women’s Health, London, UK
| | - N Le Clef
- European Society of Human Reproduction and Embryology, Brussels, Belgium
| | - A Moffett
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - N P Polyzos
- Department Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain
| | | | | | - K Sermon
- Research Group Reproduction and Genetics, Vrije Universiteit Brussel, Brussels, Belgium
| | - N Vermeulen
- European Society of Human Reproduction and Embryology, Brussels, Belgium
| | - A Pinborg
- The Fertility Department, Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
11
|
Cacciottola L, Vitale F, Donnez J, Dolmans MM. Use of mesenchymal stem cells to enhance or restore fertility potential: a systematic review of available experimental strategies. Hum Reprod Open 2023; 2023:hoad040. [PMID: 37954935 PMCID: PMC10637864 DOI: 10.1093/hropen/hoad040] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/15/2023] [Indexed: 11/14/2023] Open
Abstract
STUDY QUESTION To what extent does regenerative medicine with stem cell therapy help to address infertility issues for future clinical application? SUMMARY ANSWER Regenerative medicine using different stem cell sources is yielding promising results in terms of protecting the ovarian reserve from damage and senescence, and improving fertility potential in various preclinical settings. WHAT IS KNOWN ALREADY Regenerative medicine using stem cell therapy is emerging as a potential strategy to address a number of issues in the field of human reproduction. Indeed, different types of adult and fetal mesenchymal stem cells (MSCs) have been tested with promising results, owing to their ability to differentiate into different tissue lineages, move toward specific injured sites (homing), and generate a secretome with wound-healing, proangiogenic, and antioxidant capacities. STUDY DESIGN SIZE DURATION Guided by the checklist for preferred reporting items for systematic reviews and meta-analyses, we retrieved relevant studies from PubMed, Medline, and Embase databases until June 2023 using the following keywords: 'mesenchymal stem cells' AND 'ovarian follicles' OR 'ovarian tissue culture' OR 'ovarian follicle culture' OR 'cumulus oocyte complex'. Only peer-reviewed published articles written in English were included. PARTICIPANTS/MATERIALS SETTING METHODS The primary outcome for the experimental strategies was evaluation of the ovarian reserve, with a focus on follicle survival, number, and growth. Secondary outcomes involved analyses of other parameters associated with the follicle pool, such as hormones and growth factors, ovarian tissue viability markers including oxidative stress levels, oocyte growth and maturation rates, and of course pregnancy outcomes. MAIN RESULTS AND THE ROLE OF CHANCE Preclinical studies exploring MSCs from different animal origins and tissue sources in specific conditions were selected (n = 112), including: in vitro culture of granulosa cells, ovarian tissue and isolated ovarian follicles; ovarian tissue transplantation; and systemic or intraovarian injection after gonadotoxic or age-related follicle pool decline. Protecting the ovarian reserve from aging and gonadotoxic damage has been widely tested in vitro and in vivo using murine models and is now yielding initial data in the first ever case series of patients with premature ovarian insufficiency. Use of MSCs as feeder cells in ovarian tissue culture was found to improve follicle outcomes and oocyte competence, bringing us one step closer to future clinical application. MSCs also have proved effective at boosting revascularization in the transplantation site when grafting ovarian tissue in experimental animal models. LIMITATIONS REASONS FOR CAUTION While preclinical results look promising in terms of protecting the ovarian reserve in different experimental models (especially those in vitro using various mammal experimental models and in vivo using murine models), there is still a lot of work to do before this approach can be considered safe and successfully implemented in a clinical setting. WIDER IMPLICATIONS OF THE FINDINGS All gathered data on the one hand show that regenerative medicine techniques are quickly gaining ground among innovative techniques being developed for future clinical application in the field of reproductive medicine. After proving MSC effectiveness in preclinical settings, there is still a lot of work to do before MSCs can be safely and effectively used in different clinical applications. STUDY FUNDING/COMPETING INTERESTS This study was supported by grants from the Fonds National de la Recherche Scientifique de Belgique (FNRS-PDR T.0077.14, FNRS-CDR J.0063.20, and grant 5/4/150/5 awarded to Marie-Madeleine Dolmans), Fonds Spéciaux de Recherche, and the Fondation St Luc. None of the authors have any competing interest to disclose. REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- L Cacciottola
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - F Vitale
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - J Donnez
- Society for Research into Infertility, Brussels, Belgium
- Université Catholique de Louvain, Brussels, Belgium
| | - M M Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
- Department of Gynecology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
12
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
13
|
Liu M, Xiao B, Zhu Y, Chen M, Huang J, Guo H, Wang F. MicroRNA-144-3p protects against chemotherapy-induced apoptosis of ovarian granulosa cells and activation of primordial follicles by targeting MAP3K9. Eur J Med Res 2023; 28:264. [PMID: 37537658 PMCID: PMC10399062 DOI: 10.1186/s40001-023-01231-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 07/15/2023] [Indexed: 08/05/2023] Open
Abstract
Premature ovarian failure (POF) is defined by amenorrhea, ovarian atrophy, hypoestrogenism, elevated gonadotropin level, and infertility under the age of 40. POF is frequently induced by chemotherapeutic agents. However, the underlying mechanisms regarding chemotherapy-mediated damage to ovarian function are unclear. In this study, enhanced apoptosis of granulosa cells (GCs) and aberrant activation of primordial follicles were observed in a POF mouse model induced by cisplatin. We subsequently observed significant downregulation of miR-144-3p and upregulation of mitogen-activated protein kinase kinase kinase 9 (MAP3K9) in primary ovarian GCs from POF mice, as revealed by microarrays. Furthermore, MAP3K9 expression was higher in human ovarian granulosa cells (COV434) treated with cisplatin and was identified as a novel target of miR-144-3p. Functional analysis revealed that miR-144-3p attenuated cisplatin induced apoptosis of GCs via silencing MAP3K9 expression, which suppressed the activity of the downstream p38 mitogen activated protein kinase (MAPK) pathway. Meanwhile, miR-144-3p prevented premature primordial follicle depletion in cisplatin-induced POF mice through targeting Map3k9, which led to a decline in the phosphorylation and activation of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase b (AKT) pathway. Taken together, this study revealed the protective effects of miR-144-3p on ovarian function and shed light on the epigenetic regulatory mechanism in the development of POF, which might provide new biomarkers for the ovarian reserve.
Collapse
Affiliation(s)
- Meng Liu
- Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Bang Xiao
- Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Yiqing Zhu
- Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Meiting Chen
- Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Jinfeng Huang
- Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Haiyan Guo
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China.
| | - Fang Wang
- Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
14
|
Yuan L, Huang W, Bi Y, Chen S, Wang X, Li T, Wei P, Du J, Zhao L, Liu B, Yang Y. G-CSF-mobilized peripheral blood mononuclear cells combined with platelet-rich plasma restored the ovarian function of aged rats. J Reprod Immunol 2023; 158:103953. [PMID: 37209460 DOI: 10.1016/j.jri.2023.103953] [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: 11/16/2022] [Revised: 04/11/2023] [Accepted: 05/10/2023] [Indexed: 05/22/2023]
Abstract
BACKGROUND Regenerative medicine with peripheral blood mononuclear cell (PBMC) transplantation sheds light on the issue of premature ovarian insufficiency (POI). However, the efficiency of PBMC treatment in natural ovarian aging (NOA) remains unclear. METHODS Thirteen-month-old female Sprague-Dawley (SD) rats were used to verify the NOA model. Seventy-two NOA rats were randomly divided into three groups: the NOA control group, PBMC group, and PBMC+platelet-rich plasma (PRP) group. PBMCs and PRP were transplanted by intraovarian injection. The effects on ovarian function and fertility were measured after transplantation. RESULTS Transplantation of PBMCs could restore the normal estrous cycle, consistent with the recovery of serum sex hormone levels, increased follicle numbers at all stages, and restoration of fertility by facilitating pregnancy and live birth. Moreover, when combined with PRP injection, these effects were more significant. The male-specific SRY gene was detected in the ovary at all four time points, suggesting that PBMCs continuously survived and functioned in NOA rats. In addition, after PBMC treatment, the expression of angiogenesis-related and glycolysis-related markers in the ovaries was upregulated, which indicated that these effects were associated with angiogenesis and glycolysis. CONCLUSIONS PBMC transplantation restores the ovarian functions and fertility of NOA rats, and PRP could enhance the efficiency. Increased ovarian vascularization, follicle production, and glycolysis are likely the major mechanisms.
Collapse
Affiliation(s)
- Lifang Yuan
- Reproductive Medical Center, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Weiyu Huang
- Reproductive Medical Center, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yin Bi
- Reproductive Medical Center, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Saiqiong Chen
- Department of Obstetrics and Gynecology, the Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou, Guangxi 545005, China
| | - Xi Wang
- Reproductive Medical Center, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Ting Li
- Reproductive Medical Center, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Peiru Wei
- Reproductive Medical Center, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jiebing Du
- Guangxi Maternal and Child Healthcare Hospital, Nanning, Guangxi 530002, China
| | - Ling Zhao
- Reproductive Medical Center, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Bo Liu
- Reproductive Medical Center, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Yihua Yang
- Reproductive Medical Center, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
15
|
Lawley SD, Johnson J. Slowest first passage times, redundancy, and menopause timing. J Math Biol 2023; 86:90. [PMID: 37148411 DOI: 10.1007/s00285-023-01921-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 04/01/2023] [Accepted: 04/17/2023] [Indexed: 05/08/2023]
Abstract
Biological events are often initiated when a random "searcher" finds a "target," which is called a first passage time (FPT). In some biological systems involving multiple searchers, an important timescale is the time it takes the slowest searcher(s) to find a target. For example, of the hundreds of thousands of primordial follicles in a woman's ovarian reserve, it is the slowest to leave that trigger the onset of menopause. Such slowest FPTs may also contribute to the reliability of cell signaling pathways and influence the ability of a cell to locate an external stimulus. In this paper, we use extreme value theory and asymptotic analysis to obtain rigorous approximations to the full probability distribution and moments of slowest FPTs. Though the results are proven in the limit of many searchers, numerical simulations reveal that the approximations are accurate for any number of searchers in typical scenarios of interest. We apply these general mathematical results to models of ovarian aging and menopause timing, which reveals the role of slowest FPTs for understanding redundancy in biological systems. We also apply the theory to several popular models of stochastic search, including search by diffusive, subdiffusive, and mortal searchers.
Collapse
Affiliation(s)
- Sean D Lawley
- Department of Mathematics, University of Utah, Salt Lake City, UT, 84112, USA.
| | - Joshua Johnson
- Division of Reproductive Sciences, Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
16
|
Sakaguchi K, Kawano K, Otani Y, Yanagawa Y, Katagiri S, Telfer EE. Relationship between Amino Acid Metabolism and Bovine In Vitro Follicle Activation and Growth. Animals (Basel) 2023; 13:ani13071141. [PMID: 37048397 PMCID: PMC10093075 DOI: 10.3390/ani13071141] [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: 01/30/2023] [Revised: 03/06/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
The amino acid metabolism of bovine follicles during in vitro growth (IVG) was evaluated to identify potential indicators of health during culture. The bovine ovarian cortex was sliced, prepared as strips, and cultured for 6 days. Tissue samples were examined histologically before and after 6 days of culture, and the degree of follicle activation was classified as either high or low based on the number of growing secondary follicles present (high: 7~11; low: 0~1). In a separate experiment, secondary follicles (diameter range: 100~200 μm) were manually isolated and cultured, and their growth was monitored for 6 days. Cultured follicles were classified as growth or degenerate based on diameter change during culture (growth: +60.5~74.1 μm; degenerate: -28~15.2 μm). Free amino acids and their metabolites were measured in the spent culture medium from each group. In cultured ovarian cortical strips, the concentration of α-aminoadipic acid was significantly higher in the low activation group than in the high group (p < 0.05), while those of methionine, lysine, and arginine were higher in the high activation group. In cultured isolated secondary follicles, concentrations of methionine, tyrosine, histidine, and hydroxyproline were higher in the degenerate group (p ≤ 0.05). In conclusion, amino acid metabolism has the potential to serve as an indicator of primordial follicle activation and subsequent growth rate during bovine IVG.
Collapse
Affiliation(s)
- Kenichiro Sakaguchi
- Institute of Cell Biology, School of Biological Sciences, College of Science and Engineering, University of Edinburgh, The Hugh Robson Building, 15 George Square, Edinburgh EH8 9XD, UK
- Laboratory of Theriogenology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Kohei Kawano
- Laboratory of Theriogenology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Yuki Otani
- Laboratory of Anatomy, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Yojiro Yanagawa
- Laboratory of Theriogenology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Seiji Katagiri
- Laboratory of Theriogenology, Department of Clinical Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18 Nishi 9, Kita-ku, Sapporo 060-0818, Japan
| | - Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, College of Science and Engineering, University of Edinburgh, The Hugh Robson Building, 15 George Square, Edinburgh EH8 9XD, UK
| |
Collapse
|
17
|
Houeis L, Dolmans MM. Summary of the ISFP congress, Brussels, 10-12 November, 2022. J Assist Reprod Genet 2023; 40:433-442. [PMID: 36765026 PMCID: PMC10033808 DOI: 10.1007/s10815-023-02720-3] [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: 01/10/2023] [Accepted: 01/10/2023] [Indexed: 02/12/2023] Open
Abstract
The 7th International Congress of the ISFP was held in Brussels in November 2022. Hundreds of attendees from all over the world had the rare opportunity to hear the most distinguished leaders discuss and debate the latest advances in the field. Participants were also able to attend workshops under the guidance of skilled practitioners. Numerous topics were considered, including a recap on fertility preservation approaches in cancer and benign pathologies and a section on male factor infertility. Other aspects covered were in vitro maturation and poor responders, the impact of chemotherapy on the ovary, and future perspectives. Participants had the chance to listen to a symposium on fertility preservation techniques, and finally, a keynote lecture on fertility preservation in gynecological cancers brought this prominent and highly influential event to a close.
Collapse
Affiliation(s)
- Lara Houeis
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Marie-Madeleine Dolmans
- Gynecology Research Unit, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium.
- Department of Gynecology, Cliniques Universitaires Saint-Luc, Brussels, Belgium.
| |
Collapse
|
18
|
Grosbois J, Bailie EC, Kelsey TW, Anderson RA, Telfer EE. Spatio-temporal remodelling of the composition and architecture of the human ovarian cortical extracellular matrix during in vitro culture. Hum Reprod 2023; 38:444-458. [PMID: 36721914 PMCID: PMC9977129 DOI: 10.1093/humrep/dead008] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/29/2022] [Indexed: 02/02/2023] Open
Abstract
STUDY QUESTION How does in vitro culture alter the human ovarian cortical extracellular matrix (ECM) network structure? SUMMARY ANSWER The ECM composition and architecture vary in the different layers of the ovarian cortex and are remodelled during in vitro culture. WHAT IS KNOWN ALREADY The ovarian ECM is the scaffold within which follicles and stromal cells are organized. Its composition and structural properties constantly evolve to accommodate follicle development and expansion. Tissue preparation for culture of primordial follicles within the native ECM involves mechanical loosening; this induces undefined modifications in the ECM network and alters cell-cell contact, leading to spontaneous follicle activation. STUDY DESIGN, SIZE, DURATION Fresh ovarian cortical biopsies were obtained from six women aged 28-38 years (mean ± SD: 32.7 ± 4.1 years) at elective caesarean section. Biopsies were cut into fragments of ∼4 × 1 × 1 mm and cultured for 0, 2, 4, or 6 days (D). PARTICIPANTS/MATERIALS, SETTING, METHODS Primordial follicle activation, stromal cell density, and ECM-related protein (collagen, elastin, fibronectin, laminin) positive area in the entire cortex were quantified at each time point using histological and immunohistological analysis. Collagen and elastin content, collagen fibre characteristics, and follicle distribution within the tissue were further quantified within each layer of the human ovarian cortex, namely the outer cortex, the mid-cortex, and the cortex-medulla junction regions. MAIN RESULTS AND THE ROLE OF CHANCE Primordial follicle activation occurred concomitantly with a loosening of the ovarian cortex during culture, characterized by an early decrease in stromal cell density from 3.6 ± 0.2 × 106 at day 0 (D0) to 2.8 ± 0.1 × 106 cells/mm3 at D2 (P = 0.033) and a dynamic remodelling of the ECM. Notably, collagen content gradually fell from 55.5 ± 1.7% positive area at D0 to 42.3 ± 1.1% at D6 (P = 0.001), while elastin increased from 1.1 ± 0.2% at D0 to 1.9 ± 0.1% at D6 (P = 0.001). Fibronectin and laminin content remained stable. Moreover, collagen and elastin distribution were uneven throughout the cortex and during culture. Analysis at the sub-region level showed that collagen deposition was maximal in the outer cortex and the lowest in the mid-cortex (69.4 ± 1.2% versus 53.8 ± 0.8% positive area, respectively, P < 0.0001), and cortical collagen staining overall decreased from D0 to D2 (65.2 ± 2.4% versus 60.6 ± 1.8%, P = 0.033) then stabilized. Elastin showed the converse distribution, being most concentrated at the cortex-medulla junction (3.7 ± 0.6% versus 0.9 ± 0.2% in the outer cortex, P < 0.0001), and cortical elastin peaked at D6 compared to D0 (3.1 ± 0.5% versus 1.3 ± 0.2%, P < 0.0001). This was corroborated by a specific signature of the collagen fibre type across the cortex, indicating a distinct phenotype of the ovarian cortical ECM depending on region and culture period that might be responsible for the spatio-temporal and developmental pattern of follicular distribution observed within the cortex. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Ovarian cortical biopsies were obtained from women undergoing caesarean sections. As such, the data obtained may not accurately reflect the ECM distribution and structure of non-pregnant women. WIDER IMPLICATIONS OF THE FINDINGS Clarifying the composition and architecture signature of the human ovarian cortical ECM provides a foundation for further exploration of ovarian microenvironments. It is also critical for understanding the ECM-follicle interactions regulating follicle quiescence and awakening, leading to improvements in both in vitro activation and in vitro growth techniques. STUDY FUNDING/COMPETING INTEREST(S) Medical Research Council grant MR/R003246/1 and Wellcome Trust Collaborative Award in Science: 215625/Z/19/Z. The authors have no conflicts to declare. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Johanne Grosbois
- Institute of Cell Biology, Hugh Robson Building, University of Edinburgh, Edinburgh, UK
| | - Emily C Bailie
- Institute of Cell Biology, Hugh Robson Building, University of Edinburgh, Edinburgh, UK
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Tom W Kelsey
- School of Computer Science, University of St Andrews, St Andrews, UK
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Evelyn E Telfer
- Institute of Cell Biology, Hugh Robson Building, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
19
|
Poppe KG, Thomas AL, Kleynen P, Veltri F, Sitoris G, Autin C. Impact of Thyroid Function on Oocyte Retrieval and Cryopreservation Rate in Women Consulting for Anticipated Gamete Exhaustion. Thyroid 2023; 33:389-392. [PMID: 36515164 DOI: 10.1089/thy.2022.0528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Kris G Poppe
- Endocrine Clinic, Obstetrics and Reproductive Medicine Unit, Centre Hospitalier Universitaire (CHU) Saint-Pierre, Brussels, Belgium
- Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Anne-Laure Thomas
- Departement of Gynecology, Obstetrics and Reproductive Medicine Unit, Centre Hospitalier Universitaire (CHU) Saint-Pierre, Brussels, Belgium
| | - Pierre Kleynen
- Endocrine Clinic, Obstetrics and Reproductive Medicine Unit, Centre Hospitalier Universitaire (CHU) Saint-Pierre, Brussels, Belgium
| | - Flora Veltri
- Endocrine Clinic, Obstetrics and Reproductive Medicine Unit, Centre Hospitalier Universitaire (CHU) Saint-Pierre, Brussels, Belgium
| | - Georgiana Sitoris
- Endocrine Clinic, Obstetrics and Reproductive Medicine Unit, Centre Hospitalier Universitaire (CHU) Saint-Pierre, Brussels, Belgium
| | - Candice Autin
- Departement of Gynecology, Obstetrics and Reproductive Medicine Unit, Centre Hospitalier Universitaire (CHU) Saint-Pierre, Brussels, Belgium
| |
Collapse
|
20
|
Bai X, Wang S. Signaling pathway intervention in premature ovarian failure. Front Med (Lausanne) 2022; 9:999440. [PMID: 36507521 PMCID: PMC9733706 DOI: 10.3389/fmed.2022.999440] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Premature ovarian failure (POF) is a multifactorial disease that refers to the occurrence of secondary amenorrhea, estrogen decrease, and gonadotropin increase in women under the age of 40. The prevalence of POF is increasing year by year, and the existing instances can be categorized as primary or secondary cases. This disease has adverse effects on both the physiology and psychology of women. Hormone replacement therapy is the recommended treatment for POF, and a multidisciplinary strategy is required to enhance the quality of life of patients. According to recent studies, the primary mechanism of POF is the depletion of ovarian reserve function as a result of increased primordial follicular activation or primordial follicular insufficiency. Therefore, understanding the processes of primordial follicle activation and associated pathways and exploring effective interventions are important for the treatment of POF.
Collapse
|
21
|
The programmed death of fetal oocytes and the correlated surveillance mechanisms. REPRODUCTIVE AND DEVELOPMENTAL MEDICINE 2022. [DOI: 10.1097/rd9.0000000000000016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
22
|
Meng L, Sugishita Y, Nishimura S, Uekawa A, Suzuki-Takahashi Y, Suzuki N. Investigation of the optimal culture time for warmed bovine ovarian tissues before transplantation. Biol Reprod 2022; 107:1319-1330. [PMID: 35980811 DOI: 10.1093/biolre/ioac161] [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: 12/12/2021] [Revised: 04/25/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Ovarian tissue cryopreservation by vitrification is an effective technique, but there are still many unresolved issues related to the procedure. The aim of this study was to investigate the optimal culture time of post-warmed ovarian tissues and their viability before ovarian tissue transplantation. The bovine ovarian tissues were used to evaluate the effect of post-warming culture periods (0, 0.25, 0.5, 1, 2, 5 and 24 hours) in the levels of residual cryoprotectant, LDH release, ROS generation, gene and protein abundance, and follicle viability and its mitochondrial membrane potential. Residual cryoprotectant (CPA) concentration decreased significantly after 1 hour of culture. The warmed ovarian tissues that underwent between 0 to 2 hours of culture time showed similar LDH and ROS levels compared to fresh non-frozen tissues. The AMH transcript abundance did not differ in any of the groups. No increase in the relative transcript abundance and protein level of Caspase 3 and Cleaved-Caspase 3, respectively, in the first 2 hours of culture after warming. On the other hand, an increased protein level of double stranded DNA breaks (gamma-H2AX) was observed in post-warmed tissues disregarding the length of culture time, and a temporary reduction in pan-AKT was detected in post-warming tissues between 0 to 0.25 hours of culture time. Prolonged culture time lowered the percentage of viable follicles in warmed tissues, but it did not seem to affect the follicular mitochondrial membrane potential. In conclusion, 1 to 2 hours of culture time would be optimal for vitrified-warmed tissues before transplantation.
Collapse
Affiliation(s)
- Lingbo Meng
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yodo Sugishita
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Japan.,Department of Frontier Medicine, Institute of Medical Science, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Sandy Nishimura
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Atsushi Uekawa
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yuki Suzuki-Takahashi
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Japan.,Department of Frontier Medicine, Institute of Medical Science, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Nao Suzuki
- Department of Obstetrics and Gynecology, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
23
|
Dolmans MM, Demeestere I, Anckaert E, De Vos M. Proceedings of the Oncofertility Congress of the "Freezing Ovarian Tissue and Oocytes" (FOTO) Consortium Brussels. J Assist Reprod Genet 2022; 39:1715-1725. [PMID: 35751830 PMCID: PMC9428079 DOI: 10.1007/s10815-022-02552-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 01/19/2023] Open
Affiliation(s)
- Marie-Madeleine Dolmans
- Gynecology Department, Cliniques Universitaires St-Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium
- Gynecology Research Laboratory, Institut de Recherche Expérimentale Et Clinique, Université Catholique de Louvain, Avenue Mounier 52, bte B1.52.02, 1200, Brussels, Belgium
| | - Isabelle Demeestere
- Research Laboratory On Human Reproduction, Fertility Clinic, CUB-Hôpital Erasme, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Ellen Anckaert
- Follicle Biology Laboratory, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium
| | - Michel De Vos
- Follicle Biology Laboratory, Vrije Universiteit Brussel (VUB), 1090, Brussels, Belgium.
- Brussels IVF, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, Laarbeeklaan 101, 1090, Brussels, Belgium.
| |
Collapse
|
24
|
Méndez M, Fabregues F, Ferreri J, Calafell JM, Villarino A, Otero J, Farre R, Carmona F. Biomechanical characteristics of the ovarian cortex in POI patients and functional outcomes after drug-free IVA. J Assist Reprod Genet 2022; 39:1759-1767. [PMID: 35904669 PMCID: PMC9428073 DOI: 10.1007/s10815-022-02579-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/19/2022] [Indexed: 01/19/2023] Open
Abstract
PURPOSE There is increasing evidence that the ovarian extracellular matrix (ECM) plays a critical role in follicle development. The rigidity of the cortical ECM limits expansion of the follicle and consequently oocyte maturation, maintaining the follicle in its quiescent state. Quiescent primordial, primary, and secondary follicles still exist in primary ovarian insufficiency (POI) patients, and techniques as in vitro activation (IVA) and drug-free IVA have recently been developed aiming to activate these follicles based on the Hippo signaling disruption that is essential in mechanotransduction. In this context, we analyze the effect of drug-free IVA in POI patients, comparing the relationship between possible resumption ovarian function and biomechanical properties of ovarian tissue. METHODS Nineteen POI patients according to ESHRE criteria who underwent drug-free IVA by laparoscopy between January 2018 and December 2019 and were followed up for a year after the intervention. A sample of ovarian cortex taken during the intervention was analyzed by atomic force microscopy (AFM) in order to quantitatively measure tissue stiffness (Young's elastic modulus, E) at the micrometer scale. Functional outcomes after drug-free were analyzed. RESULTS Resumption of ovarian function was observed in 10 patients (52.6%) and two of them became pregnant with live births. There were no differences in clinical characteristics (age and duration of amenorrhea) and basal hormone parameters (FSH and AMH) depending on whether or not there was activation after surgery. However, ovarian cortex stiffness was significantly greater in patients with ovarian activity after drug-free IVA: median E = 5519 Pa (2260-11,296) vs 1501 (999-3474); p-value < 0.001. CONCLUSIONS Biomechanical properties of ovarian cortex in POI patients have a great variability, and higher ovarian tissue stiffness entails a more favorable status when drug-free IVA is applied in their treatment. This status is probably related to an ovary with more residual follicles, which would explain a greater possibility of ovarian follicular reactivations after treatment.
Collapse
Affiliation(s)
- M. Méndez
- Institute of Gynaecology Obstetrics and Neonatology (ICGON), Hospital Clínic de Barcelona, Barcelona, Spain
| | - F. Fabregues
- Institute of Gynaecology Obstetrics and Neonatology (ICGON), Hospital Clínic de Barcelona, Barcelona, Spain ,August Pi Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - J. Ferreri
- Institute of Gynaecology Obstetrics and Neonatology (ICGON), Hospital Clínic de Barcelona, Barcelona, Spain
| | - J. M. Calafell
- Institute of Gynaecology Obstetrics and Neonatology (ICGON), Hospital Clínic de Barcelona, Barcelona, Spain
| | - A. Villarino
- Biophysics and Bioengineering Unit, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - J. Otero
- Biophysics and Bioengineering Unit, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain ,CIBER of Respiratory Diseases, Madrid, Spain
| | - R. Farre
- August Pi Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain ,Biophysics and Bioengineering Unit, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain ,CIBER of Respiratory Diseases, Madrid, Spain
| | - F. Carmona
- Institute of Gynaecology Obstetrics and Neonatology (ICGON), Hospital Clínic de Barcelona, Barcelona, Spain ,August Pi Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| |
Collapse
|
25
|
Let-7a mimic transfection reduces chemotherapy-induced damage in a mouse ovarian transplantation model. Sci Rep 2022; 12:10863. [PMID: 35760952 PMCID: PMC9237019 DOI: 10.1038/s41598-022-14926-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 06/15/2022] [Indexed: 11/09/2022] Open
Abstract
Pharmacological approaches offer a non-invasive and promising option for fertility preservation in young female cancer patients undergoing gonadotoxic therapy. The GnRH-agonists are the only clinically available drugs in this indication, but their use and mechanisms of protection are still controversial. Recently, we have investigated new targeted drugs based on microRNA (miRNA) replacement therapy, and have identified the let-7a miRNA as candidate for fertility preservation strategies. Here, the effect of let-7a replacement during chemotherapy exposure on follicular growth and oocyte maturation capacity was investigated using a mouse ovarian-kidney transplantation model. Newborn mouse ovaries were cultured under different conditions; control, chemotherapy exposure (4-hydroperoxycyclophosphamide, 4-HC), and co-treatment with 4-HC and let-7a mimic transfection (4-HC + let-7a). The ovaries were then transplanted under the kidney capsule of recipient mice and follicular growth, survival, and oocyte in vitro maturation were assessed after 3 weeks. The results showed that the follicular pool was highest in the control group but higher in the 4-HC + let-7a group than the 4-HC group. DNA-damage/apoptosis ratios were higher in all 4-HC-exposed groups compared to control but were reduced in the 4-HC + let-7a group. In addition, the post-transplantation oocyte in vitro maturation rate was higher in the 4-HC + let-7a group compared to the 4-HC group, suggesting better oocyte quality. These results provide new information regarding the beneficial effects of let-7a replacement against chemotherapy-induced ovarian damage and open new perspectives for future in vivo applications.
Collapse
|
26
|
Fibroblast growth factor-2 promotes in vitro activation of cat primordial follicles. ZYGOTE 2022; 30:730-734. [PMID: 35416145 DOI: 10.1017/s0967199421000964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This study evaluated the effect of fibroblast growth factor-2 (FGF-2) on the morphology, primordial follicle activation and growth after in vitro culture of domestic cat ovarian tissue. Ovaries (n = 12) from prepubertal domestic cats were collected and fragmented. One fragment was fixed for histological analysis (fresh control). The remaining fragments were incubated in control medium alone or with 10, 50 or 100 ng/ml FGF-2 for 7 days. After in vitro culture, the following endpoints were analyzed: morphology, activation by counting primordial and developing follicles, and growth (follicle and oocyte diameters). Treatment with 100 ng/ml FGF-2 maintained (P > 0.05) the percentage of normal follicles similar to fresh control. Follicle survival was greater (P < 0.05) after culture in 100 ng/ml FGF-2 than in 50 ng/ml FGF-2. The percentage of primordial follicles decreased (P < 0.05) and the percentage of developing follicles increased (P < 0.05) in all treatments compared with fresh tissue. The proportion of developing follicles increased (P < 0.05) in tissues incubated with 100 ng/ml FGF-2 compared with control medium and other FGF-2 concentrations. Furthermore, culture in 10 or 100 ng/ml FGF-2 resulted in increased (P < 0.05) follicle and oocyte diameters compared with fresh tissues and MEM+. In conclusion, FGF-2 at 100 ng/ml maintains follicle survival and promotes the in vitro activation and growth of cat primordial follicles.
Collapse
|
27
|
Cakiroglu Y, Yuceturk A, Karaosmanoglu O, Kopuk SY, Korun ZEU, Herlihy N, Scott RT, Tiras B, Seli E. Ovarian reserve parameters and IVF outcomes in 510 women with poor ovarian response (POR) treated with intraovarian injection of autologous platelet rich plasma (PRP). Aging (Albany NY) 2022; 14:2513-2523. [PMID: 35320118 PMCID: PMC9004561 DOI: 10.18632/aging.203972] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/28/2022] [Indexed: 05/27/2023]
Abstract
The aim of the current study was to characterize ovarian reserve parameters and IVF outcomes in women with a history of poor ovarian response (POR) treated with intraovarian injection of autologous platelet rich plasma (PRP). Reproductive age women (N=510; age range 30-45yo) diagnosed with POR based on Poseidon criteria were included in the study. PRP treatment resulted in higher AFC, higher serum AMH, lower serum FSH, and a higher number of mature oocytes and cleavage and blastocyst stage embryos. After PRP injection, 22 women (4.3%) conceived spontaneously, 14 (2.7%) were lost to follow up, and 474 (92.9%) attempted IVF. Among women who attempted IVF, 312 (65.8%) generated embryos and underwent embryo transfer, 83 (17.5%) achieved a pregnancy, and 54 (11.4%) achieved sustained implantation/live birth (SI/LB). In total, of the 510 women with POR and mean age of 40.3, PRP resulted in improvement of ovarian reserve parameters, a pregnancy rate of 20.5% and SI/LB rate of 12.9%. Our findings suggest that PRP treatment may be considered in women with POR. For wider clinical application, its clinical efficacy will need to be demonstrated in prospective randomized clinical trials.
Collapse
Affiliation(s)
- Yigit Cakiroglu
- Acibadem Maslak Hospital Assisted Reproductive Technologies Unit, Sariyer, Istanbul, Turkey
- Acibadem Mehmet Ali Aydinlar University, Department of Obstetrics and Gynecology, Sariyer, Istanbul, Turkey
| | - Aysen Yuceturk
- Acibadem Maslak Hospital Assisted Reproductive Technologies Unit, Sariyer, Istanbul, Turkey
| | - Ozge Karaosmanoglu
- Acibadem Maslak Hospital Assisted Reproductive Technologies Unit, Sariyer, Istanbul, Turkey
| | - Sule Yildirim Kopuk
- Acibadem Maslak Hospital Assisted Reproductive Technologies Unit, Sariyer, Istanbul, Turkey
| | - Zeynep Ece Utkan Korun
- Acibadem Maslak Hospital Assisted Reproductive Technologies Unit, Sariyer, Istanbul, Turkey
| | - Nola Herlihy
- IVI RMA New Jersey, Basking Ridge, NJ 07920, USA
- Department of Obstetrics and Gynecology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Richard T. Scott
- IVI RMA New Jersey, Basking Ridge, NJ 07920, USA
- Department of Obstetrics and Gynecology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bulent Tiras
- Acibadem Maslak Hospital Assisted Reproductive Technologies Unit, Sariyer, Istanbul, Turkey
- Acibadem Mehmet Ali Aydinlar University, Department of Obstetrics and Gynecology, Sariyer, Istanbul, Turkey
| | - Emre Seli
- IVI RMA New Jersey, Basking Ridge, NJ 07920, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
28
|
Ntemou E, Vidal PD, Alexandri C, Van den Steen G, Lambertini M, Demeestere I. Ovarian toxicity of carboplatin and paclitaxel in mouse carriers of mutation in BRIP1 tumor suppressor gene. Sci Rep 2022; 12:1658. [PMID: 35105904 PMCID: PMC8807594 DOI: 10.1038/s41598-022-05357-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 01/11/2022] [Indexed: 11/19/2022] Open
Abstract
More than 10% of women diagnosed with breast cancer during reproductive age carry hereditary germline pathogenic variants in high-penetrance BRCA genes or in others genes involved in DNA repair mechanisms such as PALB2, BRIP or ATM. Anticancer treatments may have an additional negative impact on the ovarian reserve and subsequently on the fertility of young patients carrying such mutations. Recently, the combination of carboplatin and paclitaxel is being recommended to these BRCA-mutated patients as neoadjuvant therapy. However, the impact on the ovary is unknown. Here, we investigated their effect of on the ovarian reserve using mice carriers of BRCA1-interacting protein C-terminal helicase-1 (BRIP1) mutation that plays an important role in BRCA1-dependent DNA repair. Results revealed that the administration of carboplatin or paclitaxel did not affect the ovarian reserve although increased DNA double-strand breaks were observed with carboplatin alone. Co-administration of carboplatin and paclitaxel resulted in a significant reduction of the ovarian reserve leading to a lower IVF performance, and an activation of the PI3K-Pten pathway, irrespective of the genetic background. This study suggests that co-administration of carboplatin and paclitaxel induces cumulative ovarian damage and infertility but a heterozygote genetic predisposition for DNA damage related to BRCA1 gene function does not increase this risk.
Collapse
Affiliation(s)
- E Ntemou
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - P Diaz Vidal
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - C Alexandri
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - G Van den Steen
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - M Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genoa, Italy
- Department of Medical Oncology, UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - I Demeestere
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium.
- Obstetrics and Gynaecology Department, Erasme Hospital, Brussels, Belgium.
| |
Collapse
|
29
|
Mi X, Jiao W, Yang Y, Qin Y, Chen ZJ, Zhao S. HGF Secreted by Mesenchymal Stromal Cells Promotes Primordial Follicle Activation by Increasing the Activity of the PI3K-AKT Signaling Pathway. Stem Cell Rev Rep 2022; 18:1834-1850. [PMID: 35089464 PMCID: PMC9209380 DOI: 10.1007/s12015-022-10335-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2022] [Indexed: 01/08/2023]
Abstract
Primordial follicle activation is fundamental for folliculogenesis and for the maintenance of fertility. An effective therapeutic strategy for patients with premature ovarian insufficiency (POI) is to promote the activation of residual primordial follicles. The secretome of human umbilical cord mesenchymal stromal cells (hUC-MSC-sec) contains several components that might promote the activation of primordial follicles. In the present study, we revealed that treatment with the hUC-MSC-sec significantly increased the proportion of activated primordial follicles in mouse ovaries both in vitro and in vivo. The activating effects of hUC-MSC-sec on primordial follicles were attributed to the activation of the PI3K-AKT signaling pathway by hepatocyte growth factor (HGF). While the effect of the hUC-MSC-sec was attenuated by the neutralizing antibodies against HGF, application of exogenous HGF alone also promoted the activation of primordial follicles. Furthermore, we demonstrated that HGF promoted the expression of KITL in granulosa cells by binding with the HGF receptor c-Met, thereby increasing the activity of the PI3K-AKT signaling pathway to activate primordial follicles. Taken together, our findings demonstrate that hUC-MSC-sec promotes primordial follicle activation through the functional component HGF to increase the PI3K-AKT signaling activity, highlighting the application of the hUC-MSC-sec or HGF for the treatment of POI patients.
Collapse
Affiliation(s)
- Xin Mi
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Wenlin Jiao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yajuan Yang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yingying Qin
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China.,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China.,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, 200135, China.,Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200135, China
| | - Shidou Zhao
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, China. .,Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Key Laboratory of Reproductive Medicine, Jinan, 250012, Shandong, China. .,Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China. .,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
30
|
Xu J, Zelinski MB. Oocyte quality following in vitro follicle development†. Biol Reprod 2021; 106:291-315. [PMID: 34962509 PMCID: PMC9004734 DOI: 10.1093/biolre/ioab242] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/15/2021] [Accepted: 12/24/2021] [Indexed: 12/30/2022] Open
Abstract
In vitro follicle development (IVFD) is an adequate model to obtain basic knowledge of folliculogenesis and provides a tool for ovarian toxicity screening. IVFD yielding competent oocytes may also offer an option for fertility and species preservation. To promote follicle growth and oocyte maturation in vitro, various culture systems are utilized for IVFD in rodents, domestic animals, wild animals, nonhuman primates, and humans. Follicle culture conditions have been improved by optimizing gonadotropin levels, regulatory factors, nutrient supplements, oxygen concentration, and culture matrices. This review summarizes quality assessment of oocytes generated from in vitro-developed antral follicles from the preantral stage, including oocyte epigenetic and genetic profile, cytoplasmic and nuclear maturation, preimplantation embryonic development following in vitro fertilization, as well as pregnancy and live offspring after embryo transfer. The limitations of oocyte quality evaluation following IVFD and the gaps in our knowledge of IVFD to support proper oocyte development are also discussed. The information may advance our understanding of the requirements for IVFD, with a goal of producing competent oocytes with genetic integrity to sustain embryonic development resulting in healthy offspring.
Collapse
Affiliation(s)
- Jing Xu
- Correspondence: Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006, USA. Tel: +1 5033465411; Fax: +1 5033465585; E-mail:
| | - Mary B Zelinski
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA,Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
31
|
Learning curve of surgeons performing laparoscopic ovarian tissue transplantation in women with premature ovarian insufficiency: A statistical process control analysis. J Minim Invasive Gynecol 2021; 29:559-566. [PMID: 34958952 DOI: 10.1016/j.jmig.2021.12.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 12/15/2021] [Accepted: 12/21/2021] [Indexed: 11/22/2022]
Abstract
STUDY OBJECTIVE To analyze patient safety in laparoscopic ovarian tissue transplantation surgery by tracking the rate of postoperative complications and the learning curves of the surgeons by statistical process control (SPC) analysis. DESIGN A retrospective study. SETTING A university-affiliated hospital. PATIENTS One hundred patients with premature ovarian insufficiency (POI) who underwent ovarian tissue cryopreservation by vitrification and then autologous transplantation of frozen-thawed ovarian tissues with in vitro activation (IVA). INTERVENTION Ovarian tissue cryopreservation, in vitro activation and transplantation. MEASUREMENTS AND MAIN RESULTS We assessed the surgery complications, differences in total surgery time, transplantation time, and transplantation time per ovarian sheet in operations performed by 3 experienced laparoscopic surgeons. Surgeon A performed 80 operations; surgeon B, 29 operations; and surgeon C, 20 operations. Complications occurred in 1.55% of the procedures. While all three surgeons' performance never fell below the unacceptable failure limit, only surgeon A became competent after 66 cases. CONCLUSION The laparoscopic ovarian tissue transplantation surgery was generally safe as the postoperative complications were infrequent (1.55%). Although the performance of all 3 surgeons was acceptable, only surgeon A attained the level of competency after 66 cases. The transplantation method may not be the key factor for reducing surgery time in this surgery. An efficient OTT team is more important in reducing the surgery time than the surgeon's surgical technique alone.
Collapse
|
32
|
Abstract
The reproductive lifespan of female mammals is limited and ultimately depends on the production of a sufficient number of high quality oocytes from a pool of non-growing primordial follicles that are set aside during embryonic and perinatal development. Recent studies show multiple signaling pathways are responsible for maintaining primordial follicle arrest and regulation of activation. Identification of these pathways and their regulatory mechanisms is essential for developing novel treatments for female infertility, improving existing in vitro fertilization techniques, and more recently, restoring the function of cryopreserved ovarian tissue. This review focuses on recent developments in transforming growth factor beta (TGFβ) family signaling in ovarian follicle development and its potential application to therapeutic design. Mouse models have been an essential tool for discovering genes critical for fertility, and recent advancements in human organ culture have additionally allowed for the translation of murine discoveries into human research and clinical settings.
Collapse
|
33
|
Peek R, Eijkenboom LL, Braat DDM, Beerendonk CCM. Complete Purging of Ewing Sarcoma Metastases from Human Ovarian Cortex Tissue Fragments by Inhibiting the mTORC1 Signaling Pathway. J Clin Med 2021; 10:jcm10194362. [PMID: 34640378 PMCID: PMC8509560 DOI: 10.3390/jcm10194362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 11/24/2022] Open
Abstract
Restoration of fertility by autologous transplantation of ovarian cortex tissue in former cancer patients may lead to the reintroduction of malignancy via the graft. Pharmacological ex vivo purging of ovarian cortex fragments prior to autotransplantation may reduce the risk of reseeding the cancer. In this study we have investigated the capacity of Everolimus (EVE), an inhibitor of the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, to eradicate Ewing’s sarcoma (ES) from ovarian tissue by a short-term ex vivo treatment. Exposure of experimentally induced ES tumor foci in ovarian tissue to EVE for 24 h completely eliminated the malignant cells without detrimental effects on follicle morphology, survival or early folliculogenesis. This indicates that effective purging of ovarian cortex tissue from contaminating ES tumor foci is possible by short-term exposure to EVE.
Collapse
|
34
|
Terren C, Nisolle M, Munaut C. Pharmacological inhibition of the PI3K/PTEN/Akt and mTOR signalling pathways limits follicle activation induced by ovarian cryopreservation and in vitro culture. J Ovarian Res 2021; 14:95. [PMID: 34275490 PMCID: PMC8287691 DOI: 10.1186/s13048-021-00846-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 07/06/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cryopreservation and transplantation of ovarian tissue (OTCTP) represent a promising fertility preservation technique for prepubertal patients or for patients requiring urgent oncological management. However, a major obstacle of this technique is follicle loss due to, among others, accelerated recruitment of primordial follicles during the transplantation process, leading to follicular reserve loss in the graft and thereby potentially reducing its lifespan. This study aimed to assess how cryopreservation itself impacts follicle activation. RESULTS Western blot analysis of the PI3K/PTEN/Akt and mTOR signalling pathways showed that they were activated in mature or juvenile slow-frozen murine ovaries compared to control fresh ovaries. The use of pharmacological inhibitors of follicle signalling pathways during the cryopreservation process decreased cryopreservation-induced follicle recruitment. The second aim of this study was to use in vitro organotypic culture of cryopreserved ovaries and to test pharmacological inhibitors of the PI3K/PTEN/Akt and mTOR pathways. In vitro organotypic culture-induced activation of the PI3K/PTEN/Akt pathway is counteracted by cryopreservation with rapamycin and in vitro culture in the presence of LY294002. These results were confirmed by follicle density quantifications. Indeed, follicle development is affected by in vitro organotypic culture, and PI3K/PTEN/Akt and mTOR pharmacological inhibitors preserve primordial follicle reserve. CONCLUSIONS Our findings support the hypothesis that inhibitors of mTOR and PI3K might be an attractive tool to delay primordial follicle activation induced by cryopreservation and culture, thus preserving the ovarian reserve while retaining follicles in a functionally integrated state.
Collapse
Affiliation(s)
- Carmen Terren
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Tour de Pathologie (B23), Site Sart-Tilman, Building 23/4, Avenue Hippocrate, 13, 4000, Liege, Belgium
| | - Michelle Nisolle
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Tour de Pathologie (B23), Site Sart-Tilman, Building 23/4, Avenue Hippocrate, 13, 4000, Liege, Belgium.,Department of Obstetrics and Gynecology, Hôpital de La Citadelle, University of Liège, B-4000, Liège, Belgium
| | - Carine Munaut
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Tour de Pathologie (B23), Site Sart-Tilman, Building 23/4, Avenue Hippocrate, 13, 4000, Liege, Belgium.
| |
Collapse
|
35
|
Transplantation of cryopreserved ovarian tissue in a series of 285 women: a review of five leading European centers. Fertil Steril 2021; 115:1102-1115. [DOI: 10.1016/j.fertnstert.2021.03.008] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/19/2021] [Accepted: 03/04/2021] [Indexed: 01/18/2023]
|