1
|
Wei X, Bjarkadottir BD, Nadjaja D, Sheikh S, Fatum M, Lane S, Williams SA. Effect of AMH on primordial follicle populations in mouse ovaries and human pre-pubertal ovarian xenografts during doxorubicin treatment. Front Cell Dev Biol 2024; 12:1449156. [PMID: 39258229 PMCID: PMC11383774 DOI: 10.3389/fcell.2024.1449156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/15/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction Survival rates of the childhood cancer patients are improving, however cancer treatments such as chemotherapy may lead to infertility due to loss of the primordial follicle (PMF) reserve. Doxorubicin (DXR) is a gonadotoxic chemotherapy agent commonly used in childhood cancers. Anti-Müllerian Hormone (AMH) has been reported to have a protective effect on the mouse ovarian reserve against DXR in vivo. However, whether AMH can prevent PMF loss in conjunction with DXR in human ovarian tissue in vivo has not been determined. Methods In order to investigate this, we first established an optimum dose of DXR that induced PMF loss in cultured mouse ovaries and investigated the efficacy of AMH on reducing DXR-induced PMF loss in mice in vitro. Second, we investigated the effects of DXR on pre-pubertal human ovarian tissue and the ability of AMH to prevent DXR-induced damage comparing using a mouse xenograft model with different transplantation sites. Results Mouse ovaries treated with DXR in vitro and in vivo had reduced PMF populations and damaged follicle health. We did not observe effect of DXR-induced PMF loss or damage to follicle/stromal health in human ovarian cortex, this might have been due to an insufficient dose or duration of DXR. Although AMH does not prevent DXR-induced PMF loss in pre-pubertal and adult mouse ovaries, in mouse ovaries treated with higher concentration of AMH in vitro, DXR did not cause a significant loss in PMFs. This is the first study to illustrate an effect of AMH on DXR-induced PMF loss on pre-pubertal mouse ovaries. However, more experiments with higher doses of AMH and larger sample size are needed to confirm this finding. Discussion We did not observe that AMH could prevent DXR-induced PMF loss in mouse ovaries in vivo. Further studies are warranted to investigate whether AMH has a protective effect against DXR in xenotransplanted human ovarian tissue. Thus, to obtain robust evidence about the potential of AMH in fertility preservation during chemotherapy treatment, alternative AMH administration strategies need to be explored alongside DXR administration to fully interrogate the effect of DXR and AMH on human xenografted tissues.
Collapse
Affiliation(s)
- Xi Wei
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Briet D Bjarkadottir
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Devi Nadjaja
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Sairah Sheikh
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Muhammad Fatum
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
- Oxford Fertility, Institute of Reproductive Sciences, Oxford, United Kingdom
| | - Sheila Lane
- Department of Paediatric Oncology and Haematology, Children's Hospital Oxford, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Suzannah A Williams
- Nuffield Department of Women's and Reproductive Health, Women's Centre, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
2
|
Sun X, Xia R, Wang Y, Wang F, Liu Z, Xue G, Zhang G. Neuromedin S regulates goat ovarian granulosa cell proliferation and steroidogenesis via endoplasmic reticulum Ca 2+-YAP1-ATF4-c-Jun pathway. J Cell Physiol 2024:e31368. [PMID: 38982727 DOI: 10.1002/jcp.31368] [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: 12/22/2023] [Revised: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
Neuromedin S (NMS) plays key roles in reproductive regulation, while its function and mechanism in follicular development remain unclear. The current study aims to investigate the specific role and mechanisms of NMS and its receptors in regulating the proliferation and steroidogenesis of ovarian granulosa cells (GCs). Phenotypically, a certain concentration of NMS addition promoted the proliferation and estrogen production of goat GCs, accompanied by an increase in the G1/S cell population and upregulation of the expression levels of cyclin D1, cyclin dependent kinase 6, steroidogenic acute regulatory protein, cytochrome P450, family 11, subfamily A, polypeptide 1, 3beta-hydroxysteroid dehydrogenase, and cytochrome P450, family 11, subfamily A, polypeptide 1, while the effects of NMS treatment were effectively hindered by knockdown of neuromedin U receptor type 2 (NMUR2). Mechanistically, activation of NMUR2 with NMS maintained endoplasmic reticulum (ER) calcium (Ca2+) homeostasis by triggering the PLCG1-IP3R pathway, which helped preserve ER morphology, sustained an appropriate level of endoplasmic reticulum unfolded protein response (UPRer), and suppressed the nuclear translocation of activating transcription factor 4. Moreover, NMS maintained intracellular Ca2+ homeostasis to activate the calmodulin 1-large tumor suppressor kinase 1 pathway, ultimately orchestrating the regulation of goat GC proliferation and estrogen production through the Yes1 associated transcriptional regulator-ATF4-c-Jun pathway. Crucially, the effects of NMS were mitigated by concurrent knockdown of the NMUR2 gene. Collectively, these data suggest that activation of NMUR2 by NMS enhances cell proliferation and estrogen production in goat GCs through modulating the ER and intracellular Ca2+ homeostasis, leading to activation of the YAP1-ATF4-c-Jun pathway. These findings offer valuable insights into the regulatory mechanisms involved in follicular growth and development, providing a novel perspective for future research.
Collapse
Affiliation(s)
- Xuan Sun
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Rongxin Xia
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yifei Wang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Feng Wang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Zhipeng Liu
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
| | - Gang Xue
- Animal Husbandry and Veterinary Station of Haimen District, Nantong City, China
| | - Guomin Zhang
- Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, China
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
3
|
Kashi O, Meirow D. Overactivation or Apoptosis: Which Mechanisms Affect Chemotherapy-Induced Ovarian Reserve Depletion? Int J Mol Sci 2023; 24:16291. [PMID: 38003481 PMCID: PMC10671775 DOI: 10.3390/ijms242216291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Dormant primordial follicles (PMF), which constitute the ovarian reserve, are recruited continuously into the cohort of growing follicles in the ovary throughout female reproductive life. Gonadotoxic chemotherapy was shown to diminish the ovarian reserve pool, to destroy growing follicle population, and to cause premature ovarian insufficiency (POI). Three primary mechanisms have been proposed to account for this chemotherapy-induced PMF depletion: either indirectly via over-recruitment of PMF, by stromal damage, or through direct toxicity effects on PMF. Preventative pharmacological agents intervening in these ovotoxic mechanisms may be ideal candidates for fertility preservation (FP). This manuscript reviews the mechanisms that disrupt follicle dormancy causing depletion of the ovarian reserve. It describes the most widely studied experimental inhibitors that have been deployed in attempts to counteract these affects and prevent follicle depletion.
Collapse
Affiliation(s)
- Oren Kashi
- The Morris Kahn Fertility Preservation Center, Sheba Medical Center, Ramat Gan 5262000, Israel;
| | - Dror Meirow
- The Morris Kahn Fertility Preservation Center, Sheba Medical Center, Ramat Gan 5262000, Israel;
- Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
4
|
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
|
5
|
Tian Y, Liu X, Pei X, Gao H, Pan P, Yang Y. Mechanism of Mitochondrial Homeostasis Controlling Ovarian Physiology. Endocrinology 2022; 164:6828017. [PMID: 36378567 DOI: 10.1210/endocr/bqac189] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Indexed: 11/17/2022]
Abstract
Ovarian cells, including oocytes, granulosa/cumulus cells, theca cells, and stromal cells, contain abundant mitochondria, which play indispensable roles in the processes of ovarian follicle development. Ovarian function is closely controlled by mitochondrial proteostasis and mitostasis. While mitochondrial proteostasis and mitostasis are disturbed by several factors, leading to dysfunction of ovarian function and initiating the mitochondrial unfolded protein response (UPRmt) and mitophagy to maintain or recover ovarian function and mitochondrial function, clear interactions between the 2 pathways in the ovary have not been fully elucidated. Here, we comprehensively summarize the molecular networks or regulatory mechanisms behind further mitochondrial research in the ovary. This review provides novel insights into the interactions between the UPRmt and mitophagy in ovarian functions.
Collapse
Affiliation(s)
- Yuan Tian
- Clinical Medical College, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xinrui Liu
- Clinical Medical College, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Xiuying Pei
- Clinical Medical College, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Hui Gao
- Clinical Medical College, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Pengge Pan
- Clinical Medical College, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| | - Yanzhou Yang
- Clinical Medical College, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
6
|
Barberino RS, Macedo TJS, Lins TLBG, Menezes VG, Silva RLS, Monte APO, Palheta RC, Smitz JEJ, Matos MHT. Immunolocalization of melatonin receptor type 1 in the sheep ovary and involvement of the PI3K/Akt/FOXO3a signaling pathway in the effects of melatonin on survival and in vitro activation of primordial follicles. Mol Reprod Dev 2022; 89:485-497. [PMID: 35943024 DOI: 10.1002/mrd.23639] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/05/2022] [Accepted: 07/25/2022] [Indexed: 11/10/2022]
Abstract
This study characterized the expression of melatonin receptor type 1 (MT1 ) protein in sheep ovaries, evaluated melatonin effects on primordial follicle survival and development after in vitro culture of ovarian tissue and verified the possible involvement of the phosphatidylinositol-3-kinase/protein kinase B/forkhead box O3a (PI3K/Akt/FOXO3a) pathway in the melatonin actions. Ovine ovarian fragments were cultured in α-modified minimum essential medium alone (α-MEM+ ) or supplemented with 100, 500, or 1000 pg/ml melatonin for 7 days. PI3K inhibition was performed through pretreatment of ovarian fragments with LY294002. Thereafter, immunohistochemistry was performed to evaluate the expression of cleaved caspase-3, Akt, phosphorylated-Akt, and phosphorylated-FOXO3a (p-FOXO3a). The immunohistochemical localization of the MT1 receptor protein was documented in sheep preantral and antral follicles. After in vitro culture, 100 pg/ml melatonin showed higher follicular survival and activation than α-MEM+ and other melatonin concentrations. After PI3K inhibition, there was an increase in cleaved caspase-3-positive follicles, and a decrease in the primordial follicle activation, Akt phosphorylation, and nuclear exclusion of p-FOXO3a. In conclusion, MT1 receptor protein is present in the sheep ovary. Furthermore, 100 pg/ml melatonin maintains survival and stimulates activation of primordial follicles through the PI3K/Akt/FOXO3a signaling pathway after in vitro culture of sheep ovarian tissue.
Collapse
Affiliation(s)
- Ricássio S Barberino
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Taís J S Macedo
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Thae Lanne B G Lins
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Vanúzia G Menezes
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Regina L S Silva
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Alane P O Monte
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Raimundo C Palheta
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| | - Johan E J Smitz
- Follicle Biology Laboratory, Center for Reproductive Medicine, Free University Brussels - VUB, Brussels, Belgium
| | - Maria Helena T Matos
- Nucleus of Biotechnology Applied to Ovarian Follicle Development, Department of Veterinary Medicine, Federal University of São Francisco Valley - UNIVASF, Petrolina, Brazil
| |
Collapse
|
7
|
LIF and bFGF enhanced chicken primordial follicle activation by Wnt/β-catenin pathway. Theriogenology 2021; 176:1-11. [PMID: 34555602 DOI: 10.1016/j.theriogenology.2021.09.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022]
Abstract
The cytokines leukemia inhibitory factor (LIF) and basic fibroblast growth factor (bFGF) are closely related to the development of primordial follicles. In this study, the functions and correlation of LIF and bFGF in the development of chicken primordial follicles were examined, along with the signaling pathways including protein kinase B (AKT), extracellular regulated protein kinase (ERK) and Wnt/β-catenin signaling pathways. Ovarian tissues were collected from four-day-old chicks and incubated with LIF and bFGF alone or in combination for three days to observe the changes in follicular development. Results showed that there was a time-dependent correlation between the changes in expression of LIF/its receptor (LIFR) and the developmental process of primordial follicles. LIF and bFGF exerted a synergistic effect on the activation of primordial follicles. However, SC144 (an antagonist of LIFR) inhibited this stimulating action. The effect by LIF and bFGF were shown to operate at AKT and ERK signaling pathways to suppress cell apoptosis and promote proliferation (P < 0.05) via the Wnt/β-catenin signaling (P < 0.05). In conclusion, local cytokines LIF and bFGF functioned to enhance the activation of chicken primordial follicles by increasing cell proliferation and decreasing apoptosis in the ovary involving AKT, ERK and Wnt/β-catenin signaling.
Collapse
|
8
|
Relav L, Estienne A, Price CA. Dual-specificity phosphatase 6 (DUSP6) mRNA and protein abundance is regulated by fibroblast growth factor 2 in sheep granulosa cells and inhibits c-Jun N-terminal kinase (MAPK8) phosphorylation. Mol Cell Endocrinol 2021; 531:111297. [PMID: 33964319 DOI: 10.1016/j.mce.2021.111297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 11/20/2022]
Abstract
Growth factors regulate ovarian follicle development and they signal through intracellular pathways including mitogen-activated protein kinase (MAPK) phosphorylation, which is negatively regulated by a subfamily of 23 dual-specificity phosphatases (DUSP). Using sheep granulosa cells as a model, we detected mRNA encoding 16 DUSPs in vivo and in vitro. Stimulation of cells in vitro with FGF2 increased (p < 0.05) abundance of DUSP1, DUSP2, DUSP5 and DUSP6 mRNA, and abundance of DUSP1 and DUSP6 proteins (p < 0.05). In contrast, neither FGF8b nor FGF18 had any major effect on DUSP mRNA abundance. Inhibition of DUSP6 action with the inhibitor BCI significantly increased (p < 0.05) MAPK8 (JNK) phosphorylation but not phosphoMAPK14 (p38) or MAPK3/1 (ERK1/2) abundance. This study suggests that FGFs stimulate DUSP protein abundance, that DUSP6 regulates MAPK8 phosphorylation in granulosa cells, and DUSPs are involved in the differential MAPK signaling of individual FGF ligands.
Collapse
Affiliation(s)
- Lauriane Relav
- Centre de recherche en reproduction animale, Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, J2S 7C6, QC, Canada
| | - Anthony Estienne
- Centre de recherche en reproduction animale, Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, J2S 7C6, QC, Canada
| | - Christopher A Price
- Centre de recherche en reproduction animale, Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, J2S 7C6, QC, Canada.
| |
Collapse
|
9
|
Vo KCT, Kawamura K. In Vitro Activation Early Follicles: From the Basic Science to the Clinical Perspectives. Int J Mol Sci 2021; 22:ijms22073785. [PMID: 33917468 PMCID: PMC8038686 DOI: 10.3390/ijms22073785] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/29/2021] [Accepted: 04/03/2021] [Indexed: 12/16/2022] Open
Abstract
Development of early follicles, especially the activation of primordial follicles, is strictly modulated by a network of signaling pathways. Recent advance in ovarian physiology has been allowed the development of several therapies to improve reproductive outcomes by manipulating early folliculogenesis. Among these, in vitro activation (IVA) has been recently developed to extend the possibility of achieving genetically related offspring for patients with premature ovarian insufficiency and ovarian dysfunction. This method was established based on basic science studies of the intraovarian signaling pathways: the phosphoinositide 3-kinase (PI3K)/Akt and the Hippo signaling pathways. These two pathways were found to play crucial roles in folliculogenesis from the primordial follicle to the early antral follicle. Following the results of rodent experiments, IVA was implemented in clinical practice. There have been multiple recorded live births and ongoing pregnancies. Further investigations are essential to confirm the efficacy and safety of IVA before used widely in clinics. This review aimed to summarize the published literature on IVA and provide future perspectives for its improvement.
Collapse
|
10
|
Monte APO, Bezerra MÉS, Menezes VG, Gouveia BB, Barberino RS, Lins TLBG, Barros VRP, Santos JMS, Donfack NJ, Matos MHT. Involvement of Phosphorylated Akt and FOXO3a in the Effects of Growth and Differentiation Factor-9 (GDF-9) on Inhibition of Follicular Apoptosis and Induction of Granulosa Cell Proliferation After In Vitro Culture of Sheep Ovarian Tissue. Reprod Sci 2021; 28:2174-2185. [DOI: 10.1007/s43032-020-00409-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 11/18/2020] [Indexed: 12/18/2022]
|
11
|
Grosbois J, Devos M, Demeestere I. Implications of Nonphysiological Ovarian Primordial Follicle Activation for Fertility Preservation. Endocr Rev 2020; 41:5882019. [PMID: 32761180 DOI: 10.1210/endrev/bnaa020] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/31/2020] [Indexed: 02/06/2023]
Abstract
In recent years, ovarian tissue cryopreservation has rapidly developed as a successful method for preserving the fertility of girls and young women with cancer or benign conditions requiring gonadotoxic therapy, and is now becoming widely recognized as an effective alternative to oocyte and embryo freezing when not feasible. Primordial follicles are the most abundant population of follicles in the ovary, and their relatively quiescent metabolism makes them more resistant to cryoinjury. This dormant pool represents a key target for fertility preservation strategies as a resource for generating high-quality oocytes. However, development of mature, competent oocytes derived from primordial follicles is challenging, particularly in larger mammals. One of the main barriers is the substantial knowledge gap regarding the regulation of the balance between dormancy and activation of primordial follicles to initiate their growing phase. In addition, experimental and clinical factors also affect dormant follicle demise, while the mechanisms involved remain largely to be elucidated. Moreover, most of our basic knowledge of these processes comes from rodent studies and should be extrapolated to humans with caution, considering the differences between species in the reproductive field. Overcoming these obstacles is essential to improving both the quantity and the quality of mature oocytes available for further fertilization, and may have valuable biological and clinical applications, especially in fertility preservation procedures. This review provides an update on current knowledge of mammalian primordial follicle activation under both physiological and nonphysiological conditions, and discusses implications for fertility preservation and priorities for future research.
Collapse
Affiliation(s)
- Johanne Grosbois
- Research Laboratory in Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium.,Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Melody Devos
- Research Laboratory in Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium
| | - Isabelle Demeestere
- Research Laboratory in Human Reproduction, Université Libre de Bruxelles, Brussels, Belgium.,Obstetrics and Gynecology Department, Erasme Hospital, Brussels, Belgium
| |
Collapse
|
12
|
Molecular Basis Associated with the Control of Primordial Follicle Activation During Transplantation of Cryopreserved Ovarian Tissue. Reprod Sci 2020; 28:1257-1266. [DOI: 10.1007/s43032-020-00318-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 09/14/2020] [Indexed: 12/18/2022]
|
13
|
Yang Q, Zhu L, Jin L. Human Follicle in vitro Culture Including Activation, Growth, and Maturation: A Review of Research Progress. Front Endocrinol (Lausanne) 2020; 11:548. [PMID: 32849312 PMCID: PMC7431469 DOI: 10.3389/fendo.2020.00548] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/06/2020] [Indexed: 01/23/2023] Open
Abstract
Fertility preservation has received unprecedented attention nowadays. In addition to cryopreservation and re-implantation of embryos, oocytes, and ovarian tissue pieces, in vitro culture system for follicles/oocytes has been considered as an alternative strategy for fertility preservation. Since the metabolic dynamics and required nutrients are not entirely the same in different stages of follicular development, optimization of each culture step is needed. In this paper, literature regarding culture conditions in three steps were analyzed. Known additives in activation stage included 740Y-P, bpV(HOpic), follicle stimulating hormone (FSH), human serum albumin (HSA), ITS, growth differentiation factor 9 (GDF9), bone morphogenetic protein 15 (BMP15), and cyclic adenosine monophosphate (cAMP), with different degrees of activation promotion and potential detrimental effect on DNA integrity. For isolated follicles growth stage, actin A, FSH, basic fibroblast growth factor (bFGF), estradiol were proved to improve development or proliferation. As for maturation, addition of growth hormone, melatonin, C-type natriuretic peptide (CNP), GDF9, cilostamide, or forskolin helped to regulate maturation rate or improve oocyte quality. Based on previous sequential culture system for human follicles, optimization is needed to achieve higher maturation rate and better oocyte quality, pursuant to current review, which demonstrated the effects of various additives on different stages.
Collapse
|
14
|
Locatelli Y, Calais L, Duffard N, Lardic L, Monniaux D, Piver P, Mermillod P, Bertoldo MJ. In vitro survival of follicles in prepubertal ewe ovarian cortex cryopreserved by slow freezing or non-equilibrium vitrification. J Assist Reprod Genet 2019; 36:1823-1835. [PMID: 31376104 PMCID: PMC6731053 DOI: 10.1007/s10815-019-01532-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/09/2019] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Vitrification is a well-accepted fertility preservation procedure for cryopreservation of oocytes and embryos but little is known regarding ovarian tissue, for which slow freezing is the current convention. The aim of the present study was to assess the efficiency of non-equilibrium vitrification compared to conventional slow freezing for ovarian cortex cryopreservation. METHODS Using prepubertal sheep ovaries, the capacity of the tissue to sustain folliculogenesis following cryopreservation and in vitro culture was evaluated. Ovarian cortex fragments were cultured in wells for 9 days, immediately or after cryopreservation by conventional slow freezing or non-equilibrium vitrification in straws. During culture, follicular populations within cortex were evaluated by histology and immunohistochemistry for PCNA and TUNEL. Steroidogenic activity of the tissue was monitored by assay for progesterone and estradiol in spent media. RESULTS No significant differences in follicle morphology, PCNA, or TUNEL labeling were observed between cryopreservation methods at the initiation of culture. Similar decreases in the proportion of primordial follicle population, and increases in the proportion of growing follicles, were observed following culture of fresh or cryopreserved ovarian tissue regardless of cryopreservation method. At the end of culture, PCNA and TUNEL-positive follicles were not statistically altered by slow freezing or vitrification in comparison to fresh cultured fragments. CONCLUSIONS Overall, for both cryopreservation methods, the cryopreserved tissue showed equal capacity to fresh tissue for supporting basal folliculogenesis in vitro. Taken together, these data confirm that both non-equilibrium vitrification and slow-freezing methods are both efficient for the cryopreservation of sheep ovarian cortex fragments.
Collapse
Affiliation(s)
- Yann Locatelli
- DMJZ, Muséum National d'Histoire Naturelle, Laboratoire de la Réserve, Zoologique de la Haute Touche, Obterre, France.
- INRA UMR Physiologie de la Reproduction et des Comportements, INRA, Nouzilly, France.
| | - L Calais
- INRA UMR Physiologie de la Reproduction et des Comportements, INRA, Nouzilly, France
| | - N Duffard
- DMJZ, Muséum National d'Histoire Naturelle, Laboratoire de la Réserve, Zoologique de la Haute Touche, Obterre, France
| | - L Lardic
- INRA UMR Physiologie de la Reproduction et des Comportements, INRA, Nouzilly, France
| | - D Monniaux
- INRA UMR Physiologie de la Reproduction et des Comportements, INRA, Nouzilly, France
| | - P Piver
- Service de gynécologie-obstétrique, hôpital Mère-Enfant, CHU de Limoges, Limoges, France
| | - P Mermillod
- INRA UMR Physiologie de la Reproduction et des Comportements, INRA, Nouzilly, France
| | - M J Bertoldo
- INRA UMR Physiologie de la Reproduction et des Comportements, INRA, Nouzilly, France
- Fertility and Research Centre, School of Women's and Children's Health, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
- School of Medical Sciences, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| |
Collapse
|
15
|
Adib S, Valojerdi MR, Alikhani M. Dose optimisation of PTEN inhibitor, bpV (HOpic), and SCF for the in-vitro activation of sheep primordial follicles. Growth Factors 2019; 37:178-189. [PMID: 31646909 DOI: 10.1080/08977194.2019.1680661] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The in-vitro development of primordial follicles is critical for improving mammalian fertility and wildlife conservation. This study aimed to optimise the effective doses of bpV (HOpic) and stem cell factor (SCF) for the in-vitro activation of sheep primordial follicles. To do this, sheep ovarian cortex was treated with bpV (1.5, 15, and 150 μM) and SCF (50 and 100 ng/ml). Follicular count indicated that 15 μM bpV and 100 ng/ml SCF significantly increased normal primary follicles compared to other groups (p < 0.05). Also, a significant downregulation of P53 and PTEN, as well as the increased expression of PI3K was observed. The in-vitro maturation was more pronounced when the fragmented tissues were co-treated with selected doses of bpV and SCF. In conclusion, the combination of 15 μM bpV and 100 ng/ml SCF was the most effective treatment strategy for the activation and survival of primordial follicles in sheep ovarian fragments.
Collapse
Affiliation(s)
- Samane Adib
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Embryology at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mojtaba Rezazadeh Valojerdi
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Embryology at Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mehdi Alikhani
- Department of Molecular Systems Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
16
|
Xiong J, Wu M, Zhang Q, Zhang C, Xiong G, Ma L, Lu Z, Wang S. Proteomic analysis of mouse ovaries during the prepubertal stages. Exp Cell Res 2019; 377:36-46. [PMID: 30797753 DOI: 10.1016/j.yexcr.2019.02.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/16/2019] [Accepted: 02/18/2019] [Indexed: 12/01/2022]
Abstract
Postnatal folliculogenesis, primordial follicle activation and follicular development at early stage are important for normal ovarian function and fertility, and a comprehensive understanding of this process under physiological condition is necessary. To observe the regulation and mechanism of ovarian follicle development during the prepubertal stages, we collected the mouse ovaries from three time points, including 1 day, 7 days, and 4 weeks after birth. We then performed a proteomic analysis using tandem mass tags (TMT) labeling combined with a two-dimensional liquid chromatography-tandem mass spectrometry (2D LC-MS/MS) technique. A total of 706 proteins were determined to be significant differential abundance (P-SDA). Sixty upregulated proteins and 12 downregulated proteins that were P-SDA and 3 significant KEGG pathways (P < 0.05) were found at 7 days vs. 1 day after birth, while 237 upregulated proteins, 271 downregulated proteins and 42 significant KEGG pathways were found for 4 weeks vs. 7 days after birth. Some vital genes (Figla, Ooep, Padi6, Zp3, Hsd3b1, cyp11a1), key pathways (ECM-receptor interaction, focal adhesion, ovarian steroidogenesis, complement and coagulation cascades, PI3K/Akt/mTOR), and metabolic regulation (energy metabolism, lipid metabolism, metal ion metabolism) were found to be related to the postnatal folliculogenesis, primordial follicle activation and follicular development. Finally, qRT-PCR and western blotting verified some vital genes and further elucidated the developmental process of follicles, and the results may contribute to the understanding of the formation and activation of primordial follicle and follicular development. Significance: This study offers the first proteomic insights into mechanisms of follicle development under physiological condition during the prepubertal stages. By comparing P-SDA of mouse ovaries during various period of age, our data reveals that the regulation of primordial follicle formation and activation is significantly different from that of follicular development. These findings demonstrate that many unique molecular mechanisms underlie ovarian development could be used for ovarian disease research.
Collapse
Affiliation(s)
- Jiaqiang Xiong
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qinghua Zhang
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chun Zhang
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guoping Xiong
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lingwei Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiyong Lu
- Hubei Key Laboratory of Embryonic Stem Cell Research, Tai-He Hospital, Hubei University of Medicine, Shiyan, Hubei, China,Centre for Reproductive Medicine, Puren Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
17
|
Fabbri R, Zamboni C, Vicenti R, Macciocca M, Paradisi R, Seracchioli R. Update on oogenesis in vitro. ACTA ACUST UNITED AC 2018; 70:588-608. [PMID: 29999288 DOI: 10.23736/s0026-4784.18.04273-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Ovarian tissue is increasingly being collected from cancer patients and cryopreserved for fertility preservation. Alternately to the autologous transplantation, the development of culture systems that support oocyte development from the primordial follicle stage represent a valid strategy to restore fertility. The aim of this study is to review the most recent data regarding oogenesis in vitro and to provide an up-to-date on the contemporary knowledge of follicle growth and development in vitro. EVIDENCE ACQUISITION A comprehensive systematic MEDLINE search was performed since February 2018 for English-language reports by using the following terms: "ovary," "animal and human follicle," "in vitro growth and development," "ovarian tissue culture," "fertility preservation," "IVM," "oocyte." Previous published reviews and recent published original articles were preferred in order to meet our study scope. EVIDENCE SYNTHESIS Over time, many studies have been conducted with the aim to optimize the characteristics of ovarian tissue culture systems and to better support the three main phases: 1) activation of primordial follicles; 2) isolation and culture of growing preantral follicles; 3) removal from the follicle environment and maturation of oocyte cumulus complexes. While complete oocyte in vitro development has been achieved in mouse, with the production of live offspring, the goal of obtaining oocytes of sufficient quality to support embryo development has not been completely reached into higher mammals despite decades of effort. CONCLUSIONS Over the years, many improvements have been made on ovarian tissue cultures with the future purpose that patients will be provided with a greater number of developmentally competent oocytes for fertility preservation.
Collapse
Affiliation(s)
- Raffaella Fabbri
- Unit of Gynecology and Physiopathology of Human Reproduction, Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Chiara Zamboni
- Unit of Gynecology and Physiopathology of Human Reproduction, Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy -
| | - Rossella Vicenti
- Unit of Gynecology and Physiopathology of Human Reproduction, Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Maria Macciocca
- Unit of Gynecology and Physiopathology of Human Reproduction, Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| | | | - Renato Seracchioli
- Unit of Gynecology and Physiopathology of Human Reproduction, Department of Medical and Surgical Sciences, University of Bologna, S. Orsola-Malpighi Hospital, Bologna, Italy
| |
Collapse
|
18
|
In-vitro regulation of primordial follicle activation: challenges for fertility preservation strategies. Reprod Biomed Online 2018; 36:491-499. [PMID: 29503209 DOI: 10.1016/j.rbmo.2018.01.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 01/30/2018] [Accepted: 01/31/2018] [Indexed: 12/20/2022]
Abstract
Ovarian tissue is increasingly being collected from cancer patients and cryopreserved for fertility preservation. While the only available option to restore fertility is autologous transplantation, this treatment is not appropriate for all patients due to the risk of reintroducing cancer cells and causing disease recurrence. Harnessing the full reproductive potential of this tissue to restore fertility requires the development of culture systems that support oocyte development from the primordial follicle stage. While this has been achieved in the mouse, the goal of obtaining oocytes of sufficient quality to support embryo development has not been reached in higher mammals despite decades of effort. In vivo, primordial follicles gradually exit the resting pool, whereas when primordial follicles are placed into culture, global activation of these follicles occurs. Therefore, the addition of a factor(s) that can regulate primordial follicle activation in vitro may be beneficial to the development of culture systems for ovarian tissue from cancer patients. Several factors have been observed to inhibit follicle activation, including anti-Müllerian hormone, stromal-derived factor 1 and members of the c-Jun-N-terminal kinase pathway. This review summarizes the findings from studies of these factors and discusses their potential integration into ovarian tissue culture strategies for fertility preservation.
Collapse
|
19
|
Cadoret V, Frapsauce C, Jarrier P, Maillard V, Bonnet A, Locatelli Y, Royère D, Monniaux D, Guérif F, Monget P. Molecular evidence that follicle development is accelerated in vitro compared to in vivo. Reproduction 2017; 153:493-508. [DOI: 10.1530/rep-16-0627] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/27/2017] [Accepted: 02/02/2017] [Indexed: 12/18/2022]
Abstract
In this study, we systematically compared the morphological, functional and molecular characteristics of granulosa cells and oocytes obtained by a three-dimensional in vitro model of ovine ovarian follicular growth with those of follicles recovered in vivo. Preantral follicles of 200 µm diameter were recovered and cultured up to 950 µm over a 20-day period. Compared with in vivo follicles, the in vitro culture conditions maintained follicle survival, with no difference in the rate of atresia. However, the in vitro conditions induced a slight decrease in oocyte growth rate, delayed antrum formation and increased granulosa cell proliferation rate, accompanied by an increase and decrease in CCND2 and CDKN1A mRNA expression respectively. These changes were associated with advanced granulosa cell differentiation in early antral follicles larger than 400 µm diameter, regardless of the presence or absence of FSH, as indicated by an increase in estradiol secretion, together with decreased AMH secretion and expression, as well as increased expression of GJA1, CYP19A1, ESR1, ESR2, FSHR, INHA, INHBA, INHBB and FST. There was a decrease in the expression of oocyte-specific molecular markers GJA4, KIT, ZP3, WEE2 and BMP15 in vitro compared to that in vivo. Moreover, a higher percentage of the oocytes recovered from cultured follicles 550 to 950 µm in diameter was able to reach the metaphase II meiosis stage. Overall, this in vitro model of ovarian follicle development is characterized by accelerated follicular maturation, associated with improved developmental competence of the oocyte, compared to follicles recovered in vivo.
Collapse
|