1
|
Chen J, He Z, Xu W, Kang Y, Zhu F, Tang H, Wang J, Zhong F. Human umbilical cord mesenchymal stem cells restore chemotherapy-induced premature ovarian failure by inhibiting ferroptosis in vitro ovarian culture system. Reprod Biol Endocrinol 2024; 22:137. [PMID: 39511578 PMCID: PMC11542367 DOI: 10.1186/s12958-024-01310-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have shown potential in repairing chemotherapy-induced premature ovarian failure (POF). However, challenges such as stem cell loss and immune phagocytosis post-transplantation hinder their application. Due to easy and safe handling, in vitro ovarian culture is widely available for drug screening, pathophysiological research, and in vitro fertilization. MSCs could exhibit therapeutic capacity for ovarian injury, and avoid stem cell loss and immune phagocytosis in vitro tissue culture system. Therefore, this study utilizes an in vitro ovarian culture system to investigate the reparative potential of human umbilical cord mesenchymal stem cells (hUCMSCs) and their mechanism. METHODS In this study, a chemotherapy-induced POF model was established by introducing cisplatin in vitro ovarian culture system. The reparative effects of hUCMSCs on damaged ovarian tissue were validated through Transwell chambers. Tissue histology examination, immunohistochemical staining, Western blotting, and RT-PCR were employed to evaluate the expression effects of hUCMSCs on ferroptosis and fibrosis-related genes during the process of repairing cisplatin-induced POF. RESULTS Cisplatin was found to activate ovarian follicles in vitro POF model. Transcriptomic sequencing analysis revealed that cisplatin could activate genes associated with ferroptosis. hUCMSCs alleviated cisplatin-induced POF by suppressing the expression of ferroptosis. Moreover, inhibiting ferroptosis by hUCMSCs also ameliorated ovarian hormone levels and reduced the expression of fibrosis-related factors α-SMA and COL-I in the ovaries. CONCLUSIONS This study confirms that cisplatin-induced ovarian damage via ferroptosis in vitro POF model, and hUCMSCs repair ovarian injury by inhibiting the ferroptosis pathway and suppressing fibrosis. This research contributes to evaluating the effectiveness of hUCMSCs in treating chemotherapy-induced POF by inhibiting ferroptosis in an in vitro ovarian culture system and provides a potential therapeutic strategy for chemotherapy-induced POF.
Collapse
Affiliation(s)
- Jiaqi Chen
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Zhuoying He
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Wenjuan Xu
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China
| | - Yumiao Kang
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Fengyu Zhu
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China
| | - Heng Tang
- Wanbei Coal Electric Group General Hospital, Suzhou, Anhui Province, 234011, China.
| | - Jianye Wang
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China.
| | - Fei Zhong
- Department of Oncology, Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China.
- NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, No 81 Meishan Road, Hefei, Anhui, 230032, China.
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, No 218 Jixi Road, Hefei, Anhui, 230022, China.
| |
Collapse
|
2
|
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: 57] [Impact Index Per Article: 28.5] [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
|
3
|
Giovannopoulou E, Karakasi MV, Kouroupi M, Giatromanolaki A, Tsikouras P, Pavlidis P. Safety and efficacy of ovarian tissue autotransplantation: A systematic literature review. Folia Med (Plovdiv) 2023; 65:362-370. [PMID: 38351810 DOI: 10.3897/folmed.65.e89198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/30/2022] [Indexed: 02/16/2024] Open
Abstract
Ovarian tissue autotransplantation is an innovative fertility preservation technique that has provoked ongoing investigations. The purpose of the present study was to assess the safety and reproductive performance of ovarian tissue autotransplantation. This review is conducted according to PRISMA guidelines. Seven studies met the inclusion criteria. A total of 3427 patients underwent ovarian tissue cryopreservation and 205 received an autotransplantation. Tissue retrieval was mainly performed by laparoscopy and only one major complication occurred. Transplantations were predominantly performed by open procedures and data on safety were insufficient. A total of 295 autotransplantations were analyzed, resulting in 104 pregnancies. Sixty-five pregnancies led to live births, while nine were ongoing at that time. A pregnancy rate (PR) of 50.7% and a live-birth rate (LBR) of 32.7% were observed. Natural conception accounted for 46.3% of live births. No birth deficits were recorded. Ovarian tissue autotransplantation seems to be a safe procedure with acceptable pregnancy rates.
Collapse
Affiliation(s)
| | | | - Maria Kouroupi
- General University Hospital of Alexandroupolis, Alexandroupolis, Greece
| | | | | | | |
Collapse
|
4
|
Johnson J, Emerson JW, Lawley SD. Recapitulating human ovarian aging using random walks. PeerJ 2022; 10:e13941. [PMID: 36032944 PMCID: PMC9406804 DOI: 10.7717/peerj.13941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/02/2022] [Indexed: 01/19/2023] Open
Abstract
Mechanism(s) that control whether individual human primordial ovarian follicles (PFs) remain dormant, or begin to grow, are all but unknown. One of our groups has recently shown that activation of the Integrated Stress Response (ISR) pathway can slow follicular granulosa cell proliferation by activating cell cycle checkpoints. Those data suggest that the ISR is active and fluctuates according to local conditions in dormant PFs. Because cell cycle entry of (pre)granulosa cells is required for PF growth activation (PFGA), we propose that rare ISR checkpoint resolution allows individual PFs to begin to grow. Fluctuating ISR activity within individual PFs can be described by a random process. In this article, we model ISR activity of individual PFs by one-dimensional random walks (RWs) and monitor the rate at which simulated checkpoint resolution and thus PFGA threshold crossing occurs. We show that the simultaneous recapitulation of (i) the loss of PFs over time within simulated subjects, and (ii) the timing of PF depletion in populations of simulated subjects equivalent to the distribution of the human age of natural menopause can be produced using this approach. In the RW model, the probability that individual PFs grow is influenced by regionally fluctuating conditions, that over time manifests in the known pattern of PFGA. Considered at the level of the ovary, randomness appears to be a key, purposeful feature of human ovarian aging.
Collapse
Affiliation(s)
- Joshua Johnson
- Department of Obstetrics and Gynecology, University of Colorado-Anschutz Medical Center, Aurora, Colorado, United States
| | - John W. Emerson
- Department of Statistics and Data Science, Yale University, New Haven, Connecticut, United States
| | - Sean D. Lawley
- Department of Mathematics, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
5
|
Effects of low-dose X-ray medical diagnostics on female gonads: Insights from large animal oocytes and human ovaries as complementary models. PLoS One 2021; 16:e0253536. [PMID: 34166427 PMCID: PMC8224917 DOI: 10.1371/journal.pone.0253536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 06/08/2021] [Indexed: 11/19/2022] Open
Abstract
Diagnostic imaging has significantly grown over the last thirty years as indispensable support for diagnostic, prognostic, therapeutic and monitoring procedures of human diseases. This study explored the effects of low-dose X-ray medical diagnostics exposure on female fertility. To aim this, cumulus-oocyte complexes (COCs) recovered from the ovaries of juvenile sheep and human ovaries were used as complementary models for in vitro studies. In the sheep model, the effects of low-dose X-rays on oocyte viability and developmental competence were evaluated. In human ovaries originated from two age group (21–25 and 33–36 years old) subjects with gender dysphoria, X-rays effects on tissue morphology, follicular density and expression of apoptosis-related (NOXA, PUMA, Bcl2, Bak, γH2AX) and cell cycle-related genes (p21 and ki67) were investigated. It was noted that in sheep, the minimum dose of 10 mGy did not influence most of examined parameters at oocyte and embryo levels, whereas 50 and 100 mGy X-ray exposure reduced oocyte bioenergetic/oxidative activity but without any visible effects on oocyte and embryo development. In addition, blastocyst bioenergetic/oxidative status was reduced with all used doses. Overall data on human ovaries showed that low-dose X-rays, similarly as in sheep, did not alter any of examined parameters. However, in women belonging to the 33–36 year group, significantly reduced follicular density was observed after exposure to 50 and 100 mGy, and increased NOXA and Bax expression after exposure at 50 mGy. In conclusion, used low-doses of X-ray exposure, which resemble doses used in medical diagnostics, produce weak damaging effects on female fertility with increased susceptibility in advanced age.
Collapse
|
6
|
Vallet N, Boissel N, Elefant E, Chevillon F, Pasquer H, Calvo C, Dhedin N, Poirot C. Can Some Anticancer Treatments Preserve the Ovarian Reserve? Oncologist 2021; 26:492-503. [PMID: 33458904 DOI: 10.1002/onco.13675] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Preventing premature ovarian failure (POF) is a major challenge in oncology. With conventional regimens, cytotoxicity-associated POF involves primordial follicles (PF) pool depletion by apoptosis or overactivation mechanisms, notably mediated by the ABL/TAp63 and PI3K/Akt/mTOR pathways. New anticancer treatments have been designed to target pathways implicated in tumor growth. Although concerns regarding fertility arise with these targeted therapies, we hypothesized that targeted therapies may exert off-tumor effects on PF that might delay POF. We provide an overview of evidence concerning these off-tumor effects on PF. Limitations and future potential implications of these findings are discussed. DESIGN PubMed was searched by combining Boolean operators with the following keywords: fertility, ovarian, follicle, anti-tumoral, cancer, targeted, cytotoxic, and chemotherapy. RESULTS Cisplatin-related PF apoptosis via the ABL/TAp63 pathway was targeted with a tyrosine kinase inhibitor, imatinib, in mice, but effects were recently challenged by findings on human ovarian xenografts in mice. In cyclophosphamide-treated mice, PI3K/Akt/mTOR pathway inhibition with mTOR inhibitors and AS101 preserved the PF pool. Proteasome and GSK3 inhibitors were evaluated for direct and indirect follicle DNA damage prevention. Surprisingly, evidence for cytotoxic drug association with PF pool preservation was found. We also describe selected non-anticancer molecules that may minimize gonadotoxicity. CONCLUSION Not all anticancer treatments are associated with POF, particularly since the advent of targeted therapies. The feasibility of associating a protective drug targeting PF exhaustion mechanisms with cytotoxic treatments should be evaluated, as a way of decreasing the need for conventional fertility preservation techniques. Further evaluations are required for transfer into clinical practice. IMPLICATIONS FOR PRACTICE Anticancer therapies are associated with infertility in 10%-70% of patients, which is the result of primordial follicles pool depletion. Alone or associated with gonadotoxic treatments, some targeted therapies may exert favorable off-targets effects on the primordial follicle pool by slowing down their exhaustion. Current evidence of these effects relies on murine models or human in vitro models. Evaluation of these protective strategies in humans is challenging; however, if these results are confirmed with clinical and biological data, it not only could be a new approach to female fertility preservation but also would change standard fertility strategies.
Collapse
Affiliation(s)
- Nicolas Vallet
- Department of Hematology and Cellular Therapy, Tours University Hospital, Tours, France
| | - Nicolas Boissel
- Department of Hematology, Adolescent and Young Adults Unit, Fertility Preservation, Saint Louis Hospital, AP-, HP, Paris, France.,Paris University, Paris, France
| | - Elisabeth Elefant
- Centre de Référence sur les Agents Tératogènes (CRAT), Armand Trousseau Hospital, AP-, HP, Paris, France.,Faculty of Medicine, Sorbonne University, Paris, France
| | - Florian Chevillon
- Department of Hematology, Adolescent and Young Adults Unit, Fertility Preservation, Saint Louis Hospital, AP-, HP, Paris, France
| | - Hélène Pasquer
- Department of Hematology, Adolescent and Young Adults Unit, Fertility Preservation, Saint Louis Hospital, AP-, HP, Paris, France
| | - Charlotte Calvo
- Pediatric Hematology Department, Robert Debré Hospital, AP-, HP, Paris, France
| | - Nathalie Dhedin
- Department of Hematology, Adolescent and Young Adults Unit, Fertility Preservation, Saint Louis Hospital, AP-, HP, Paris, France
| | - Catherine Poirot
- Department of Hematology, Adolescent and Young Adults Unit, Fertility Preservation, Saint Louis Hospital, AP-, HP, Paris, France.,Faculty of Medicine, Sorbonne University, Paris, France
| |
Collapse
|
7
|
Moura GAD, Monteiro PB. Cytotoxic Activity of Antineoplastic Agents on Fertility: A Systematic Review. REVISTA BRASILEIRA DE GINECOLOGIA E OBSTETRÍCIA 2020; 42:759-768. [PMID: 33254272 PMCID: PMC10309244 DOI: 10.1055/s-0040-1713911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVE To analyze the long-term effects of antineoplastic treatments on patient fertility. SELECTION OF STUDIES The studies were selected through the New PubMed, Scielo and Lilacs databases along with references used for the creation of the present work. For the selection of studies, articles published between the periods from January 1, 2015 to April 6, 2020 in the English, Portuguese and Spanish languages were used. As inclusion criteria: cohort studies and studies conducted in vitro. As exclusion criteria: review articles, reported cases, studies that do not address thematic reproduction, studies that do not address the cancer theme, articles that used animals, articles that address the preservation of fertility and articles in duplicate in the bases. DATA COLLECTION The collected data included: age of the patient at the beginning of treatment, type of neoplasm, type of antineoplastic treatment, chemotherapy used, radiotherapy dosage, radiotherapy site, effect of antineoplastic agents on fertility and number of patients in the study. DATA SYNTHESIS Thirty studies were evaluated, antineoplastic chemotherapy agents and radiotherapy modulate serum hormone levels, reduces germ cell quantities and correlated with an increase in sterility rates. The effects mentioned occur in patients in the prepubertal and postpubertal age. CONCLUSION Antineoplastic treatments have cytotoxic effects on the germ cells leading to hormonal modulation, and pubertal status does not interfere with the cytotoxic action of therapies.
Collapse
|
8
|
Pampanini V, Wagner M, Asadi-Azarbaijani B, Oskam IC, Sheikhi M, Sjödin MOD, Lindberg J, Hovatta O, Sahlin L, Björvang RD, Otala M, Damdimopoulou P, Jahnukainen K. Impact of first-line cancer treatment on the follicle quality in cryopreserved ovarian samples from girls and young women. Hum Reprod 2020; 34:1674-1685. [PMID: 31411325 PMCID: PMC6736429 DOI: 10.1093/humrep/dez125] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 06/07/2019] [Indexed: 01/18/2023] Open
Abstract
STUDY QUESTION Does first-line chemotherapy affect the quality of ovarian pre-antral follicles and stromal tissue in a population of young patients? SUMMARY ANSWER Exposure to first-line chemotherapy significantly impacts follicle viability, size of residual intact follicles, steroid secretion in culture and quality of the stromal compartment. WHAT IS KNOWN ALREADY First-line chemotherapy is considered to have a low gonadotoxic potential, and as such, does not represent an indication for fertility preservation. Studies investigating the effects of chemotherapy on the quality of ovarian tissue stored for fertility preservation in young patients are limited and the results sometimes contradictory. STUDY DESIGN, SIZE, DURATION We conducted a retrospective cohort study including young patients referred to three centers (Helsinki, Oslo and Tampere) to perform ovarian tissue cryopreservation for fertility preservation between 2003 and 2018. PARTICIPANTS/MATERIALS, SETTING, METHODS A total of 43 patients (age 1-24 years) were included in the study. A total of 25 were exposed to first-line chemotherapy before cryopreservation, whereas 18 patients were not. Density and size of follicles divided by developmental stages, prevalence of atretic follicles, health of the stromal compartment and functionality of the tissue in culture were evaluated and related to age and chemotherapy exposure. Activation of dormant follicles and DNA damage were also assessed. MAIN RESULTS AND THE ROLE OF CHANCE Patients exposed to first-line chemotherapy showed a significantly higher density of atretic primordial and intermediary follicles than untreated patients. The intact primordial and intermediary follicles were significantly smaller in size in patients exposed to chemotherapy. Production of steroids in culture was also significantly impaired and a higher content of collagen and DNA damage was observed in the stromal compartment of treated patients. Collectively, these observations may indicate reduced quality and developmental capacity of follicles as a consequence of first-line chemotherapy exposure. Neither increased activation of dormant follicles nor elevated levels of DNA damage in oocyte nuclei were found in patients exposed to chemotherapy. LIMITATIONS, REASONS FOR CAUTION The two groups were not homogeneous in terms of age and the patients were exposed to different treatments, which did not allow us to distinguish the effect of specific agents. The limited material availability did not allow us to perform all the analyses on the entire set of patients. WIDER IMPLICATION OF THE FINDINGS This study provides for the first time a comprehensive analysis of the effects of first-line chemotherapy on the health, density and functionality of follicles categorized according to the developmental stage in patients under 24 years of age. When exposed to these treatments, patients were considered at low/medium risk of infertility. Our data suggest a profound impact of these relatively safe therapies on ovarian health and encourages further exploration of this effect in follow-up studies in order to optimize fertility preservation for young cancer patients. STUDY FUNDING/COMPETING INTEREST(S) This study was funded by the Swedish Childhood Cancer Foundation, the Finnish Cancer Society, the Finnish Pediatric Research Foundation, the Väre Foundation for Pediatric Cancer Research, The Swedish Research Council, the Stockholm County Council (ALF project) and Karolinska Institutet. The authors have no conflict of interest to declare.
Collapse
Affiliation(s)
- Valentina Pampanini
- Department of Women's and Children's Health, NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Solna, Sweden
| | - Magdalena Wagner
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet and University Hospital, Huddinge, Stockholm, Sweden
| | | | - Irma C Oskam
- The Animal Production Experimental Centre at the Norwegian University for Life Sciences, Oslo Norway
| | - Mona Sheikhi
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet and University Hospital, Huddinge, Stockholm, Sweden.,Stockholm IVF-Eugin, Stockholm, Sweden
| | - Marcus O D Sjödin
- Unit of Toxicological Sciences, Swetox, Karolinska Institutet, Södertälje, Sweden
| | - Johan Lindberg
- Unit of Toxicological Sciences, Swetox, Karolinska Institutet, Södertälje, Sweden
| | - Outi Hovatta
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet and University Hospital, Huddinge, Stockholm, Sweden
| | - Lena Sahlin
- Department of Women's and Children's Health, NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Solna, Sweden
| | - Richelle D Björvang
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet and University Hospital, Huddinge, Stockholm, Sweden.,Unit of Toxicological Sciences, Swetox, Karolinska Institutet, Södertälje, Sweden
| | - Marjut Otala
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Pauliina Damdimopoulou
- Department of Clinical Science, Intervention and Technology, Division of Obstetrics and Gynecology, Karolinska Institutet and University Hospital, Huddinge, Stockholm, Sweden.,Unit of Toxicological Sciences, Swetox, Karolinska Institutet, Södertälje, Sweden
| | - Kirsi Jahnukainen
- Department of Women's and Children's Health, NORDFERTIL Research Lab Stockholm, Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Solna, Sweden.,Division of Hematology-Oncology and Stem Cell Transplantation, New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
9
|
Quan N, Mara JN, Grover AR, Pavone ME, Duncan FE. Spatial Analysis of Growing Follicles in the Human Ovary to Inform Tissue Engineering Strategies. Tissue Eng Part A 2020; 26:733-746. [PMID: 32598235 DOI: 10.1089/ten.tea.2020.0051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cancer survivorship has increased considerably, but common cancer treatments may threaten female reproductive health and fertility. In females, standard fertility preservation techniques include egg and embryo banking and ovarian tissue cryopreservation, but these methods are not suitable for all individuals. Emerging fertility preservation technologies include in vitro follicle growth and ovarian bioprosthetics. Although these platforms hold tremendous promise, they remain in the preclinical phase likely because of our inability to adequately phenocopy the complexity of the in vivo ovarian environment. The goal of this study was to use an established research archive of fixed human ovarian tissue established through the Oncofertility Consortium to better understand the dynamics and milieu of growing follicles within the human ovary. We performed a histological analysis of the immediate surroundings of primary and secondary stage follicles. We evaluated oocyte and follicle diameters of these growing follicles, analyzed their growth trajectories, and mapped their precise relationships to other stage follicles within a defined area. We also stratified our findings according to participant age and previous treatment history. Our results serve as in vivo benchmarks for follicles grown in vitro and provide insight into how follicles should be seeded spatially within bioprosthetic ovaries, potentially improving the efficacy and clinical translation of these emerging technologies. Impact statement Life-preserving cancer treatments have greatly increased survivorship. However, treatments often have off-target health consequences that threaten female reproductive health and fertility. Although several standard fertility preservation options exist, there is a constant need to explore and expand options for all populations. In vitro follicle growth and ovarian bioprosthetics are new experimental procedures, which are currently limited to proof of concept. In this study, we analyzed human ovarian tissue from a deidentified biospecimen repository to characterize the growing follicle landscape with the ultimate goal of informing bioengineering practices. This spatial analysis pinpoints the geometry of growing follicles within the human ovary and provides a framework for paralleling this environment in ex vivo platforms.
Collapse
Affiliation(s)
- Natalie Quan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jamie N Mara
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Allison R Grover
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mary Ellen Pavone
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
10
|
Winship AL, Willson C, Hansen KR, Hutt KJ, Hickey M. Do BRCA1 and BRCA2 gene mutation carriers have a reduced ovarian reserve? Protocol for a prospective observational study. BMJ Open 2019; 9:e033810. [PMID: 31772111 PMCID: PMC6887091 DOI: 10.1136/bmjopen-2019-033810] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION BRCA1/2 gene mutations increase risk of breast and/or ovarian cancer and may have implications for reproductive health. Indirect biomarkers of the ovarian primordial follicle pool (anti-Müllerian hormone (AMH)) and one small study in female cadavers suggest that ovarian reserve may be reduced in BRCA mutation carriers, but findings are conflicting and association between circulating AMH and primordial follicle number is not established. The aim of this study is to measure primordial follicle density in premenopausal ovarian tissue samples from women with BRCA1/2 gene mutations versus age-matched comparison group. METHODS AND ANALYSIS Prospective observational study measuring associations between BRCA gene mutation status, premenopausal ovarian primordial follicle density and serum AMH concentrations versus age-matched premenopausal women from the general population. Primordial follicle density will be measured in cortical sections from ovarian tissue collected at the time of risk-reducing bilateral salpingo-oophorectomy (RRBSO) in 88 BRCA1 gene mutation carriers, 65 BRCA2 gene mutation carriers and 157 non-mutation carriers. Primordial follicle density will be determined by counting follicles in a known volume of ovarian cortical tissue using light microscopy. Follicles will be identified by immunohistochemical staining for oocyte marker mouse vasa homologue. To inform the mechanisms underlying reduced ovarian reserve, the proportion of follicles containing oocytes with DNA damage will be determined by immunohistochemical staining for phosphorylated histone H2AX and terminal deoxynucleotidyl transferase dUTP nick end labelling assay to identify apoptotic cells. Follicle density will be correlated with circulating AMH concentrations quantified in the same cohort, using an electrochemiluminescence immunoassay on an automated platform. ETHICS AND DISSEMINATION Ethics approval has been granted by Peter MacCallum Cancer Centre to access biobanks, including; The Kathleen Cuningham Foundation Consortium for Research into Familial Breast Cancer (kConFab-HREC#97_27) and the What Happens after Menopause? (HREC12PMCC24-12/90) and Melbourne IVF.
Collapse
Affiliation(s)
- Amy Louise Winship
- Department of Anatomy and Developmental Biology, Monash University Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia
| | - Chris Willson
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
- Gynaecology Research Centre, The Royal Women's Hospital, Melbourne, Victoria, Australia
| | - Karl R Hansen
- College of Medicine, University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Karla J Hutt
- Department of Anatomy and Developmental Biology, Monash University Monash Biomedicine Discovery Institute, Clayton, Victoria, Australia
| | - Martha Hickey
- Department of Obstetrics and Gynaecology, The University of Melbourne, Melbourne, Victoria, Australia
- Gynaecology Research Centre, The Royal Women's Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Wang Y, Liu M, Johnson SB, Yuan G, Arriba AK, Zubizarreta ME, Chatterjee S, Nagarkatti M, Nagarkatti P, Xiao S. Doxorubicin obliterates mouse ovarian reserve through both primordial follicle atresia and overactivation. Toxicol Appl Pharmacol 2019; 381:114714. [PMID: 31437492 DOI: 10.1016/j.taap.2019.114714] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/12/2019] [Accepted: 08/17/2019] [Indexed: 12/22/2022]
Abstract
Ovarian toxicity and infertility are major side effects of cancer therapy in young female cancer patients. We and others have previously demonstrated that doxorubicin (DOX), one of the most widely used chemotherapeutic chemicals, has a dose-dependent toxicity on growing follicles. However, it is not fully understood if the primordial follicles are the direct or indirect target of DOX. Using both prepubertal and young adult female mouse models, we comprehensively investigated the effect of DOX on all developmental stages of follicles, determined the impact of DOX on primordial follicle survival, activation, and development, as well as compared the impact of age on DOX-induced ovarian toxicity. Twenty-one-day-old CD-1 female mice were intraperitoneally injected with PBS or clinically relevant dose of DOX at 10 mg/kg once. Results indicated that DOX primarily damaged granulosa cells in growing follicles and oocytes in primordial follicles and DOX-induced growing follicle apoptosis was associated with the primordial follicle overactivation. Using the 5-day-old female mice with a more uniform primordial follicle population, our data revealed that DOX also directly promoted primordial follicle death and the DNA damage-TAp63α-C-CASP3 pathway was involved in DOX-induced primordial follicle oocyte apoptosis. Compared to 21-day- and 8-week-old female mice that were treated with the same dose of DOX, the 5-day-old mice had the most severe primordial follicle loss as well as the least degree of primordial follicle overactivation. Taken together, these results demonstrate that DOX obliterates mouse ovarian reserve through both primordial follicle atresia and overactivation and the DOX-induced ovarian toxicity is age dependent.
Collapse
Affiliation(s)
- Yingzheng Wang
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Mingjun Liu
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Sarah B Johnson
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Gehui Yuan
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA; Department of Hygienic Analysis and Detection, School of Public Health, Nanjing Medical University, Nanjing 21009, China
| | - Alana K Arriba
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Maria E Zubizarreta
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Saurabh Chatterjee
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA
| | - Mitzi Nagarkatti
- Department of Microbiology and Immunology, School of Medicine, University of South Carolina, SC 29208, USA
| | - Prakash Nagarkatti
- Department of Microbiology and Immunology, School of Medicine, University of South Carolina, SC 29208, USA
| | - Shuo Xiao
- Department of Environmental Health Sciences, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
12
|
Akahori T, Woods DC, Tilly JL. Female Fertility Preservation through Stem Cell-based Ovarian Tissue Reconstitution In Vitro and Ovarian Regeneration In Vivo. CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119848007. [PMID: 31191070 PMCID: PMC6540489 DOI: 10.1177/1179558119848007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 03/27/2019] [Indexed: 12/13/2022]
Abstract
Historically, approaches designed to offer women diagnosed with cancer the prospects of having a genetically matched child after completion of their cytotoxic treatments focused on the existing oocyte population as the sole resource available for clinical management of infertility. In this regard, elective oocyte and embryo cryopreservation, as well as autologous ovarian cortical tissue grafting posttreatment, have gained widespread support as options for young girls and reproductive-age women who are faced with cancer to consider. In addition, the use of ovarian protective therapies, including gonadotropin-releasing hormone agonists and sphingosine-1-phosphate analogs, has been put forth as an alternative way to preserve fertility by shielding existing oocytes in the ovaries in vivo from the side-effect damage caused by radiotherapy and many chemotherapeutic regimens. This viewpoint changed with the publication of now numerous reports that adult ovaries of many mammalian species, including humans, contain a rare population of oocyte-producing germ cells-referred to as female germline or oogonial stem cells (OSCs). This new line of study has fueled research into the prospects of generating new oocytes, rather than working with existing oocytes, as a novel approach to sustain or restore fertility in female cancer survivors. Here, we overview the history of work from laboratories around the world focused on improving our understanding of the biology of OSCs and how these cells may be used to reconstitute "artificial" ovarian tissue in vitro or to regenerate damaged ovarian tissue in vivo as future fertility-preservation options.
Collapse
Affiliation(s)
- Taichi Akahori
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, USA.,On leave from the Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Dori C Woods
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, USA
| | - Jonathan L Tilly
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, USA
| |
Collapse
|
13
|
Somigliana E, Terenziani M, Filippi F, Bergamini A, Martinelli F, Mangili G, Peccatori F, Vercellini P. Chemotherapy-related damage to ovarian reserve in childhood cancer survivors: interpreting the evidence. J Assist Reprod Genet 2019; 36:341-348. [PMID: 30362055 PMCID: PMC6420530 DOI: 10.1007/s10815-018-1345-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/12/2018] [Indexed: 01/23/2023] Open
Abstract
Chemotherapy during childhood damages ovarian reserve and can affect future fertility. However, recent large epidemiological studies showed that the detrimental impact on fertility is less severe if women seek for pregnancy at a younger age. To explain this observation, we hypothesize that the detrimental effects of previous chemotherapy on the ovarian reserve may be attenuated in young adults for two main reasons. Firstly, recent evidence showed that the amount of ovarian reserve is not a critical factor for effective natural conceptions. Provided that the residual ovarian reserve allows regular ovulatory cycles, the chances of pregnancy are similar in women with intact or reduced ovarian reserve. Secondly, ovarian reserve depletion appears to be a phenomenon that is inversely related to the residual ovarian reserve rather than to age. From a mathematical perspective, this kind of regulation intrinsically attenuates the effects of an early loss of a significant amount of primordial follicles. In conclusion, the detrimental effects of chemotherapy on natural fertility may be less severe if women with a history of chemotherapy during childhood seek for pregnancy early. This information should be part of the counseling.
Collapse
Affiliation(s)
- Edgardo Somigliana
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy.
- Obstet-Gynecol Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
- Infertility Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via M. Fanti, 6, 20122, Milan, Italy.
| | - Monica Terenziani
- Pediatric Oncology Unit, Fondazione IRCCS National Cancer Institute, Milan, Italy
| | - Francesca Filippi
- Obstet-Gynecol Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Alice Bergamini
- Obstet-Gynecol Department, San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Martinelli
- Gynecologic Oncology Unit, Fondazione IRCCS National Cancer Institute, Milan, Italy
| | - Giorgia Mangili
- Obstet-Gynecol Department, San Raffaele Scientific Institute, Milan, Italy
| | - Fedro Peccatori
- Fertility and Procreation Unit, Division of Gynaecologic Oncology, European Institute of Oncology, Milan, Italy
| | - Paolo Vercellini
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Obstet-Gynecol Department, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
14
|
Winship AL, Bakai M, Sarma U, Liew SH, Hutt KJ. Dacarbazine depletes the ovarian reserve in mice and depletion is enhanced with age. Sci Rep 2018; 8:6516. [PMID: 29695822 PMCID: PMC5917018 DOI: 10.1038/s41598-018-24960-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/05/2018] [Indexed: 01/06/2023] Open
Abstract
Dacarbazine is commonly administered for the treatment of cancers prevalent in reproductive age females. However, investigations of off-target effects of dacarbazine on the ovary are limited. We assessed the impact of dacarbazine on the ovarian reserve of primordial follicles, essential for fertility. Eight week and 6 month old C57BL/6 J mice were administered with dacarbazine or saline on day (d)0 and d7, then sacrificed after 12 hours (h), or 14d (n = 4-5/group). Follicle numbers, follicle density, serum AMH and corpora lutea were quantified and estrous cyclicity monitored. In reproductively young mice, dacarbazine did not affect primordial follicle numbers at 12 h, but resulted in a 36% reduction at 14d (p < 0.05). Dacarbazine-mediated primordial follicle depletion was accelerated with age, with a 24% (p < 0.05) and 36% (p < 0.01) reduction at 12 h and 14d. Follicle density remained unchanged between treatment groups at either age. Dacarbazine depleted antral follicles at 14d (p < 0.05), at both ages. Despite partial reduction of antral follicles, serum AMH, estrous cyclicity and corpora lutea (indicative of ovulation) remained unchanged between treatment groups, at both ages. Importantly, diminished ovarian reserve can result in premature ovarian insufficiency and infertility, thus, fertility preservation options should be considered for young female patients prior to dacarbazine treatment.
Collapse
Affiliation(s)
- Amy L Winship
- Ovarian Biology Laboratory, Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Monika Bakai
- Ovarian Biology Laboratory, Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Urooza Sarma
- Ovarian Biology Laboratory, Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Seng H Liew
- Ovarian Biology Laboratory, Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Karla J Hutt
- Ovarian Biology Laboratory, Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia.
| |
Collapse
|
15
|
Talevi R, Sudhakaran S, Barbato V, Merolla A, Braun S, Di Nardo M, Costanzo V, Ferraro R, Iannantuoni N, Catapano G, Gualtieri R. Is oxygen availability a limiting factor for in vitro folliculogenesis? PLoS One 2018; 13:e0192501. [PMID: 29425251 PMCID: PMC5806880 DOI: 10.1371/journal.pone.0192501] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/24/2018] [Indexed: 02/06/2023] Open
Abstract
Transplantation of ovarian tissue for the preservation of fertility in oncological patients is becoming an accepted clinical practice. However, the risk of re-introducing tumour cells at transplantation has stirred an increased interest for complete in vitro folliculogenesis. This has not yet been achieved in humans possibly for the lack of knowledge on the environmental milieu that orchestrates folliculogenesis in vivo. The main aim of this study was to investigate the effect of oxygen availability on follicle health and growth during in vitro culture of ovarian tissue strips. To this end, a model was developed to predict the dissolved oxygen concentration in tissue under varying culture conditions. Ovarian cortical strips of bovine, adopted as an animal model, and human tissue were cultured in conventional (CD) and gas permeable (PD) dishes under different media column heights and gaseous oxygen tensions for 3, 6 and 9 days. Follicle quality, activation of primordial follicles to the primary stage, and progression to the secondary stage were analysed through histology. Follicle viability was assessed through a live-dead assay at the confocal scanning laser microscope. Findings showed a higher follicle quality and viability after culture of bovine ovarian strips in PD in adequate medium height and oxygen tensions. The best culture conditions found in the bovine were adopted for human ovarian strip culture and promoted a higher follicle quality, viability and progression. Overall, data demonstrated that modulation of oxygen availability in tissue plays a key role in maintaining follicles' health and their ability to survive and progress to the secondary stage during ovarian tissue in vitro culture. Such culture conditions could increase the yield of healthy secondary follicles for subsequent dissection and individual culture to obtain competent oocytes.
Collapse
Affiliation(s)
- Riccardo Talevi
- Dipartimento di Biologia, Università di Napoli "Federico II", Complesso Universitario di Monte S Angelo, Napoli, Italy
| | - Sam Sudhakaran
- St Bartholomew's Hospital, W Smithfield, London, United Kingdom
| | - Vincenza Barbato
- Dipartimento di Biologia, Università di Napoli "Federico II", Complesso Universitario di Monte S Angelo, Napoli, Italy
| | - Anna Merolla
- Dipartimento di Biologia, Università di Napoli "Federico II", Complesso Universitario di Monte S Angelo, Napoli, Italy
| | - Sabrina Braun
- Dipartimento di Biologia, Università di Napoli "Federico II", Complesso Universitario di Monte S Angelo, Napoli, Italy
| | - Maddalena Di Nardo
- Dipartimento di Biologia, Università di Napoli "Federico II", Complesso Universitario di Monte S Angelo, Napoli, Italy
| | - Valentina Costanzo
- Dipartimento di Biologia, Università di Napoli "Federico II", Complesso Universitario di Monte S Angelo, Napoli, Italy
| | | | - Nicola Iannantuoni
- Ospedale S. Maria delle Grazie, ASL Napoli 2 Nord, Località La Schiana, Pozzuoli, Italy
| | - Gerardo Catapano
- Department of Environmental and Chemical Engineering, University of Calabria, Rende, Italy
| | - Roberto Gualtieri
- Dipartimento di Biologia, Università di Napoli "Federico II", Complesso Universitario di Monte S Angelo, Napoli, Italy
| |
Collapse
|
16
|
Bhartiya D, Patel H. Ovarian stem cells-resolving controversies. J Assist Reprod Genet 2017; 35:393-398. [PMID: 29128912 DOI: 10.1007/s10815-017-1080-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/01/2017] [Indexed: 12/13/2022] Open
Abstract
A recent review on ovarian stem cells by Horan and Williams entitled "Oocyte Stem Cells: Fact or Fantasy?" suggests that the debate on ovarian stem cells (OSCs) is still not over. They did not even discuss the presence of two distinct populations of stem cells in the ovary in their review. OSCs are located in the ovary surface epithelium and Tilly's group reported them in the size range of 5-8 μm whereas Virant-Klun's group has reported pluripotent, 2-4 μm OSCs. Our group reported OSCs of two distinct sizes including pluripotent very small embryonic-like stem cells (VSELs) which are smaller in size than RBCs (similar to those reported by Virant-Klun's group) and slightly bigger (similar to those reported by Tilly's group) tissue committed progenitors (OSCs) that presumably differentiate from VSELs. These stem/progenitor cells express receptors for follicle stimulating hormone (FSH) and are activated by FSH. Our opinion article provides explanation to several open-ended questions raised in the review on OSCs by Horan and Williams. VSELs survive chemotherapy; maintain life-long homeostasis; loss of their function due to a compromised niche results in age-related senescence and presence of overlapping pluripotent markers suggest that they may also be implicated in epithelial ovarian cancers.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India.
| | - Hiren Patel
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| |
Collapse
|
17
|
Horan CJ, Williams SA. Oocyte stem cells: fact or fantasy? Reproduction 2017; 154:R23-R35. [PMID: 28389520 DOI: 10.1530/rep-17-0008] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/29/2017] [Accepted: 04/07/2017] [Indexed: 01/08/2023]
Abstract
For many decades, the dogma prevailed that female mammals had a finite pool of oocytes at birth and this was gradually exhausted during a lifetime of reproductive function. However, in 2004, a new era began in the field of female oogenesis. A study was published that appeared to detect oocyte-stem cells capable of generating new eggs within mouse ovaries. This study was highly controversial and the years since this initial finding have produced extensive research and even more extensive debate into their possibility. Unequivocal evidence testifying to the existence of oocyte-stem cells (OSCs) has yet to be produced, meanwhile the spectrum of views from both sides of the debate are wide-ranging and surprisingly passionate. Although recent studies have presented some convincing results that germ cells exist and are capable of creating new oocytes, many questions remain. Are these cells present in humans? Do they exist in physiological conditions in a dormant state? This comprehensive review first examines where and how the dogma of a finite pool was established, how this has been challenged over the years and addresses the most pertinent questions as to the current status of their existence, their role in female fertility, and perhaps most importantly, if they do exist, how can we harness these cells to improve a woman's oocyte reserve and treat conditions such as premature ovarian insufficiency (POI: also known as premature ovarian failure, POF).
Collapse
Affiliation(s)
- Corrina J Horan
- Nuffield Department of Obstetrics and GynaecologyUniversity of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - Suzannah A Williams
- Nuffield Department of Obstetrics and GynaecologyUniversity of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|