1
|
Morales-Sánchez E, Campuzano-Caballero JC, Cervantes A, Martínez-Ibarra A, Cerbón M, Vital-Reyes VS. Which Side of the Coin Are You on Regarding Possible Postnatal Oogenesis? Arch Med Res 2024; 55:103071. [PMID: 39236439 DOI: 10.1016/j.arcmed.2024.103071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 08/14/2024] [Indexed: 09/07/2024]
Abstract
It is well known that oocytes are produced during fetal development and that the total number of primary follicles is determined at birth. In humans, there is a constant loss of follicles after birth until about two years of age. The number of follicles is preserved until the resumption of meiosis at puberty and there is no renewal of the oocytes; this dogma was maintained in the last century because there were no suitable techniques to detect and obtain stem cells. However, following stem cell markers, several scientists have detected them in developing and adult human ovarian tissues, especially in the ovarian surface epithelial cells. Furthermore, many authors using different methodological strategies have indicated this possibility. This evidence has led many scientists to explore this hypothesis; there is no definitive consensus to accept this idea. Interestingly, oocyte retrieval from mature ovaries and other tissue sources of stem cells has contributed to the development of strategies for the retrieval of mature oocytes, useful for assisted reproductive technology. Here, we review the evidence and controversies on oocyte neooogenesis in adult women; in addition, we agree with the idea that this process may occur in adulthood and that its alteration may be related to various pathologies in women, such as polycystic ovary syndrome, premature ovarian insufficiency, diminished ovarian reserve and several infertility and genetic disorders.
Collapse
Affiliation(s)
- Elizabeth Morales-Sánchez
- Unidad de Histología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Juan Carlos Campuzano-Caballero
- Departamento de Biología Comparada, Facultad de Ciencias, Laboratorio de Biología de la Reproducción Animal, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alicia Cervantes
- Servicio de Genética, Hospital General de México, Eduardo Liceaga, Mexico City, Mexico; Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Alejandra Martínez-Ibarra
- Departmento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Av. Universidad 3000, Circuito Escolar, Mexico City, Coyoacán 04510, Mexico
| | - Marco Cerbón
- Departmento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad Universitaria, Av. Universidad 3000, Circuito Escolar, Mexico City, Coyoacán 04510, Mexico.
| | | |
Collapse
|
2
|
Rashidian P. An update on oncofertility in prepubertal females. J Gynecol Obstet Hum Reprod 2024; 53:102742. [PMID: 38341083 DOI: 10.1016/j.jogoh.2024.102742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/12/2024]
Abstract
Cancer is a life-threatening event for pediatric patients. Treatment advancements in pediatric cancer have improved prognosis, but some of these treatments have gonadotoxic potential and may affect fertility in different ways. Due to the growing interest of the research community in the life prospects of young cancer survivors, there has been a demand to intersect reproductive medicine and oncology, which is referred to as "oncofertility". There are various fertility preservation options according to gender and pubertal status, and shared decisions must take place at the time of diagnosis. This study aims to provide a critical review of current and emerging strategies for preserving and restoring fertility in prepubertal females, ranging from established methods to experimental approaches that can be offered before, during, and after anticancer therapies. Additionally, the author aims to review how clinicians' awareness of oncofertility options and the latest advancements in this field, timely referral, and proper consultations with patients and their families are vital in addressing their concerns, providing emotional support, and guiding them through the decision-making process, as well as potential barriers that may hinder the fertility preservation process.
Collapse
Affiliation(s)
- Pegah Rashidian
- Reproductive Health Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
3
|
Future regenerative medicine developments and their therapeutic applications. Biomed Pharmacother 2023; 158:114131. [PMID: 36538861 DOI: 10.1016/j.biopha.2022.114131] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Although the currently available pharmacological assays can cure most pathological disorders, they have limited therapeutic value in relieving certain disorders like myocardial infarct, peripheral vascular disease, amputated limbs, or organ failure (e.g. renal failure). Pilot studies to overcome such problems using regenerative medicine (RM) delivered promising data. Comprehensive investigations of RM in zebrafish or reptilians are necessary for better understanding. However, the precise mechanisms remain poorly understood despite the tremendous amount of data obtained using the zebrafish model investigating the exact mechanisms behind their regenerative capability. Indeed, understanding such mechanisms and their application to humans can save millions of lives from dying due to potentially life-threatening events. Recent studies have launched a revolution in replacing damaged human organs via different approaches in the last few decades. The newly established branch of medicine (known as Regenerative Medicine aims to enhance natural repair mechanisms. This can be done through the application of several advanced broad-spectrum technologies such as organ transplantation, tissue engineering, and application of Scaffolds technology (support vascularization using an extracellular matrix), stem cell therapy, miRNA treatment, development of 3D mini-organs (organoids), and the construction of artificial tissues using nanomedicine and 3D bio-printers. Moreover, in the next few decades, revolutionary approaches in regenerative medicine will be applied based on artificial intelligence and wireless data exchange, soft intelligence biomaterials, nanorobotics, and even living robotics capable of self-repair. The present work presents a comprehensive overview that summarizes the new and future advances in the field of RM.
Collapse
|
4
|
Tian C, An Y, Zhao J, Zhu X, Wei W, Ruan G, Li Y, Pan X. Bone Marrow Mesenchymal Stem Cells Reversed Ovarian Aging-related m6A RNA Methylation Modification Profile in Aged Granulosa Cells. Stem Cell Rev Rep 2023; 19:953-967. [PMID: 36609903 DOI: 10.1007/s12015-022-10485-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Ovarian ageing causes endocrine disturbances and the degeneration of systemic tissue and organ functions to seriously affect women's physical and mental health, and effective treatment methods are urgently needed. Based on our previous studies using juvenile rhesus monkey bone marrow mesenchymal stem cells (BMMSCs) to treat ovarian ageing in rhesus monkey, we found that BMMSCs improved ovarian structure and function. This study continues to explore the mechanism by which BMMSCs reversed granulosa cell (GC) ageing. METHODS A GC ageing model and coculture system of BMMSCs were established, changes in the level of the N6-methyladenosine (m6A) methylation modification were detected, m6A-modified RNA immunoprecipitation sequencing (MeRIP-seq) were performed, correlations between m6A peaks and mRNA expression were determined, and the expression of hub genes was identified using Q-PCR, immunofluorescence staining, and western blot. RESULTS Our results showed that H2O2 successfully induced GC ageing and that BMMSCs reversed measures of GC ageing. BMMSCs increased the expression of the FTO protein and reduced the overall level of m6A. We identified 797 m6A peaks (348 hypomethylated and 449 hypermethylated peaks) and 817 differentially expressed genes (DEGs) (412 upregulated and 405 downregulated) after aged GCs were cocultured with BMMSCs, which significantly associated with ovarian function and epigenetic modification. The epigenetic repressive mark and important cell cycle regulator lysine demethylase 8 (KDM8) was downregulated at both the mRNA and protein levels, histone H3 was upregulated in aged GCs after BMMSC coculture, and KDM8 was upregulated after FTO was inhibited through FB23. CONCLUSIONS Our study revealed an essential role for m6A in BMMSCs in reversing GC ageing, and FTO regulated KDM8 mediates histone H3 changes may as a novel regulatory mechanism in BMMSCs to reverse GC ageing.
Collapse
Affiliation(s)
- Chuan Tian
- The Basic Medical Laboratory of the 920Th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
| | - Yuanyuan An
- The Affiliated Stomatology of Kunming Medical University, Kunming, 650106, Yunnan Province, China
| | - Jing Zhao
- The Basic Medical Laboratory of the 920Th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
| | - Xiangqing Zhu
- The Basic Medical Laboratory of the 920Th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
| | - Wei Wei
- The Basic Medical Laboratory of the 920Th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
| | - Guangping Ruan
- The Basic Medical Laboratory of the 920Th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
| | - Ye Li
- The Basic Medical Laboratory of the 920Th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
| | - Xinghua Pan
- The Basic Medical Laboratory of the 920Th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China.
| |
Collapse
|
5
|
Alberico H, Fleischmann Z, Bobbitt T, Takai Y, Ishihara O, Seki H, Anderson RA, Telfer EE, Woods DC, Tilly JL. Workflow Optimization for Identification of Female Germline or Oogonial Stem Cells in Human Ovarian Cortex Using Single-Cell RNA Sequence Analysis. Stem Cells 2022; 40:523-536. [PMID: 35263439 PMCID: PMC9199849 DOI: 10.1093/stmcls/sxac015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/15/2022] [Indexed: 11/16/2022]
Abstract
In 2004, the identification of female germline or oogonial stem cells (OSCs) that can support post-natal oogenesis in ovaries of adult mice sparked a major paradigm shift in reproductive biology. Although these findings have been independently verified, and further extended to include identification of OSCs in adult ovaries of many species ranging from pigs and cows to non-human primates and humans, a recent study rooted in single-cell RNA sequence analysis (scRNA-seq) of adult human ovarian cortical tissue claimed that OSCs do not exist, and that other groups working with OSCs following isolation by magnetic-assisted or fluorescence-activated cell sorting have mistaken perivascular cells (PVCs) for germ cells. Here we report that rare germ lineage cells with a gene expression profile matched to OSCs but distinct from that of other cells, including oocytes and PVCs, can be identified in adult human ovarian cortical tissue by scRNA-seq after optimization of analytical workflow parameters. Deeper cell-by-cell expression profiling also uncovered evidence of germ cells undergoing meiosis-I in adult human ovaries. Lastly, we show that, if not properly controlled for, PVCs can be inadvertently isolated during flow cytometry protocols designed to sort OSCs because of inherently high cellular autofluorescence. However, human PVCs and human germ cells segregate into distinct clusters following scRNA-seq due to non-overlapping gene expression profiles, which would preclude the mistaken identification and use of PVCs as OSCs during functional characterization studies.
Collapse
Affiliation(s)
- Hannah Alberico
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Zoë Fleischmann
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Tyler Bobbitt
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Yasushi Takai
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, Saitama 350-0495, Japan
| | - Osamu Ishihara
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, Saitama 350-0495, Japan
| | - Hiroyuki Seki
- Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, Saitama 350-0495, Japan
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH14 1DJ, UK
| | - Evelyn E Telfer
- Institute of Cell Biology, University of Edinburgh, Edinburgh EH14 1DJ, UK
| | - Dori C Woods
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA 02115, USA
| | - Jonathan L Tilly
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
6
|
Silvestris E, Minoia C, Guarini A, Opinto G, Negri A, Dellino M, Tinelli R, Cormio G, Paradiso AV, De Palma G. Ovarian Stem Cells (OSCs) from the Cryopreserved Ovarian Cortex: A Potential for Neo-Oogenesis in Women with Cancer-Treatment Related Infertility: A Case Report and a Review of Literature. Curr Issues Mol Biol 2022; 44:2309-2320. [PMID: 35678686 PMCID: PMC9164018 DOI: 10.3390/cimb44050157] [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: 03/16/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer treatment related infertility (CTRI) affects more than one third of young women undergoing anti-cancer protocols, inducing a premature exhaustion of the ovarian reserve. In addition to ovarian suppression by GnRHa, oocyte and cortex cryopreservation has gained interest in patients with estrogen-sensitive tumors for whom the hormonal burst to prompt the multiple follicular growth could provide a further pro-life tumor pulsing. On the other hand, cortex reimplantation implies a few drawbacks due to the unknown consistency of the follicles to be reimplanted or the risk of reintroducing malignant cells. The capability of ovarian stem cells (OCSs) from fresh ovarian cortex fragments to differentiate in vitro to mature oocytes provides a tool to overcome these drawbacks. In fact, since ovarian cortex sampling and cryopreservation is practicable before gonadotoxic treatments, the recruitment of OSCs from defrosted fragments could provide a novel opportunity to verify their suitability to be expanded in vitro as oocyte like cells (OLCs). Here, we describe in very preliminary experiments the consistency of an OSC population from a single cryopreserved ovarian cortex after thawing as well as both their viability and their suitability to be further explored in their property to differentiate in OLCs, thus reinforcing interest in stemness studies in the treatment of female CTRI.
Collapse
Affiliation(s)
- Erica Silvestris
- Gynecologic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy
- Correspondence:
| | - Carla Minoia
- Haematology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (C.M.); (A.G.); (G.O.); (A.N.)
| | - Attilio Guarini
- Haematology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (C.M.); (A.G.); (G.O.); (A.N.)
| | - Giuseppina Opinto
- Haematology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (C.M.); (A.G.); (G.O.); (A.N.)
| | - Antonio Negri
- Haematology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (C.M.); (A.G.); (G.O.); (A.N.)
| | - Miriam Dellino
- Department of Obstetrics and Gynecology, “San Paolo” Hospital, 70123 Bari, Italy;
| | - Raffaele Tinelli
- Department of Obstetrics and Gynecology, “Valle d’Itria” Hospital, 74015 Martina Franca, Italy;
| | - Gennaro Cormio
- Unit of Obstetrics and Gynecology, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Angelo Virgilio Paradiso
- Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (A.V.P.); (G.D.P.)
| | - Giuseppe De Palma
- Institutional BioBank, Experimental Oncology and Biobank Management Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (A.V.P.); (G.D.P.)
| |
Collapse
|
7
|
The process of ovarian aging: it is not just about oocytes and granulosa cells. J Assist Reprod Genet 2022; 39:783-792. [PMID: 35352316 PMCID: PMC9051003 DOI: 10.1007/s10815-022-02478-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ovarian age is classically considered the main cause of female reproductive infertility. In women, the process proceeds as an ongoing decline in the primordial follicle stockpile and it is associated with reduced fertility in the mid-thirties, irregular menstruation from the mid-forties, cessation of fertility, and, eventually, menopause in the early fifties. Reproductive aging is historically associated with changes in oocyte quantity and quality. However, besides the oocyte, other cellular as well as environmental factors have been the focus of more recent investigations suggesting that ovarian decay is a complex and multifaceted process. Among these factors, we will consider mitochondria and oxidative stress as related to nutrition, changes in extracellular matrix molecules, and the associated ovarian stromal compartment where immune cells of both the native and adaptive systems seem to play an important role. Understanding such processes is crucial to design treatment strategies to slow down ovarian aging and consequently prolong reproductive lifespan and, more to this, alleviaingt side effects of menopause on the musculoskeletal, cardiovascular, and nervous systems.
Collapse
|
8
|
Durkina VB, Chapman JW, Demchenko NL. First observations of ovary regeneration in an amphipod, Ampelisca eschrichtii Krøyer, 1842. PeerJ 2022; 10:e12950. [PMID: 35291485 PMCID: PMC8918206 DOI: 10.7717/peerj.12950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/26/2022] [Indexed: 01/11/2023] Open
Abstract
Background Females of the gammaridean amphipod Ampelisca eschrichtii with signs of regenerating, previously atrophied ovaries were recovered from the northeastern shelf of Sakhalin Island (Okhotsk Sea, Russia). Ovarian regeneration was previously unknown for any amphipod species. A. eschrichtii have a predominantly 2-year life cycle (from embryo to adult death) and reproduce once between late winter or early spring at the age of 2 years. Occasionally, females survive to a third year. An adaptive value of extended survival among these females is likely to require that they are also reproductive. Methods Histological sections from a second-year female with ovarian atrophy, a female with normal ovaries, a third-year female with ovarian regeneration, as well as testes of an immature and a sexually mature male were compared to determine the sources of cells of the germinal and somatic lines necessary for ovarian regeneration. Results Ovarian regeneration in the third-year female began with the formation of a new germinal zone from germ cells preserved in the atrophied ovaries and eosinophilic cells of the previously starving second-year female. Eosinophilic cells form the mesodermal component of the germinal zone. A mass of these cells appeared in the second-year female that had atrophied ovaries and in large numbers on the intestine wall of the third-year female with regenerating ovaries. These eosinophilic cells appear to migrate into the regenerating ovaries. Conclusions All germ cells of the second-year female are not lost during ovarian atrophy and can be involved in subsequent ovarian regeneration. Eosinophilic cells involved in ovarian regeneration are of mesodermal origin. The eosinophilic cell morphologies are similar to those of quiescence cells (cells in a reversible state that do not divide but retain the ability to re-enter cell division and participate in regeneration). These histological data thus indicate that eosinophilic and germ cells of third-year females can participate in the regeneration of the ovaries to reproduce a second brood. The precursors of these third-year females (a small number the second-year females with an asynchronous [summer] breeding period and ovaries that have atrophied due to seasonal starvation) appear to possess sources of somatic and germ cells that are sufficient for ovarian regeneration and that may be adaptations to starvation stress.
Collapse
Affiliation(s)
- Valentina B. Durkina
- Laboratory of Physiology, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Primorsky Krai, Russia
| | - John W. Chapman
- Department of Fisheries, Wildlife and Conservation, Oregon State University, Newport, Oregon, United States of America
| | - Natalia L. Demchenko
- Laboratory of Marine Ecosystem Dynamics, A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Primorsky Krai, Russia
| |
Collapse
|
9
|
Assessment of female fertility preservation in Auvergne 3 years after implementation of the PREFERA platform (PREservation FERtilité Auvergne). J Gynecol Obstet Hum Reprod 2022; 51:102342. [PMID: 35181543 DOI: 10.1016/j.jogoh.2022.102342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Fertility preservation (FP) in patients with cancer or pathology at risk of gonadotoxicity is now according to legislation, an integral part of the treatment protocol. for this reason, clinical-biological platforms have emerged with the aim of developing and improving this practice, such as the PREFERA platform (PREservation FERtilité Auvergne) MATERIAL ET METHOD: This is an observational cohort study to evaluate female fertility preservation activity in Auvergne at the AMP-CECOS center of the Clermont-Ferrand University Hospital from March 2013 to March 2019. This period covering 3 years before and after the creation of PREFERA in 2015. RESULTS 205 patients were referred for fertility preservation consultations, including 77 before the platform was set up and 128 after, corresponding to an increase of 66%. 190 patients (92.7%) referred were eligible for FP, of whom 169 (88.9%) received treatment. Thirty-nine patients underwent oocyte vitrification before the platform was set up and 74 after (+89.7%), twenty patients underwent ovarian cortex freezing before the platform was set up and 27 after (+35%). Only 54 patients (26.2%) were seen for follow-up with an increased number of consultations following the implementation of PREFERA. (8% vs 33%, p<0.001). CONCLUSION Creation of the PREFERA platform facilitated patient access and management of fertility preservation procedures. However, at the regional level, it is necessary to continue to raise awareness of fertility issues, particularly in the context of post-cancer follow-up, both among patients and health professionals.
Collapse
|
10
|
Alberico HC, Woods DC. Role of Granulosa Cells in the Aging Ovarian Landscape: A Focus on Mitochondrial and Metabolic Function. Front Physiol 2022; 12:800739. [PMID: 35153812 PMCID: PMC8829508 DOI: 10.3389/fphys.2021.800739] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/29/2021] [Indexed: 01/11/2023] Open
Abstract
Mitochondria are at the intersection of aging and fertility, with research efforts centered largely on the role that these specialized organelles play in the relatively rapid decline in oocyte quality that occurs as females approach reproductive senescence. In addition to various roles in oocyte maturation, fertilization, and embryogenesis, mitochondria are critical to granulosa cell function. Herein, we provide a review of the literature pertaining to the role of mitochondria in granulosa cell function, with emphasis on how mitochondrial aging in granulosa cells may impact reproduction in female mammals.
Collapse
|
11
|
Wang J, Liu W, Yu D, Yang Z, Li S, Sun X. Research Progress on the Treatment of Premature Ovarian Failure Using Mesenchymal Stem Cells: A Literature Review. Front Cell Dev Biol 2021; 9:749822. [PMID: 34966738 PMCID: PMC8710809 DOI: 10.3389/fcell.2021.749822] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/29/2021] [Indexed: 12/31/2022] Open
Abstract
Premature ovarian failure (POF) has become one of the main causes of infertility in women of childbearing age and the incidence of POF is increasing year by year, seriously affecting the physical and mental health of patients and increasing the economic burden on families and society as a whole. The etiology and pathogenesis of POF are complex and not very clear at present. Currently, hormone replacement therapy is mainly used to improve the symptoms of low estrogen, but cannot fundamentally solve the fertility problem. In recent years, stem cell (SC) transplantation has become one of the research hotspots in the treatment of POF. The results from animal experiments bring hope for the recovery of ovarian function and fertility in patients with POF. In this article, we searched the published literature between 2000 and 2020 from the PubMed database (https://pubmed.ncbi.nlm.nih.gov), and summarized the preclinical research data and possible therapeutic mechanism of mesenchymal stem cells (MSCs) in the treatment of POF. Our aim is to provide useful information for understanding POF and reference for follow-up research and treatment of POF.
Collapse
Affiliation(s)
- Jing Wang
- Department of Reproductive Medicine, Department of Prenatal Diagnosis, The First Hospital of Jilin University, Changchun, China
| | - Wanru Liu
- Department of Reproductive Medicine, Department of Prenatal Diagnosis, The First Hospital of Jilin University, Changchun, China
| | - Dehai Yu
- The Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zongxing Yang
- Department of Clinical Laboratory, The First Hospital of Jilin University, Changchun, China
| | - Sijie Li
- Department of Breast Surgery, The First Hospital of Jilin University, Changchun, China
| | - Xiguang Sun
- Hand Surgery Department, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Tian C, He J, An Y, Yang Z, Yan D, Pan H, Lv G, Li Y, Wang Y, Yang Y, Zhu G, He Z, Zhu X, Pan X. Bone marrow mesenchymal stem cells derived from juvenile macaques reversed ovarian ageing in elderly macaques. Stem Cell Res Ther 2021; 12:460. [PMID: 34407863 PMCID: PMC8371769 DOI: 10.1186/s13287-021-02486-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/01/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Female sex hormone secretion and reproductive ability decrease with ageing. Bone marrow mesenchymal stem cells (BMMSCs) have been postulated to play a key role in treating ovarian ageing. METHODS We used macaque ovarian ageing models to observe the structural and functional changes after juvenile BMMSC treatment. Moreover, RNA-seq was used to analyse the ovarian transcriptional expression profile and key pathways through which BMMSCs reverse ovarian ageing. RESULTS In the elderly macaque models, the ovaries were atrophied, the regulation ability of sex hormones was reduced, the ovarian structure was destroyed, and only local atretic follicles were observed, in contrast with young rhesus monkeys. Intravenous infusion of BMMSCs in elderly macaques increased ovarian volume, strengthened the regulation ability of sex hormones, reduced the degree of pulmonary fibrosis, inhibited apoptosis, increased density of blood vessels, and promoted follicular regeneration. In addition, the ovarian expression characteristics of ageing-related genes of the elderly treatment group reverted to that of the young control group, 1258 genes that were differentially expressed, among which 415 genes upregulated with age were downregulated, 843 genes downregulated with age were upregulated after BMMSC treatment, and the top 20 differentially expressed genes (DEGs) in the protein-protein interaction (PPI) network were significantly enriched in oocyte meiosis and progesterone-mediated oocyte maturation pathways. CONCLUSION The BMMSCs derived from juvenile macaques can reverse ovarian ageing in elderly macaques.
Collapse
Affiliation(s)
- Chuan Tian
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
- Guizhou Medical University, Tissue Engineering and Stem Cell Experimental Center, Guizhou Provinc, Guiyang, 550004, China
| | - Jie He
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
- Kunming Medical University, Guizhou Province, Kunming, 650032, China
| | - Yuanyuan An
- Kunming Medical University, Guizhou Province, Kunming, 650032, China
| | - Zailing Yang
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
- Guizhou Medical University, Tissue Engineering and Stem Cell Experimental Center, Guizhou Provinc, Guiyang, 550004, China
| | - Donghai Yan
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
| | - Hang Pan
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
- Guizhou Medical University, Tissue Engineering and Stem Cell Experimental Center, Guizhou Provinc, Guiyang, 550004, China
| | - Guanke Lv
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
- Kunming Medical University, Guizhou Province, Kunming, 650032, China
| | - Ye Li
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
- Kunming Medical University, Guizhou Province, Kunming, 650032, China
| | - Yanying Wang
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
| | - Yukun Yang
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China
| | - Gaohong Zhu
- Kunming Medical University, Guizhou Province, Kunming, 650032, China
| | - Zhixu He
- Guizhou Medical University, Tissue Engineering and Stem Cell Experimental Center, Guizhou Provinc, Guiyang, 550004, China
| | - Xiangqing Zhu
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China.
| | - Xinghua Pan
- The Basic Medical Laboratory of the 920th Hospital of Joint Logistics Support Force of PLA, The Transfer Medicine Key Laboratory of Cell Therapy Technology of Yunan Province, The Integrated Engineering Laboratory of Cell Biological Medicine of State and Regions, Kunming, 650032, Yunnan Province, China.
- Guizhou Medical University, Tissue Engineering and Stem Cell Experimental Center, Guizhou Provinc, Guiyang, 550004, China.
- Kunming Medical University, Guizhou Province, Kunming, 650032, China.
| |
Collapse
|
13
|
Qu J, Li Y, Liao S, Yan J. The Effects of Negative Elements in Environment and Cancer on Female Reproductive System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1300:283-313. [PMID: 33523439 DOI: 10.1007/978-981-33-4187-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
With the development of human society, factors that contribute to the impairment of female fertility is accumulating. Lifestyle-related risk factors, occupational risk factors, and iatrogenic factors, including cancer and anti-cancer treatments, have been recognized with their negative effects on the function of female reproductive system. However, the exact influences and their possible mechanism have not been elucidated yet. It is impossible to accurately estimate the indexes of female fertility, but many researchers have put forward that the general fertility has inclined through the past decades. Thus the demand for fertility preservation has increased more and more dramatically. Here we described some of the factors which may influence female reproductive system and methods for fertility preservation in response to female infertility.
Collapse
Affiliation(s)
- Jiangxue Qu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yuehan Li
- Department of Gynaecology and Obstetrics, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shujie Liao
- Department of Gynaecology and Obstetrics, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Jie Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
14
|
Tandulwadkar S, Karthick MS. Combined Use of Autologous Bone Marrow-derived Stem Cells and Platelet-rich Plasma for Ovarian Rejuvenation in Poor Responders. J Hum Reprod Sci 2020; 13:184-190. [PMID: 33311903 PMCID: PMC7727891 DOI: 10.4103/jhrs.jhrs_130_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 04/26/2020] [Indexed: 12/30/2022] Open
Abstract
Background: The management of poor responders is still a challenge in modern-assisted reproductive technology. Several researches are showing encouraging results with autologous bone marrow-derived stem cells (ABMDSCs) and platelet-rich plasma (PRP) individually. Hence, we decided to study the synergistic effect of ABMDSCs with PRP. Aims and Objective: The aim of the study was to assess the safety and efficacy of intraovarian instillation of ABMDSCs combined with PRP in poor responders. Design: This was an interventional pilot study. Study Period: January 2017 to January 2019. Materials and Methods: We designed a pilot study using Patient-oriented Strategies Encompassing IndividualizeD Oocyte Number (POSEIDON) Group 3 and 4 poor responder patients (n = 20). The study group underwent laparoscopic/transvaginal intraovarian instillation of ABMDSCs combined with PRP and the outcome was analyzed – primary outcome – antral follicular count (AFC) and mature MII oocytes and secondary outcome – Anti-Mullerian hormone (AMH) levels and number of Grade A and B embryos frozen on day 3. The Wilcoxon signed-rank test and Pearson correlation were used for the statistical analysis and P < 0.05 was considered statistically significant. Results: After 6 weeks of intraovarian instillation ABMDSCs mixed with PRP, patients were reassessed for AFC and AMH and their response to subsequent controlled ovarian stimulation (COS) cycle was observed. Statistically significant improvement was seen in AFC, MII oocytes, and Grade A and Grade B embryos. AMH was also increased in some patients, but the result was not statistically significant. Conclusion: Our results suggest that intraovarian instillation of ABMDSCs combined with PRP is safe and it optimized the recruitment of existing dormant primordial follicles to improve oocyte yield and hence the number and quality of embryos after COS in POSEIDON Group 3 and 4 poor responders.
Collapse
Affiliation(s)
- Sunita Tandulwadkar
- Ruby Hall IVF and Endoscopy Center, Ruby Hall Clinic and Solo Stem Cells, Pune, Maharashtra, India
| | - M Selva Karthick
- Ruby Hall IVF and Endoscopy Center, Ruby Hall Clinic and Solo Stem Cells, Pune, Maharashtra, India
| |
Collapse
|
15
|
Cho HW, Lee S, Min KJ, Hong JH, Song JY, Lee JK, Lee NW, Kim T. Advances in the Treatment and Prevention of Chemotherapy-Induced Ovarian Toxicity. Int J Mol Sci 2020; 21:E7792. [PMID: 33096794 PMCID: PMC7589665 DOI: 10.3390/ijms21207792] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 10/20/2020] [Indexed: 12/24/2022] Open
Abstract
Due to improvements in chemotherapeutic agents, cancer treatment efficacy and cancer patient survival rates have greatly improved, but unfortunately gonadal damage remains a major complication. Gonadotoxic chemotherapy, including alkylating agents during reproductive age, can lead to iatrogenic premature ovarian insufficiency (POI), and loss of fertility. In recent years, the demand for fertility preservation has increased dramatically among female cancer patients. Currently, embryo and oocyte cryopreservation are the only established options for fertility preservation in women. However, there is growing evidence for other experimental techniques including ovarian tissue cryopreservation, oocyte in vitro maturation, artificial ovaries, stem cell technologies, and ovarian suppression. To prevent fertility loss in women with cancer, individualized fertility preservation options including established and experimental techniques that take into consideration the patient's age, marital status, chemotherapy regimen, and the possibility of treatment delay should be provided. In addition, effective multidisciplinary oncofertility strategies that involve a highly skilled and experienced oncofertility team consisting of medical oncologists, gynecologists, reproductive biologists, surgical oncologists, patient care coordinators, and research scientists are necessary to provide cancer patients with high-quality care.
Collapse
Affiliation(s)
| | - Sanghoon Lee
- Department of Obstetrics and Gynecology, Korea University College of Medicine, Seoul 02841, Korea; (H.-W.C.); (K.-J.M.); (J.H.H.); (J.Y.S.); (J.K.L.); (N.W.L.); (T.K.)
| | | | | | | | | | | | | |
Collapse
|
16
|
Dolmans MM, Donnez J. Fertility preservation in women for medical and social reasons: Oocytes vs ovarian tissue. Best Pract Res Clin Obstet Gynaecol 2020; 70:63-80. [PMID: 32800711 DOI: 10.1016/j.bpobgyn.2020.06.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 06/13/2020] [Indexed: 01/18/2023]
Abstract
Approximately 10% of cancers occur in women under 45 years of age. Chemotherapy, radiotherapy, and bone marrow transplantation cure more than 90% of cancer in women, but can result in premature ovarian insufficiency depending on follicular reserve, age, and drugs used. Some benign diseases are also indications for fertility preservation, particularly those requiring chemotherapy (like thalassemia and lupus), recurrent endometriosis, and family history of premature menopause. Social reasons also account for a large proportion of women who wish to postpone pregnancy. This article discusses the two main strategies for fertility preservation, namely oocyte vitrification and ovarian tissue cryopreservation, examining the indications and results of these options. Oocyte cryopreservation is an effective approach, but further studies are needed in cancer patients to ensure the excellent outcomes obtained in women without cancer or in egg donation programs. For prepubertal girls or cases where immediate therapy is required, cryopreservation of ovarian tissue is the only available option.
Collapse
Affiliation(s)
- Marie-Madeleine Dolmans
- Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvain, 1200, Brussels, Belgium; Gynecology Department, Cliniques Universitaires Saint Luc, Avenue Hippocrate 10, 1200, Brussels, Belgium.
| | - Jacques Donnez
- Université Catholique de Louvain, Belgium; Société de Recherche pour l'Infertilité (SRI), 143 Avenue Grandchamp, 1150, Brussels, Belgium.
| |
Collapse
|
17
|
Na J, Kim GJ. Recent trends in stem cell therapy for premature ovarian insufficiency and its therapeutic potential: a review. J Ovarian Res 2020; 13:74. [PMID: 32576209 PMCID: PMC7313218 DOI: 10.1186/s13048-020-00671-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
Abstract
Stem cell therapy is attracting attention in the field of regenerative medicine and is advancing rapidly. Many recent studies have applied stem cell therapy to treat reproductive system diseases; however, data are not yet available as to whether this therapy shows enhanced therapeutic effects. This paper analyzes recent preclinical studies on stem cell therapy for ovarian dysfunction in several types of animal models. Several clinical trials and pending projects are also discussed. This review will provide a background for developing stem cell therapies to enhance ovarian function.
Collapse
Affiliation(s)
- Jeeyoon Na
- Department of Biology, College of Arts & Sciences, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gi Jin Kim
- Department of Biomedical Science, CHA University, Seongnam, 13488, Republic of Korea.
| |
Collapse
|
18
|
Plasticity in Ovarian Cancer: The Molecular Underpinnings and Phenotypic Heterogeneity. J Indian Inst Sci 2020. [DOI: 10.1007/s41745-020-00174-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
19
|
Restoration of estrous cycles by co-transplantation of mouse ovarian tissue with MSCs. Cell Tissue Res 2020; 381:509-525. [PMID: 32424509 DOI: 10.1007/s00441-020-03204-x] [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: 07/04/2019] [Accepted: 03/12/2020] [Indexed: 12/13/2022]
Abstract
This study investigates the effect of bone marrow (BM-MSCs) and visceral peritoneum (VP-MSCs)-derived mesenchymal stem cells on the transplanted ovary. VP-MSCs and BM-MSCs were obtained from green fluorescent protein-expressing mice (GFP+). Six- to eight-week-old female NMRI mice were divided into four experimental groups, autograft ovarian tissue fragments (AO), autograft ovarian tissue fragments encapsulated in fibrin-collagen hydrogel (AO-H), autograft ovarian tissue fragments encapsulated in fibrin-collagen hydrogel containing BM-MSCs (AO-HB) and autograft ovarian tissue fragments encapsulated in fibrin-collagen hydrogel containing VP-MSCs (AO-HP). Intact ovary (IO) was the control group. The estrous cycles resumption time was monitored and at the third estrous cycle, the blood samples and grafted ovaries were evaluated using hormonal, histological and gene expression analysis. Onset of estrous cycles, especially at the second cycle, was earlier in AO-HB and AO-HP groups than in the AO-H group (P < 0.05). Moreover, E2 and FSH levels in AO-HB and AO-HP groups were returned to those of the intact group. However, folliculogenesis was still retarded as compared with the IO group. The gene expression of theca (Lhcgr, Cyp17a1, Gli2, Gli3 and Ptch1), granulosa (Amh and Fshr), oocyte (Zp3 and Gdf9), germ cells (Stella and Prdm1), angiogenesis (VEGF and bFGF) and apoptosis (Bax/Bcl2 and Caspase3) markers was similar in both AO-HB and AO-HP groups. Expression of Amh, Fshr, Gdf9 and VEGF increased only in the AO-HP group whereas expression of Ptch1 increased only in the AO-HB group, as compared with the AO group (P < 0.05). In conclusion, BM-MSCs or VP-MSCs can improve ovarian autotransplantation in mice with no superiority over each other.
Collapse
|
20
|
Hummitzsch K, Hatzirodos N, Macpherson AM, Schwartz J, Rodgers RJ, Irving-Rodgers HF. Transcriptome analyses of ovarian stroma: tunica albuginea, interstitium and theca interna. Reproduction 2020; 157:545-565. [PMID: 30925461 DOI: 10.1530/rep-18-0323] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 03/29/2019] [Indexed: 01/15/2023]
Abstract
The ovary has specialised stromal compartments, including the tunica albuginea, interstitial stroma and theca interna, which develops concurrently with the follicular antrum. To characterise the molecular determinants of these compartments, stroma adjacent to preantral follicles (pre-theca), interstitium and tunica albuginea were laser microdissected (n = 4 per group) and theca interna was dissected from bovine antral follicles (n = 6). RNA microarray analysis showed minimal differences between interstitial stroma and pre-theca, and these were combined for some analyses and referred to as stroma. Genes significantly upregulated in theca interna compared to stroma included INSL3, LHCGR, HSD3B1, CYP17A1, ALDH1A1, OGN, POSTN and ASPN. Quantitative RT-PCR showed significantly greater expression of OGN and LGALS1 in interstitial stroma and theca interna versus tunica and greater expression of ACD in tunica compared to theca interna. PLN was significantly higher in interstitial stroma compared to tunica and theca. Ingenuity pathway, network and upstream regulator analyses were undertaken. Cell survival was also upregulated in theca interna. The tunica albuginea was associated with GPCR and cAMP signalling, suggesting tunica contractility. It was also associated with TGF-β signalling and increased fibrous matrix. Western immunoblotting was positive for OGN, LGALS1, ALDH1A1, ACD and PLN with PLN and OGN highly expressed in tunica and interstitial stroma (each n = 6), but not in theca interna from antral follicles (n = 24). Immunohistochemistry localised LGALS1 and POSTN to extracellular matrix and PLN to smooth muscle cells. These results have identified novel differences between the ovarian stromal compartments.
Collapse
Affiliation(s)
- Katja Hummitzsch
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Nicholas Hatzirodos
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Anne M Macpherson
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Jeff Schwartz
- School of Medicine, Griffith University, Gold Coast, Queensland, Australia
| | - Raymond J Rodgers
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia
| | - Helen F Irving-Rodgers
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, South Australia, Australia.,School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
21
|
Human Ovarian Cortex biobanking: A Fascinating Resource for Fertility Preservation in Cancer. Int J Mol Sci 2020; 21:ijms21093245. [PMID: 32375324 PMCID: PMC7246700 DOI: 10.3390/ijms21093245] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/21/2022] Open
Abstract
Novel anti-cancer treatments have improved the survival rates of female young patients, reopening pregnancy issues for female cancer survivors affected by the tumor treatment-related infertility. This condition occurs in approximately one third of women of fertile age and is mainly dependent on gonadotoxic protocols, including radiation treatments. Besides routine procedures such as the hormonal induction of follicular growth and subsequent cryopreservation of oocytes or embryos, the ovarian protection by gonadotropin-releasing hormone (GnRH) agonists during chemotherapy as well as even gonadal shielding during radiotherapy, other innovative techniques are available today and need to be optimized to support their introduction into the clinical practice. These novel methods are hormone stimulation-free and include the ovarian cortex cryopreservation before anti-cancer treatments and its subsequent autologous reimplantation and a regenerative medicine approach using oocytes derived in vitro from ovarian stem cells (OSCs). For both procedures, the major benefit is related to the prompt recruitment and processing of the ovarian cortex fragments before gonadotoxic treatments. However, while the functional competence of oocytes within the cryopreserved cortex is not assessable, the in vitro maturation of OSCs to oocytes, allows to select the most competent eggs to be cryopreserved for fertility restoration.
Collapse
|
22
|
Bothun AM, Woods DC. Inherent mitochondrial activity influences specification of the germ line in pluripotent stem cells. Heliyon 2020; 6:e03651. [PMID: 32258510 PMCID: PMC7118317 DOI: 10.1016/j.heliyon.2020.e03651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 01/12/2020] [Accepted: 03/19/2020] [Indexed: 12/14/2022] Open
Abstract
Herein we investigated whether inherent differences in mitochondrial activity in mouse pluripotent cells could be used to identify populations with an intrinsic ability to differentiate into primordial germ cells (PGCs). Notably, we determined that stem cells sorted based on differences in mitochondrial membrane activity exhibited altered germline differentiation capacity, with low-mitochondrial membrane potential associated with an increase in PGC-like cells. This specification was not further enhanced by hypoxia. We additionally noted differences between these populations in metabolism, transcriptome, and cell-cycle. These data contribute to a growing body of work demonstrating that pluripotent cells exhibit a large range of mitochondrial activity, which impacts cellular function and differentiation potential. Furthermore, pluripotent cells possess a subpopulation of cells with an improved ability to differentiate into the germ lineage that can be identified based on differences in mitochondrial membrane potential.
Collapse
Affiliation(s)
- Alisha M Bothun
- Department of Biology, Laboratory for Aging and Infertility Research, Northeastern University, Boston, MA 02115, USA
| | - Dori C Woods
- Department of Biology, Laboratory for Aging and Infertility Research, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
23
|
Laisk T, Tšuiko O, Jatsenko T, Hõrak P, Otala M, Lahdenperä M, Lummaa V, Tuuri T, Salumets A, Tapanainen JS. Demographic and evolutionary trends in ovarian function and aging. Hum Reprod Update 2020; 25:34-50. [PMID: 30346539 DOI: 10.1093/humupd/dmy031] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/03/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The human female reproductive lifespan is regulated by the dynamics of ovarian function, which in turn is influenced by several factors: from the basic molecular biological mechanisms governing folliculogenesis, to environmental and lifestyle factors affecting the ovarian reserve between conception and menopause. From a broader point of view, global and regional demographic trends play an additional important role in shaping the female reproductive lifespan, and finally, influences on an evolutionary scale have led to the reproductive senescence that precedes somatic senescence in humans. OBJECTIVE AND RATIONALE The narrative review covers reproductive medicine, by integrating the molecular mechanisms of ovarian function and aging with short-term demographic and long-term evolutionary trends. SEARCH METHODS PubMed and Google Scholar searches were performed with relevant keywords (menopause, folliculogenesis, reproductive aging, reproductive lifespan and life history theory). The reviewed articles and their references were restricted to those written in English. OUTCOMES We discuss and summarize the rapidly accumulating information from large-scale population-based and single-reproductive-cell genomic studies, their constraints and advantages in the context of female reproductive aging as well as their possible evolutionary significance on the life history trajectory from foetal-stage folliculogenesis until cessation of ovarian function in menopause. The relevant environmental and lifestyle factors and demographic trends are also discussed in the framework of predominant evolutionary hypotheses explaining the origin and maintenance of menopause. WIDER IMPLICATIONS The high speed at which new data are generated has so far raised more questions than it has provided solid answers and has been paralleled by a lack of satisfactory interpretations of the findings in the context of human life history theory. Therefore, the recent flood of data could offer an unprecedented tool for future research to possibly confirm or rewrite human evolutionary reproductive history, at the same time providing novel grounds for patient counselling and family planning strategies.
Collapse
Affiliation(s)
- Triin Laisk
- Competence Centre on Health Technologies, Tiigi 61b, Tartu, Estonia.,Institute of Clinical Medicine, Department of Obstetrics and Gynaecology, University of Tartu, L. Puusepa 8, Tartu, Estonia
| | - Olga Tšuiko
- Competence Centre on Health Technologies, Tiigi 61b, Tartu, Estonia.,Institute of Biomedicine and Translational Medicine, Department of Biomedicine, University of Tartu, Ravila 19, Tartu, Estonia
| | - Tatjana Jatsenko
- Competence Centre on Health Technologies, Tiigi 61b, Tartu, Estonia
| | - Peeter Hõrak
- Department of Zoology, University of Tartu, Vanemuise 46, Tartu, Estonia
| | - Marjut Otala
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, Helsinki, Finland
| | - Mirkka Lahdenperä
- Department of Biology, University of Turku, Turun yliopisto, Turku, Finland
| | - Virpi Lummaa
- Department of Biology, University of Turku, Turun yliopisto, Turku, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, Helsinki, Finland
| | - Andres Salumets
- Competence Centre on Health Technologies, Tiigi 61b, Tartu, Estonia.,Institute of Clinical Medicine, Department of Obstetrics and Gynaecology, University of Tartu, L. Puusepa 8, Tartu, Estonia.,Institute of Biomedicine and Translational Medicine, Department of Biomedicine, University of Tartu, Ravila 19, Tartu, Estonia.,Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, Helsinki, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 2, Helsinki, Finland.,Department of Obstetrics and Gynecology, University Hospital of Oulu, University of Oulu, Medical Research Center Oulu and PEDEGO Research Unit, OYS Oulu, Finland
| |
Collapse
|
24
|
Silvestris E, D’Oronzo S, Cafforio P, Kardhashi A, Dellino M, Cormio G. In Vitro Generation of Oocytes from Ovarian Stem Cells (OSCs): In Search of Major Evidence. Int J Mol Sci 2019; 20:ijms20246225. [PMID: 31835581 PMCID: PMC6940822 DOI: 10.3390/ijms20246225] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/04/2019] [Accepted: 12/07/2019] [Indexed: 02/07/2023] Open
Abstract
The existence of ovarian stem cells (OSCs) in women as well as their physiological role in post-menopausal age are disputed. However, accumulating evidence demonstrated that, besides the animal models including primarily mice, even in adult women putative OSCs obtained from ovarian cortex are capable to differentiate in vitro into oocyte-like cells (OLCs) expressing molecular markers typical of terminal stage of oogonial cell lineage. Recent studies describe that, similarly to mature oocytes, the OSC-derived OLCs also contain haploid karyotype. As proof of concept of their stem commitment, OSCs from mice differentiated to oocytes in vitro are suitable to be fertilized and implanted in sterilized animals resulting in embryo development. Despite enthusiasm for these data, which definitely require extended confirmation before considering potential application in humans for treatment of ovarian insufficiency, OSCs appear suitable for other clinical uses, restoring the endocrine derangements in premature ovarian failure or for fertility preservation in oncologic patients after anti-cancer treatments. In this context, the selection of viable oocytes generated from OSCs before chemotherapy protocols would overcome the potential adjunct oncogenic risk in women bearing hormone-dependent tumors who are repeatedly stimulated with high dose estrogens to induce oocyte maturation for their egg recruitment and cryopreservation.
Collapse
Affiliation(s)
- Erica Silvestris
- Gynecologic Oncology Unit, National Cancer Center, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (A.K.); (M.D.); (G.C.)
- Correspondence:
| | - Stella D’Oronzo
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, 70124 Bari, Italy; (S.D.); (P.C.)
- National Cancer Center, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy
| | - Paola Cafforio
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro, 70124 Bari, Italy; (S.D.); (P.C.)
| | - Anila Kardhashi
- Gynecologic Oncology Unit, National Cancer Center, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (A.K.); (M.D.); (G.C.)
| | - Miriam Dellino
- Gynecologic Oncology Unit, National Cancer Center, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (A.K.); (M.D.); (G.C.)
| | - Gennaro Cormio
- Gynecologic Oncology Unit, National Cancer Center, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (A.K.); (M.D.); (G.C.)
- Department of Biomedical Sciences and Human Oncology, Unit of Obstetrics and Gynecology, University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
25
|
Salama M, Anazodo A, Woodruff TK. Preserving fertility in female patients with hematological malignancies: a multidisciplinary oncofertility approach. Ann Oncol 2019; 30:1760-1775. [PMID: 31418765 DOI: 10.1093/annonc/mdz284] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
Oncofertility is a new interdisciplinary field at the intersection of oncology and reproductive medicine that expands fertility options for young cancer patients. The most common forms of hematological malignancies that occur in girls and young women and therefore necessitate oncofertility care are acute lymphocytic leukemia, acute myeloid leukemia, non-Hodgkin's lymphoma, and Hodgkin's lymphoma. Aggressive gonadotoxic anticancer regimens including alkylating chemotherapy and total body irradiation are used often in treating girls and young women with hematological malignancies. The risks of gonadotoxicity and subsequent iatrogenic premature ovarian insufficiency and fertility loss depend mainly on the type and stage of the disease, dose of anticancer therapy as well as the age of the patient at the beginning of treatment. To avoid or at least mitigate the devastating complications of anticancer therapy-induced gonadotoxicity, effective and comprehensive strategies that integrate different options for preserving and restoring fertility ranging from established to experimental strategies should be offered before, during, and after chemotherapy or radiotherapy. A multidisciplinary approach that involves strong coordination and collaboration between hemato-oncologists, gynecologists, reproductive biologists, research scientists, and patient navigators is essential to guarantee high standard of care.
Collapse
Affiliation(s)
- M Salama
- Department of Obstetrics and Gynecology, Feinberg School of Medicine-Northwestern University, Chicago, USA
| | - A Anazodo
- Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia; Nelune Cancer Centre, Prince of Wales Hospital, Sydney, Australia; School of Women's and Children's Health, University of New South Wales, Sydney, Australia
| | - T K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine-Northwestern University, Chicago, USA.
| |
Collapse
|
26
|
Insights into the Pathophysiology of Infertility in Females with Classical Galactosaemia. Int J Mol Sci 2019; 20:ijms20205236. [PMID: 31652573 PMCID: PMC6834160 DOI: 10.3390/ijms20205236] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/14/2019] [Accepted: 10/16/2019] [Indexed: 11/26/2022] Open
Abstract
Classical galactosaemia (CG) (OMIM 230400) is a rare inborn error of galactose metabolism caused by the deficiency of the enzyme galactose-1-phosphate uridylyltransferase (GALT, EC 2.7.7.12). Primary ovarian insufficiency (POI) is the most common long-term complication experienced by females with CG, presenting with hypergonadotrophic hypoestrogenic infertility affecting at least 80% of females despite new-born screening and lifelong galactose dietary restriction. In this review, we describe the hypothesized pathophysiology of POI from CG, implications of timing of the ovarian dysfunction, and the new horizons and future prospects for treatments and fertility preservation.
Collapse
|
27
|
Guo H, Du X, Zhang Y, Wu J, Wang C, Li M, Hua X, Zhang XA, Yan J. Specific miRNA-G Protein-Coupled Receptor Networks Regulate Sox9a/Sox9b Activities to Promote Gonadal Rejuvenation in Zebrafish. Stem Cells 2019; 37:1189-1199. [DOI: 10.1002/stem.3040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 04/17/2019] [Accepted: 05/04/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Huiping Guo
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Xinlu Du
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Ying Zhang
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Jiacheng Wu
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
| | - Chenghui Wang
- Department of Aquaculture; Shanghai Ocean University; Lingang New City, Shanghai People's Republic of China
| | - Mingyou Li
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources; Ministry of Education; Shanghai People's Republic of China
| | - Xianxin Hua
- Department of Cancer Biology; University of Pennsylvania Perelman School of Medicine; Philadelphia, Pennsylvania USA
| | - Xin A. Zhang
- Stephenson Cancer Center and Department of Physiology; The University of Oklahoma Health Sciences Center; Oklahoma City Oklahoma USA
| | - Jizhou Yan
- Department of Developmental Biology; Institute for Marine Biosystem and Neurosciences; People's Republic of China
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources; Ministry of Education; Shanghai People's Republic of China
| |
Collapse
|
28
|
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
|
29
|
Tammiste T, Kask K, Padrik P, Idla K, Rosenstein K, Jatsenko T, Veerus P, Salumets A. A case report and follow-up of the first live birth after heterotopic transplantation of cryopreserved ovarian tissue in Eastern Europe. BMC WOMENS HEALTH 2019; 19:65. [PMID: 31088441 PMCID: PMC6518657 DOI: 10.1186/s12905-019-0764-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 05/07/2019] [Indexed: 02/06/2023]
Abstract
Background Ovarian insufficiency is a major concern for long-term cancer survivors. Although semen freezing is well established to preserve male fertility, the possibilities to secure post-cancer female fertility are mostly limited to oocyte or embryo freezing. These methods require time-consuming ovarian stimulation with or without in vitro fertilization (IVF) that evidently delays cancer therapy. Ovarian tissue cryopreservation and subsequent thawed tissue autotransplantation are considered the most promising alternative strategy for restoring the fertility of oncology patients, which has not yet received the full clinical acceptance. Therefore, all successful cases are needed to prove its reliability and safety. Case presentation Here we report a single case in Estonia, where a 28-year-old woman with malignant breast neoplasm had ovarian cortex cryopreserved before commencing gonadotoxic chemo- and radiotherapy. Two years after cancer therapy, the patient underwent heterotopic ovarian tissue transplantation into the lateral pelvic wall. The folliculogenesis was stimulated in the transplanted tissue by exogenous follicle-stimulating hormone and oocytes were collected under ultrasound guidance for IVF and embryo transfer. The healthy boy was born after full-term gestation in 2014, first in Eastern Europe. Conclusion Despite many countries have reported the first implementation of the ovarian tissue freezing and transplantation protocols, the data is still limited on the effectiveness of heterotopic ovarian transplant techniques. Thus, all case reports of heterotopic ovarian tissue transplantation and long-term follow-ups to describe the children’s health are valuable source of clinical experience.
Collapse
Affiliation(s)
- Triin Tammiste
- West Tallinn Central Hospital Women's Clinic, Tallinn, Estonia.,Internal Medicine Clinic, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Keiu Kask
- Women's Clinic, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Peeter Padrik
- Cancer Centre of Tartu University Hospital, Tartu, Estonia.,Department of Hematology and Oncology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Külli Idla
- West Tallinn Central Hospital Women's Clinic, Tallinn, Estonia
| | | | | | - Piret Veerus
- West Tallinn Central Hospital Women's Clinic, Tallinn, Estonia
| | - Andres Salumets
- Women's Clinic, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia. .,Competence Centre on Health Technologies, Tartu, Estonia. .,Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia. .,Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
30
|
A report on three live births in women with poor ovarian response following intra-ovarian injection of platelet-rich plasma (PRP). Mol Biol Rep 2019; 46:1611-1616. [DOI: 10.1007/s11033-019-04609-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/16/2019] [Indexed: 11/27/2022]
|
31
|
Tomaszewski CE, Constance E, Lemke MM, Zhou H, Padmanabhan V, Arnold KB, Shikanov A. Adipose-derived stem cell-secreted factors promote early stage follicle development in a biomimetic matrix. Biomater Sci 2019; 7:571-580. [PMID: 30608082 PMCID: PMC6351215 DOI: 10.1039/c8bm01253a] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Development of primary follicles in vitro benefits from a three-dimensional matrix that is enriched with paracrine factors secreted from feeder cells and mimics the in vivo environment. In this study, we investigated the role of paracrine signaling from adipose-derived stem cells (ADSCs) in supporting primary follicle development in a biomimetic poly(ethylene glycol) (PEG)-based matrix. Follicles co-cultured with ADSCs and follicles cultured in conditioned medium from ADSCs encapsulated in gels (3D CM) exhibited significantly (p < 0.01 and p = 0.09, respectively) improved survival compared to follicles cultured in conditioned medium collected from ADSCs cultured in flasks (2D CM) and follicles cultured without paracrine support. The gene expression of ADSCs suggested that the stem cells maintained their multipotency in the 3D PEG environment over the culture period, regardless of the presence of the follicles, while under 2D conditions the multipotency markers were downregulated. The differences in cytokine signatures of follicles exposed to 3D and 2D ADSC paracrine factors suggest that early cytokine interactions are key for follicle survival. Taken together, the biomimetic PEG scaffold provides a three-dimensional, in vivo-like environment to induce ADSCs to secrete factors which promote early stage ovarian follicle development and survival.
Collapse
|
32
|
Martin JJ, Woods DC, Tilly JL. Implications and Current Limitations of Oogenesis from Female Germline or Oogonial Stem Cells in Adult Mammalian Ovaries. Cells 2019; 8:E93. [PMID: 30696098 PMCID: PMC6407002 DOI: 10.3390/cells8020093] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/16/2019] [Indexed: 12/15/2022] Open
Abstract
A now large body of evidence supports the existence of mitotically active germ cells in postnatal ovaries of diverse mammalian species, including humans. This opens the possibility that adult stem cells naturally committed to a germline fate could be leveraged for the production of female gametes outside of the body. The functional properties of these cells, referred to as female germline or oogonial stem cells (OSCs), in ovaries of women have recently been tested in various ways, including a very recent investigation of the differentiation capacity of human OSCs at a single cell level. The exciting insights gained from these experiments, coupled with other data derived from intraovarian transplantation and genetic tracing analyses in animal models that have established the capacity of OSCs to generate healthy eggs, embryos and offspring, should drive constructive discussions in this relatively new field to further exploring the value of these cells to the study, and potential management, of human female fertility. Here, we provide a brief history of the discovery and characterization of OSCs in mammals, as well as of the in-vivo significance of postnatal oogenesis to adult ovarian function. We then highlight several key observations made recently on the biology of OSCs, and integrate this information into a broader discussion of the potential value and limitations of these adult stem cells to achieving a greater understanding of human female gametogenesis in vivo and in vitro.
Collapse
Affiliation(s)
- Jessica J Martin
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA 02115, USA.
| | - Dori C Woods
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA 02115, USA.
| | - Jonathan L Tilly
- Laboratory of Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Ubaldi FM, Cimadomo D, Vaiarelli A, Fabozzi G, Venturella R, Maggiulli R, Mazzilli R, Ferrero S, Palagiano A, Rienzi L. Advanced Maternal Age in IVF: Still a Challenge? The Present and the Future of Its Treatment. Front Endocrinol (Lausanne) 2019; 10:94. [PMID: 30842755 PMCID: PMC6391863 DOI: 10.3389/fendo.2019.00094] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 02/01/2019] [Indexed: 12/13/2022] Open
Abstract
Advanced maternal age (AMA; >35 year) is associated with a decline in both ovarian reserve and oocyte competence. At present, no remedies are available to counteract the aging-related fertility decay, however different therapeutic approaches can be offered to women older than 35 year undergoing IVF. This review summarizes the main current strategies proposed for the treatment of AMA: (i) oocyte cryopreservation to conduct fertility preservation for medical reasons or "social freezing" for non-medical reasons, (ii) personalized controlled ovarian stimulation to maximize the exploitation of the ovarian reserve in each patient, (iii) enhancement of embryo selection via blastocyst-stage preimplantation genetic testing for aneuploidies and frozen single embryo transfer, or (iv) oocyte donation in case of minimal/null residual chance of pregnancy. Future strategies and tools are in the pipeline that might minimize the risks of AMA through non-invasive approaches for embryo selection (e.g., molecular analyses of leftover products of IVF, such as spent culture media). These are yet challenging but potentially ground-breaking perspectives promising a lower clinical workload with a higher cost-effectiveness. We also reviewed emerging experimental therapeutic approaches to attempt at restoring maternal reproductive potential, e.g., spindle-chromosomal complex, pronuclear or mitochondrial transfer, and chromosome therapy. In vitro generation of gametes is also an intriguing challenge for the future. Lastly, since infertility is a social issue, social campaigns, and education among future generations are desirable to promote the awareness of the impact of age and lifestyle habits upon fertility. This should be a duty of the clinical operators in this field.
Collapse
Affiliation(s)
- Filippo Maria Ubaldi
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| | - Danilo Cimadomo
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
- *Correspondence: Danilo Cimadomo
| | - Alberto Vaiarelli
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| | - Gemma Fabozzi
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| | - Roberta Venturella
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Roberta Maggiulli
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| | - Rossella Mazzilli
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
- Andrology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Susanna Ferrero
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| | - Antonio Palagiano
- Department of Gynecological, Obstetrical and Reproductive Sciences, University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Laura Rienzi
- Clinica Valle Giulia, G.en.e.r.a. Centers for Reproductive Medicine, Rome, Italy
| |
Collapse
|
34
|
Premature Ovarian Insufficiency: Procreative Management and Preventive Strategies. Biomedicines 2018; 7:biomedicines7010002. [PMID: 30597834 PMCID: PMC6466184 DOI: 10.3390/biomedicines7010002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/22/2018] [Accepted: 12/24/2018] [Indexed: 02/08/2023] Open
Abstract
Premature ovarian insufficiency (POI) is the loss of normal hormonal and reproductive function of ovaries in women before age 40 as the result of premature depletion of oocytes. The incidence of POI increases with age in reproductive-aged women, and it is highest in women by the age of 40 years. Reproductive function and the ability to have children is a defining factor in quality of life for many women. There are several methods of fertility preservation available to women with POI. Procreative management and preventive strategies for women with or at risk for POI are reviewed.
Collapse
|
35
|
Genes responsible for proliferation, differentiation, and junction adhesion are significantly up-regulated in human ovarian granulosa cells during a long-term primary in vitro culture. Histochem Cell Biol 2018; 151:125-143. [PMID: 30382374 PMCID: PMC6394675 DOI: 10.1007/s00418-018-1750-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2018] [Indexed: 01/10/2023]
Abstract
The human ovarian granulosa cells (GCs) surround the oocyte and form the proper architecture of the ovarian follicle. The ability of GCs to proliferate and differentiate in the conditions of in vitro culture has been proven. However, there is still a large field for extensive investigation of molecular basics, as well as marker genes, responsible for these processes. This study aimed to find the new marker genes, encoding proteins that regulate human GCs in vitro capability for proliferation and differentiation during long-term primary culture. The human follicular GCs were collected from hyper-stimulated ovarian follicles during IVF procedures and transferred to a long-term in vitro culture. The culture lasted for 30 days, with RNA samples isolated at days 1, 7, 15, 30. Transcriptomic analysis was then performed with the use of Affymetrix microarray. Obtained results were then subjected to bioinformatical evaluation and sorting. After subjecting the datasets to KEGG analysis, three differentially expressed ontology groups "cell differentiation" (GO:0030154), "cell proliferation" (GO:0008283) and "cell-cell junction organization" (GO:0045216) were chosen for further investigation. All three of those ontology groups are involved in human GCs' in vitro lifespan, proliferation potential, and survival capability. Changes in expression of genes of interest belonging to the chosen GOs were validated with the use of RT-qPCR. In this manuscript, we suggest that VCL, PARVA, FZD2, NCS1, and COL5A1 may be recognized as new markers of GC in vitro differentiation, while KAT2B may be a new marker of their proliferation. Additionally, SKI, GLI2, FERMT2, and CDH2 could also be involved in GC in vitro proliferation and differentiation processes. We demonstrated that, in long-term in vitro culture, GCs exhibit markers that suggest their ability to differentiate into different cells types. Therefore, the higher expression profile of these genes may also be associated with the induction of cellular differentiation processes that take place beyond the long-term primary in vitro culture.
Collapse
|
36
|
Bothun AM, Woods DC. Dynamics of WNT signaling components in the human ovary from development to adulthood. Histochem Cell Biol 2018; 151:115-123. [PMID: 30284608 DOI: 10.1007/s00418-018-1729-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2018] [Indexed: 12/20/2022]
Abstract
WNT signaling has been shown to play a pivotal role in mammalian gonad development and sex differentiation; however, its role in the developing human ovary has not been investigated. We analyzed a quantitative mass spectrometry dataset to determine the expression of WNT signaling components between 47 and 137 days of development and in adult ovarian cortex tissue. WNT signaling was identified within the top ten canonical pathways of proteins detected at every developmental stage examined. We further examined the specific localization of WNT signaling components glycogen synthase kinase 3 (GSK3B), frizzled 2 (FZD2), and β-catenin (CTNNB1) within ovarian tissue. GSK3B was nearly ubiquitously expressed during fetal development, while FZD2 was specific to germ cell nests during early development. β-catenin exhibited translocation from primarily membrane bound during early ovarian development to cytoplasmic and nuclear staining specifically in early primordial follicles in the fetal ovary. This cytoplasmic and nuclear β-catenin persisted in primordial follicles in adult ovarian tissue, but returned to membrane-bound localization in secondary follicles. We conclude that WNT signaling components are expressed in the human ovary from early to mid-gestation and remain in the adult ovary, and observed evidence for canonical WNT signaling only in the oocytes of primordial follicles. Together, these data are indicative of a role for canonical WNT signaling via β-catenin nuclear translocation during human follicle formation and follicle maintenance.
Collapse
Affiliation(s)
- Alisha M Bothun
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, 02115, USA
| | - Dori C Woods
- Laboratory for Aging and Infertility Research, Department of Biology, Northeastern University, Boston, MA, 02115, USA.
| |
Collapse
|
37
|
Orkhon B, Kobayashi K, Javzan B, Sasaki K. Astragalus root induces ovarian β‑oxidation and suppresses estrogen‑dependent uterine proliferation. Mol Med Rep 2018; 18:5198-5206. [PMID: 30272268 DOI: 10.3892/mmr.2018.9493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/26/2018] [Indexed: 11/06/2022] Open
Abstract
Continuous estrogen stimulation in the uterus has been known to cause excess proliferation of the functional layer of endometrium, resulting in endometrial hyperplasia and leading to infertility. Estrogens can modulate other nuclear receptor signaling pathways, such as peroxisome proliferator‑activated receptors (PPARs). Astragalus root (AsR) has exhibited strong PPARα agonistic activity. Female Imprinting Control Region mice were fed a powder diet that included 5% AsR hot water extract or 0.1% bezafibrate as a positive control for 56 days to investigate AsR effects on the reproductive tract, ovary and uterus. AsR resulted in upregulation of the expression of uterine and ovarian PPARα mRNA by 2.5‑fold, and 1.5‑fold, respectively, compared with controls. AsR significantly increased ovarian expression levels of mitochondrial 2,4‑dienoyl‑CoA reductase (mDECR), an auxiliary enzyme involved in β‑oxidation. AsR‑fed mice also exhibited a significant increase in blood estradiol levels and tended to have higher ovary weight. AsR resulted in significantly decreased uterine weight and mDECR expression levels. It has been reported that a PPARα agonist suppresses the development of estrogen‑dependent endometrial hyperplasia. These findings raise the possibility that AsR suppresses estrogen‑dependent endometrial hyperplasia and ovarian dysfunction leading to infertility.
Collapse
Affiliation(s)
- Banzragchgarav Orkhon
- Department of Pharmacognosy, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981‑8558, Japan
| | - Kyoko Kobayashi
- Department of Pharmacognosy, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981‑8558, Japan
| | - Batkhuu Javzan
- School of Engineering and Applied Sciences, National University of Mongolia, Ulaanbaatar 14201, Mongolia
| | - Kenroh Sasaki
- Department of Pharmacognosy, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi 981‑8558, Japan
| |
Collapse
|
38
|
Saadeldin IM, Swelum AAA, Elsafadi M, Mahmood A, Alfayez M, Alowaimer AN. Cumulus cells of camel (Camelus dromedarius) antral follicles are multipotent stem cells. Theriogenology 2018; 118:233-242. [PMID: 30100012 DOI: 10.1016/j.theriogenology.2018.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 05/14/2018] [Accepted: 06/17/2018] [Indexed: 01/10/2023]
Abstract
The mammalian ovary is a highly dynamic organ, in which proliferation and differentiation occur constantly during the entire life span, particularly in camels that are characterized by a follicular wave pattern and induced ovulation. Granulosa cells are the main cells of mature follicles. Two distinct cell types, namely, the mural and cumulus granulosa cells are distinguished on the basis of antral fluid increase. The multipotency of follicular fluid and the luteinizing cell were recently demonstrated. However, reports regarding the plasticity of cumulus cells are lacking. We obtained cumulus cells from cumulus-oocyte complexes and showed that camel cumulus cells expressed stem cell mRNA transcripts (POU5A1, KLF4, SOX2, and MYC) and were able to differentiate into other non-ovarian follicular cell types in vitro, such as neurons, osteoblasts, and adipocytes. In contrast, removal of the ooplasm (oocytectemy) showed no effect on cumulus cell proliferation and differentiation. This is the first report to identify an invaluable source of multipotent stem cells, which is routinely discarded during in vitro embryo production. The plasticity and transdifferentiation capability of camel cumulus cells definitely requires attention as it provides a cheap biological experimental model for basic research in stem cells and for understanding ovarian differentiation, both of which are relevant for use in regenerative medicine and tissue engineering in humans and animals.
Collapse
Affiliation(s)
- Islam M Saadeldin
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, 11451, Riyadh, Saudi Arabia; Department of Physiology, Faculty of Veterinary Medicine, Zagazig University, 44519, Zagazig, Egypt.
| | - Ayman Abdel-Aziz Swelum
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, 11451, Riyadh, Saudi Arabia; Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, 44519, Zagazig, Egypt
| | - Mona Elsafadi
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Amer Mahmood
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Musaad Alfayez
- Stem Cell Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Saudi Society for Camel Studies, Saudi Arabia
| | - Abdullah N Alowaimer
- Department of Animal Production, College of Food and Agricultural Sciences, King Saud University, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
39
|
Patel H, Bhartiya D, Parte S. Further characterization of adult sheep ovarian stem cells and their involvement in neo-oogenesis and follicle assembly. J Ovarian Res 2018; 11:3. [PMID: 29304868 PMCID: PMC5755409 DOI: 10.1186/s13048-017-0377-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 12/26/2017] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Stem cells in the ovary comprise of two distinct populations including very small embryonic-like stem cells (VSELs) and slightly bigger progenitors termed ovarian stem cells (OSCs). They are lodged in ovary surface epithelium (OSE) and are expected to undergo neo-oogenesis and primordial follicle (PF) assembly in adult ovaries. The ovarian stem cells express follicle stimulating hormone (FSH) receptors and are directly activated by FSH resulting in formation of germ cell nests (GCN) in vitro. Present study was undertaken to further characterize adult sheep OSCs and to understand their role during neo-oogenesis and PF assembly. METHODS Stem cells were collected by gently scraping the OSE cells and were characterized by H&E staining, immuno-localization, immuno-phenotyping and RT-PCR studies. Expression of FSH receptors and markers specific for stem cells (OCT-4, SSEA-4) and proliferation (PCNA) were studied on stem/progenitor cells in OSE culture and on adult sheep ovarian cortical tissue sections. Effect of FSH on stem cells was also studied in vitro. Asymmetric cell division (ACD) was monitored by studying expression of OCT-4 and NUMB. RESULTS Additional evidence was generated on the presence of two populations of stem cells in the OSE including VSELs and OSCs. FSHR expression was observed on both VSELs and OSCs by immuno-localization and immuno-phenotyping studies. FSH treatment in vitro stimulated VSELs that underwent ACD to self-renew and give rise to OSCs which divided rapidly by symmetric cell divisions (SCD) and clonal expansion with incomplete cytokinesis to form GCN. ACD was further confirmed by differential expression of OCT-4 in VSELs and NUMB in the OSCs. Immuno-histochemical expression of OCT-4, PCNA and FSHR was noted on stem cells located in the OSE in sheep ovarian sections. GCN and cohort of PF were observed in the ovarian cortex and provided evidence in support of neo-oogenesis from the stem cells. CONCLUSION Results of present study provide further evidence in support of two stem cells populations in adult sheep ovary. Both VSELs, OSCs and GCN express FSH receptors and FSH possibly regulates their function to undergo neo-oogenesis and primordial follicle assembly.
Collapse
Affiliation(s)
- Hiren Patel
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012 India
| | - Deepa Bhartiya
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012 India
| | - Seema Parte
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012 India
| |
Collapse
|
40
|
Affiliation(s)
- Jacques Donnez
- From Société de Recherche pour l'Infertilité and Université Catholique de Louvain (J.D.), and Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, and the Department of Gynecology, Cliniques Universitaires Saint-Luc (M.-M.D.) - all in Brussels
| | - Marie-Madeleine Dolmans
- From Société de Recherche pour l'Infertilité and Université Catholique de Louvain (J.D.), and Pôle de Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, and the Department of Gynecology, Cliniques Universitaires Saint-Luc (M.-M.D.) - all in Brussels
| |
Collapse
|
41
|
Thakur M, Feldman G, Puscheck EE. Primary ovarian insufficiency in classic galactosemia: current understanding and future research opportunities. J Assist Reprod Genet 2017; 35:3-16. [PMID: 28932969 DOI: 10.1007/s10815-017-1039-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 09/03/2017] [Indexed: 02/06/2023] Open
Abstract
Classic galactosemia is an inborn error of the metabolism with devastating consequences. Newborn screening has been successful in markedly reducing the acute neonatal symptoms from this disorder. The dramatic response to dietary treatment is one of the major success stories of newborn screening. However, as children with galactosemia achieve adulthood, they face long-term complications. A majority of women with classic galactosemia develop primary ovarian insufficiency and resulting morbidity. The underlying pathophysiology of this complication is not clear. This review focuses on the reproductive issues seen in girls and women with classic galactosemia. Literature on the effects of classic galactosemia on the female reproductive system was reviewed by an extensive Pubmed search (publications from January 1975 to January 2017) using the keywords: galactosemia, ovarian function/dysfunction, primary ovarian insufficiency/failure, FSH, oxidative stress, fertility preservation. In addition, articles cited in the search articles and literature known to the authors was also included in the review. Our understanding of the role of galactose metabolism in the ovary is limited and the pathogenic mechanisms involved in causing primary ovarian insufficiency are unclear. The relative rarity of galactosemia makes it difficult to accumulate data to determine factors defining timing of ovarian dysfunction or treatment/fertility preservation options for this group of women. In this review, we present reproductive challenges faced by women with classic galactosemia, highlight the gaps in our understanding of mechanisms leading to primary ovarian insufficiency in this population, discuss new advances in fertility preservation options, and recommend collaboration between reproductive medicine and metabolic specialists to improve fertility in these women.
Collapse
Affiliation(s)
- Mili Thakur
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, 48201, USA. .,Division of Genetic, Genomic and Metabolic Disorders, Department of Pediatrics and Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA. .,The Fertility Center, 3230 Eagle Park Dr. NE, Suite 100, Grand Rapids, MI, 49525, USA.
| | - Gerald Feldman
- Division of Genetic, Genomic and Metabolic Disorders, Department of Pediatrics and Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, 48201, USA.,Department of Pathology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Elizabeth E Puscheck
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, The C.S. Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| |
Collapse
|
42
|
Liu J, Shang D, Xiao Y, Zhong P, Cheng H, Zhou R. Isolation and characterization of string-forming female germline stem cells from ovaries of neonatal mice. J Biol Chem 2017; 292:16003-16013. [PMID: 28827310 DOI: 10.1074/jbc.m117.799403] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/19/2017] [Indexed: 12/30/2022] Open
Abstract
Germline stem cells are essential in the generation of both male and female gametes. In mammals, the male testis produces sperm throughout the entire lifetime, facilitated by testicular germline stem cells. Oocyte renewal ceases in postnatal or adult life in mammalian females, suggesting that germline stem cells are absent from the mammalian ovary. However, studies in mice, rats, and humans have recently provided evidence for ovarian female germline stem cells (FGSCs). A better understanding of the role of FGSCs in ovaries could help improve fertility treatments. Here, we developed a rapid and efficient method for isolating FGSCs from ovaries of neonatal mice. Notably, our FGSC isolation method could efficiently isolate on average 15 cell "strings" per ovary from mice at 1-3 days postpartum. FGSCs isolated from neonatal mice displayed the string-forming cell configuration at mitosis (i.e. a "stringing" FGSC (sFGSC) phenotype) and a disperse phenotype in postnatal mice. We also found that sFGSCs undergo vigorous mitosis especially at 1-3 days postpartum. After cell division, the sFGSC membranes tended to be connected to form sFGSCs. Moreover, F-actin filaments exhibited a cell-cortex distribution in sFGSCs, and E-cadherin converged in cell-cell connection regions, resulting in the string-forming morphology. Our new method provides a platform for isolating FGSCs from the neonatal ovary, and our findings indicate that FGCSs exhibit string-forming features in neonatal mice. The sFGSCs represent a valuable resource for analysis of ovary function and an in vitro model for future clinical use to address ovarian dysfunction.
Collapse
Affiliation(s)
- Jing Liu
- From the Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Dantong Shang
- From the Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yao Xiao
- From the Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Pei Zhong
- From the Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Hanhua Cheng
- From the Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Rongjia Zhou
- From the Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| |
Collapse
|
43
|
Restricting the induction of NGF in ovarian stroma engenders selective follicular activation through the mTOR signaling pathway. Cell Death Dis 2017; 8:e2817. [PMID: 28542147 PMCID: PMC5520698 DOI: 10.1038/cddis.2017.168] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/09/2017] [Accepted: 03/14/2017] [Indexed: 02/08/2023]
Abstract
In mammalian ovaries, primordial follicles remain in a quiescent state until activation by the surrounding microenvironment. Ovarian intervention, for example, ovarian cystectomy, ovarian wedge resection or laser drilling therapies for polycystic ovarian syndrome, has long been reported to change follicular development by an unknown mechanism(s). Herein, we established a murine model with partial ovarian resection of one ovary unilaterally, with the contralateral ovary undamaged. We found the injury accelerated follicular activation and development through the mTORC1 signaling pathway. Moreover, the stimulation of primordial follicles was restricted near the incision site where the mTORC1 pathway showed sequential activation beginning at the interstitial cells and proceeding to the primordial follicles. Total and polysome-associated RNA-seq revealed the increase of the nerve growth factor (NGF) family member, in both two fractions and immunostaining showed the restricted induction of NGF near the incision site. In cultured newborn ovaries, NGF demonstrated increase of follicular activation, and moreover, the NGF inhibitor K252a effectively blocked activation of primordial follicles stimulated by the surgery. We liken ovulation in mammals to minor tissue trauma, which happens naturally and cyclically in the body. As the increase in NGF accompanied the accumulation of activated primordial follicles after ovulation, our study may represent a common mechanism for selective follicular activation induced by a localized increase in NGF in interstitial cells and mediated via the mTOR signaling pathway. In addition, the NGF inhibitor K252a and the mTOR inhibitor rapamycin constitute good candidates for protecting follicular reserve against over exhaustion after ovarian surgery.
Collapse
|
44
|
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, National Institute for Research in Reproductive Health, Parel, Mumbai 400 012, India.
| |
Collapse
|