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] [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
|
A Simplified and Effective Approach for the Isolation of Small Pluripotent Stem Cells Derived from Human Peripheral Blood. Biomedicines 2023; 11:biomedicines11030787. [PMID: 36979766 PMCID: PMC10045871 DOI: 10.3390/biomedicines11030787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/24/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Pluripotent stem cells are key players in regenerative medicine. Embryonic pluripotent stem cells, despite their significant advantages, are associated with limitations such as their inadequate availability and the ethical dilemmas in their isolation and clinical use. The discovery of very small embryonic-like (VSEL) stem cells addressed the aforementioned limitations, but their isolation technique remains a challenge due to their small cell size and their efficiency in isolation. Here, we report a simplified and effective approach for the isolation of small pluripotent stem cells derived from human peripheral blood. Our approach results in a high yield of small blood stem cell (SBSC) population, which expresses pluripotent embryonic markers (e.g., Nanog, SSEA-3) and the Yamanaka factors. Further, a fraction of SBSCs also co-express hematopoietic markers (e.g., CD45 and CD90) and/or mesenchymal markers (e.g., CD29, CD105 and PTH1R), suggesting a mixed stem cell population. Finally, quantitative proteomic profiling reveals that SBSCs contain various stem cell markers (CD9, ITGA6, MAPK1, MTHFD1, STAT3, HSPB1, HSPA4), and Transcription reg complex factors (e.g., STAT5B, PDLIM1, ANXA2, ATF6, CAMK1). In conclusion, we present a novel, simplified and effective isolating process that yields an abundant population of small-sized cells with characteristics of pluripotency from human peripheral blood.
Collapse
|
3
|
Wang J, Fang J, Feng M, Li L, Ma L, Zhao X, Dai Y. Inhibition of EED activity enhances cell survival of female germline stem cell and improves the oocytes production during oogenesis in vitro. Open Biol 2023; 13:220211. [PMID: 36695089 PMCID: PMC9874982 DOI: 10.1098/rsob.220211] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Ovarian organoids, based on female germline stem cells (FGSCs), are nowadays widely applied for reproductive medicine screening and exploring the potential mechanisms during mammalian oogenesis. However, there are still key issues that urgently need to be resolved in ovarian organoid technology, one of which is to establish a culture system that effectively expands FGSCs in vitro, as well as maintaining the unipotentcy of FGSCs to differentiate into oocytes. Here, FGSCs were EED226 treated and processed for examination of proliferation and differentiation in vitro. According to the results, EED226 specifically increased FGSC survival by decreasing the enrichment of H3K27me3 on Oct4 promoter and exon, as well as enhancing OCT4 expression and inhibiting P53 and P63 expression. Notably, we also found that FGSCs with EED226 treatment differentiated into more oocytes during oogenesis in vitro, and the resultant oocytes maintained a low level of P63 versus control at early stage development. These results demonstrated that inhibition of EED activity appeared to promote the survival of FGSCs and markedly inhibited their apoptosis during in vitro differentiation. As a result of our study, we propose an effective culture strategy to culture FGSCs and obtain oocytes in vitro, which provides a new vision for oogenesis in vitro.
Collapse
Affiliation(s)
- Jiapeng Wang
- College of Life Sciences, Inner Mongolia University, Xilingol South Road No. 49, Hohhot 010020, People's Republic of China
| | - Junxian Fang
- College of Life Sciences, Inner Mongolia University, Xilingol South Road No. 49, Hohhot 010020, People's Republic of China
| | - Mingqian Feng
- College of Life Sciences, Inner Mongolia University, Xilingol South Road No. 49, Hohhot 010020, People's Republic of China
| | - Liping Li
- College of Life Sciences, Inner Mongolia University, Xilingol South Road No. 49, Hohhot 010020, People's Republic of China
| | - Lixin Ma
- College of Life Sciences, Inner Mongolia University, Xilingol South Road No. 49, Hohhot 010020, People's Republic of China
| | - Xiaorong Zhao
- College of Life Sciences, Inner Mongolia University, Xilingol South Road No. 49, Hohhot 010020, People's Republic of China
| | - Yanfeng Dai
- College of Life Sciences, Inner Mongolia University, Xilingol South Road No. 49, Hohhot 010020, People's Republic of China
| |
Collapse
|
4
|
Proteomic Analysis of Murine Bone Marrow Very Small Embryonic-like Stem Cells at Steady-State Conditions and after In Vivo Stimulation by Nicotinamide and Follicle-Stimulating Factor Reflects their Germ-Lineage Origin and Multi Germ Layer Differentiation Potential. Stem Cell Rev Rep 2023; 19:120-132. [PMID: 35986128 PMCID: PMC9823037 DOI: 10.1007/s12015-022-10445-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/09/2022] [Indexed: 01/29/2023]
Abstract
Very small embryonic-like stem cells (VSELs) are a dormant population of development early stem cells deposited in adult tissues that as demonstrated contribute to tissue/organ repair and regeneration. We postulated developmental relationship of these cells to migrating primordial germ cells (PGCs) and explained the quiescent state of these cells by the erasure of differently methylated regions (DMRs) at some of the paternally imprinted genes involved in embryogenesis. Recently, we reported that VSELs began to proliferate and expand in vivo in murine bone marrow (BM) after exposure to nicotinamide (NAM) and selected pituitary and gonadal sex hormones. In the current report, we performed proteomic analysis of VSELs purified from murine bone marrow (BM) after repeated injections of NAM + Follicle-Stimulating Hormone (FSH) that in our previous studies turned out to be an effective combination to expand these cells. By employing the Gene Ontology (GO) resources, we have performed a combination of standard GO annotations (GO-CAM) to produce a network between BM steady-state conditions VSELs (SSC-VSELS) and FSH + NAM expanded VSELs (FSH + NAM VSELs). We have identified several GO biological processes regulating development, organogenesis, gene expression, signal transduction, Wnt signaling, insulin signaling, cytoskeleton organization, cell adhesion, inhibiting apoptosis, responses to extra- and intracellular stimuli, protein transport and stabilization, protein phosphorylation and ubiquitination, DNA repair, immune response, and regulation of circadian rhythm. We report that VSELs express a unique panel of proteins that only partially overlapped with the proteome of BM - derived hematopoietic stem cells (HSCs) and hematopoietic mononuclear cells (MNCs) and respond to FSH + NAM stimulation by expressing proteins involved in the development of all three germ layers. Thus, our current data supports further germ-lineage origin and multi germ layer differentiation potential of these cells.
Collapse
|
5
|
Kelsey T. Models and Biomarkers for Ovarian Ageing. Subcell Biochem 2023; 103:185-199. [PMID: 37120469 DOI: 10.1007/978-3-031-26576-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
The human ovarian reserve is defined by the number of non-growing follicles (NGFs) in the ovary, with the age-related decline in NGF population determining age at menopause for healthy women. In this chapter, the concept of ovarian reserve is explored in detail, with a sequence of models described that in principle allow any individual to be compared to the general population. As there is no current technology that can count the NGFs in a living ovary, we move our focus to biomarkers for the ovarian reserve. Using serum analysis and ultrasound it is possible to measure anti-Müllerian hormone (AMH), follicle-stimulating hormone (FSH), and ovarian volume (OV) and to count numbers of antral follicles (AFC). These are compared, with ovarian volume being the closest to a true biomarker for a wide range of ages and with AMH and AFC being the most popular for post-pubertal and pre-menopausal ages. The study of genetic and subcellular biomarkers for the ovarian reserve has produced less concrete results. Recent advances are described and compared in terms of limitations and potential. The chapter concludes with an overview of the future study indicated by our current knowledge and by current controversy in the field.
Collapse
Affiliation(s)
- Tom Kelsey
- School of Computer Science, University of St Andrews, St Andrews, UK.
| |
Collapse
|
6
|
Dadashzadeh A, Moghassemi S, Grubliauskaité M, Vlieghe H, Brusa D, Amorim CA. Medium supplementation can influence the human ovarian cells in vitro. J Ovarian Res 2022; 15:137. [PMID: 36572931 PMCID: PMC9791781 DOI: 10.1186/s13048-022-01081-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 12/16/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Cells are an essential part of the triple principles of tissue engineering and a crucial component of the engineered ovary as they can induce angiogenesis, synthesize extracellular matrix and influence follicle development. Here, we hypothesize that by changing the medium supplementation, we can obtain different cell populations isolated from the human ovary to use in the engineered ovary. To this end, we have in vitro cultured cells isolated from the menopausal ovarian cortex using different additives: KnockOut serum replacement (KO), fetal bovine serum (FBS), human serum albumin (HSA), and platelet lysate (PL). RESULTS Our results showed that most cells soon after isolation (pre-culture, control) and cells in KO and FBS groups were CD31- CD34- (D0: vs. CD31-CD34+, CD31 + CD34+, and CD31 + CD34- p < 0.0001; KO: vs. CD31-CD34+, CD31 + CD34+, and CD31 + CD34- p < 0.0001; FBS: vs. CD31-CD34+ and CD31 + CD34+ p < 0.001, and vs. CD31 + CD34- p < 0.01). Moreover, a deeper analysis of the CD31-CD34- population demonstrated a significant augmentation (more than 86%) of the CD73+ and CD90+ cells (possibly fibroblasts, mesenchymal stem cells, or pericytes) in KO- and FBS-based media compared to the control (around 16%; p < 0.001). Still, in the CD31-CD34- population, we found a higher proportion (60%) of CD90+ and PDPN+ cells (fibroblast-like cells) compared to the control (around 7%; vs PL and KO p < 0.01 and vs FBS p < 0.001). Additionally, around 70% of cells in KO- and FBS-based media were positive for CD105 and CD146, which may indicate an increase in the number of pericytes in these media compared to a low percentage (4%) in the control group (vs KO and FBS p < 0.001). On the other hand, we remarked a significant decrease of CD31- CD34+ cells after in vitro culture using all different medium additives (HSA vs D0 p < 0.001, PL, KO, and FBS vs D0 P < 0.01). We also observed a significant increase in epithelial cells (CD326+) when the medium was supplemented with KO (vs D0 p < 0.05). Interestingly, HSA and PL showed more lymphatic endothelial cells compared to other groups (CD31 + CD34+: HSA and PL vs KO and FBS p < 0.05; CD31 + CD34 + CD90 + PDPN+: HSA and PL vs D0 p < 0.01). CONCLUSION Our results demonstrate that medium additives can influence the cell populations, which serve as building blocks for the engineered tissue. Therefore, according to the final application, different media can be used in vitro to favor different cell types, which will be incorporated into a functional matrix.
Collapse
Affiliation(s)
- Arezoo Dadashzadeh
- grid.7942.80000 0001 2294 713XPôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Hippocrate 55, bte B1.55.03, 1200 Brussels, Belgium
| | - Saeid Moghassemi
- grid.7942.80000 0001 2294 713XPôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Hippocrate 55, bte B1.55.03, 1200 Brussels, Belgium
| | - Monika Grubliauskaité
- grid.459837.40000 0000 9826 8822Department of Biobank, National Cancer Institute, 08660 Vilnius, Lithuania
| | - Hanne Vlieghe
- grid.7942.80000 0001 2294 713XPôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Hippocrate 55, bte B1.55.03, 1200 Brussels, Belgium
| | - Davide Brusa
- grid.7942.80000 0001 2294 713XCytoFlux-Flow Cytometry and Cell Sorting Platform, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Christiani A. Amorim
- grid.7942.80000 0001 2294 713XPôle de Recherche en Physiopathologie de la Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Avenue Hippocrate 55, bte B1.55.03, 1200 Brussels, Belgium
| |
Collapse
|
7
|
Szczerba A, Śliwa A, Pieta PP, Jankowska A. The Role of Circulating Tumor Cells in Ovarian Cancer Dissemination. Cancers (Basel) 2022; 14:cancers14246030. [PMID: 36551515 PMCID: PMC9775737 DOI: 10.3390/cancers14246030] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Metastatic ovarian cancer is the main reason for treatment failures and consequent deaths. Ovarian cancer is predisposed to intraperitoneal dissemination. In comparison to the transcoelomic route, distant metastasis via lymph vessels and blood is less common. The mechanisms related to these two modes of cancer spread are poorly understood. Nevertheless, the presence of tumor cells circulating in the blood of OC patients is a well-established phenomenon confirming the significant role of lymphatic and hematogenous metastasis. Thus, the detection of CTCs may provide a minimally invasive tool for the identification of ovarian cancer, monitoring disease progression, and treatment effectiveness. This review focuses on the biology of ovarian CTCs and the role they may play in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Anna Szczerba
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznan, Poland
| | - Aleksandra Śliwa
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznan, Poland
| | - Pawel P. Pieta
- Department of Bionic and Experimental Medical Biology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Anna Jankowska
- Chair and Department of Cell Biology, Poznan University of Medical Sciences, Rokietnicka 5D, 60-806 Poznan, Poland
- Correspondence: ; Tel.: +48-618-547-190
| |
Collapse
|
8
|
Davies DM, van den Handel K, Bharadwaj S, Lengefeld J. Cellular enlargement - A new hallmark of aging? Front Cell Dev Biol 2022; 10:1036602. [PMID: 36438561 PMCID: PMC9688412 DOI: 10.3389/fcell.2022.1036602] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/17/2022] [Indexed: 12/03/2023] Open
Abstract
Years of important research has revealed that cells heavily invest in regulating their size. Nevertheless, it has remained unclear why accurate size control is so important. Our recent study using hematopoietic stem cells (HSCs) in vivo indicates that cellular enlargement is causally associated with aging. Here, we present an overview of these findings and their implications. Furthermore, we performed a broad literature analysis to evaluate the potential of cellular enlargement as a new aging hallmark and to examine its connection to previously described aging hallmarks. Finally, we highlight interesting work presenting a correlation between cell size and age-related diseases. Taken together, we found mounting evidence linking cellular enlargement to aging and age-related diseases. Therefore, we encourage researchers from seemingly unrelated areas to take a fresh look at their data from the perspective of cell size.
Collapse
Affiliation(s)
- Daniel M. Davies
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Kim van den Handel
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Soham Bharadwaj
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Jette Lengefeld
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
- Center for Hematology and Regenerative Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Chhabria S, Takle V, Sharma N, Kharkar P, Pansare K, Tripathi A, Tripathi A, Bhartiya D. Extremely Active Nano-formulation of Resveratrol (XAR™) attenuates and reverses chemotherapy-induced damage in mice ovaries and testes. J Ovarian Res 2022; 15:115. [PMID: 36271409 PMCID: PMC9585716 DOI: 10.1186/s13048-022-01043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/23/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Fertility preservation and restoration in cancer patients/survivors is the need of present times when increased numbers of patients get cured of cancer but face infertility as a serious side effect. Resveratrol has beneficial effects on chemoablated ovaries and testes in mice but has failed to enter the clinics because of extremely poor bioavailability. The present study was undertaken to evaluate the protective and curative effects of Extremely active Resveratrol (XAR™)- a nano-formulation of resveratrol with significantly improved bioavailability- on mouse ovary and testis after chemotherapy. Effects of XAR™ and FSH were compared on stimulation of follicle growth in adult mice ovaries. XAR™ (25 mg/kg) was administered for two days prior to chemotherapy to study the protective effects on the mouse gonads. XAR™ was also administered for 14 days post chemoablation to study the regenerative effects. Besides effect on numbers of primordial and growing follicles and spermatogenesis, the effect of XAR™ was also evaluated on the transcripts specific for ovarian/testicular stem/progenitor/germ cells, their proliferation, differentiation, meiosis, and the antioxidant indices. RESULTS Similar to FSH, XAR™ increased the numbers of primordial follicles (PF) as well as growing follicles. It protected the gonads from the adverse effects of chemotherapy and showed the ability to regenerate non-functional, chemoablated gonads. Besides stimulating follicle growth in adult ovaries similar to FSH, XAR™ also protected the testes from the adverse effects of chemotherapy and improved spermatogenesis. This was accompanied by improved anti-oxidant indices. CONCLUSIONS The results of the present study potentiate the use of XAR™ in pilot clinical studies to protect gonadal function during oncotherapy and also regenerate non-functional gonads in cancer survivors by improving antioxidant indices and stem cell-based tissue regeneration.
Collapse
Affiliation(s)
- Sagar Chhabria
- Epigeneres Biotech Pvt. Ltd., Sun Mill Compound, Ikon House, B-Block, Senapati Bapat Marg, Lower Parel, Mumbai, Maharashtra, 400013, India
| | - Vaishnavi Takle
- Epigeneres Biotech Pvt. Ltd., Sun Mill Compound, Ikon House, B-Block, Senapati Bapat Marg, Lower Parel, Mumbai, Maharashtra, 400013, India
| | - Nripen Sharma
- Epigeneres Biotech Pvt. Ltd., Sun Mill Compound, Ikon House, B-Block, Senapati Bapat Marg, Lower Parel, Mumbai, Maharashtra, 400013, India
| | - Prashant Kharkar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Matunga, Mumbai, 400 019, India
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, Vile Parle (West), Mumbai, 400 056, India
| | - Kshama Pansare
- Epigeneres Biotech Pvt. Ltd., Sun Mill Compound, Ikon House, B-Block, Senapati Bapat Marg, Lower Parel, Mumbai, Maharashtra, 400013, India
| | - Anish Tripathi
- Epigeneres Biotech Pvt. Ltd., Sun Mill Compound, Ikon House, B-Block, Senapati Bapat Marg, Lower Parel, Mumbai, Maharashtra, 400013, India
| | - Ashish Tripathi
- Epigeneres Biotech Pvt. Ltd., Sun Mill Compound, Ikon House, B-Block, Senapati Bapat Marg, Lower Parel, Mumbai, Maharashtra, 400013, India
| | - Deepa Bhartiya
- Epigeneres Biotech Pvt. Ltd., Sun Mill Compound, Ikon House, B-Block, Senapati Bapat Marg, Lower Parel, Mumbai, Maharashtra, 400013, India.
| |
Collapse
|
10
|
ÜNAL MS, SEÇME M. Does the ovarian surface epithelium differentiate into primordial follicle and primary follicle precursor structures? CUKUROVA MEDICAL JOURNAL 2022. [DOI: 10.17826/cumj.1134852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Purpose: The aim of this study is to investigate the differentiation capacity of ovarian surface epithelial cells both in cell culture conditions and in ovarian tissue sections.
Materials and Methods: The ovaries of two prepubertal (4 weeks old) female rats were divided into small pieces and explant cell culture was created. Ovarian surface epithelium proliferating together with ovarian stromal cells in mixed cell culture was isolated and reproduced. In addition, ovarian surface epithelium was examined in histological sections of ovarian tissue and images were taken under the microscope.
Results: The morphological appearance of the ovarian surface epithelium was found to be cobblestone. In the count performed under phase contrast microscopy, it was observed that 2x106 and 3x106 cells were grown in the culture dishes, respectively. Primordial follicle-like structures were observed in some areas of the petri dishes. On the histological sections, primordial and primary follicle precursor structures were observed on the basement membrane.
Conclusion: Showing oocyte markers (Gdf-9, C-Mos, Zpc, Stella) and germ cell markers (Dazl, Vasa, Blimp1, Fragilis) both in cell cultures and in histological sections can give us valuable information in terms of monitoring the differentiation capacity of these cells.
Collapse
|
11
|
Isolation of Female Germline Stem Cells from Mouse and Human Ovaries by Differential Adhesion. Int J Cell Biol 2022; 2022:5224659. [PMID: 36120418 PMCID: PMC9473869 DOI: 10.1155/2022/5224659] [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: 05/14/2022] [Accepted: 08/05/2022] [Indexed: 11/18/2022] Open
Abstract
Spermatogonial stem cell (SSC) counterparts known as female germline stem cells (fGSCs) were found in the mammalian ovary in 2004. Although the existence of fGSCs in the mammalian postnatal ovary is still under controversy, fGSC discovery encourages investigators to better understand the various aspects of these cells. However, their existence is not accepted by all scientists in the field because isolation of fGSCs by fluorescent activated cell sorting (FACS) has not been reproducible. In this study, we used differential adhesion to isolate and enrich fGSCs from mouse and human ovaries and subsequently cultured them in vitro. fGSCs were able to proliferate in vitro and expressed germ cell-specific markers Vasa, Dazl, Blimp1, Fragilis, Stella, and Oct4, at the protein level. Moreover, mouse and human fGSCs were, respectively, cultured for more than four months and one month in culture. Both mouse and human fGSCs maintained the expression of germ cell-specific markers over these times. In vitro cultured fGSCs spontaneously produced oocyte-like cells (OLCs) which expressed oocyte-relevant markers. Our results demonstrated that differential adhesion allows reproducible isolation of fGSCs that are able to proliferate in vitro over time. This source of fGSCs can serve as a suitable material for studying mechanisms underlying female germ cell development and function.
Collapse
|
12
|
Sharma D, Bhartiya D. Dysfunctional Ovarian Stem Cells Due to Neonatal Endocrine Disruption Result in PCOS and Ovarian Insufficiency in Adult Mice. Stem Cell Rev Rep 2022; 18:2912-2927. [PMID: 35834052 DOI: 10.1007/s12015-022-10414-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 12/13/2022]
Abstract
Polycystic ovarian syndrome (PCOS) is a common global cause of anovulatory infertility but underlying etiology leading to PCOS still remains elusive. Fetal and perinatal endocrine disruption reportedly affects germ cell nests (GCN) breakdown, meiosis, and primordial follicle (PF) assembly with unassembled oocytes in neonatal ovaries. We recently reported that very small embryonic-like stem cells (VSELs) and ovarian stem cells (OSCs) express ERα, ERβ and FSHR, undergo distinct cyclic changes and neo-oogenesis encompassing GCN formation, meiosis, and primordial follicle (PF) assembly on regular basis in adult mice ovaries and these GCN are arrested in pre-meiotic or early meiotic stage in aged ovaries. Present study was undertaken to evaluate whether neonatal exposure to endocrine disruption (estradiol E2 or diethylstilbestrol DES) affects ovarian stem cells and their differentiation (neo-oogenesis) and PF assembly in adult 100 days old ovaries. Neonatal exposure to E2 resulted in typical features of PCOS including hyperandrogenism, infertility, increased stromal compartment, absent corpus lutea, and cystic follicles whereas DES treated ovaries showed rapid recruitment of follicles in young ovaries and multi-ovular/cystic follicles. Ovary surface epithelial cells smears showed large numbers of growth-arrested GCN in zygotene/pachytene with increased expression of Mlh-1 and Scp-1 suggesting defects at synapsis and recombination stages during prophase-1 of meiosis. Being immortal and expression of ERα and ERβ makes VSELs directly vulnerable to carry developmental endocrine insults to adult life. Dysfunction of VSELs/OSCs possibly results in oocyte defects observed in our study in PCOS/POI besides the widely reported defects in granulosa cells.
Collapse
Affiliation(s)
- Diksha Sharma
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, 400 012, Mumbai, India
| | - Deepa Bhartiya
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive and Child Health, Jehangir Merwanji Street, Parel, 400 012, Mumbai, India.
| |
Collapse
|
13
|
Oktem O. In response to: why double ovarian stimulation in an in vitro fertilization cycle is potentially unsafe? Hum Reprod 2022; 37:1945-1947. [DOI: 10.1093/humrep/deac127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Ozgur Oktem
- Department of Obstetrics and Gynecology, Division Reproductive Endocrinology and Infertility, Assisted Reproduction Unit, Koç University School of Medicine , Istanbul, Turkey
- The Graduate School of Health Sciences, Koç University , Istanbul, Turkey
- Research Center for Translational Medicine, Koç University , Istanbul, Turkey
| |
Collapse
|
14
|
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
|
15
|
Eijkenboom L, Saedt E, Zietse C, Braat D, Beerendonk C, Peek R. Strategies to safely use cryopreserved ovarian tissue to restore fertility after cancer: A systematic review. Reprod Biomed Online 2022; 45:763-778. [DOI: 10.1016/j.rbmo.2022.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
|
16
|
Tocci A. Reply to: Neither rationale nor scientific evidence exist to support that double stimulation is potentially unsafe. Hum Reprod 2022; 37:1951-1952. [PMID: 35394502 DOI: 10.1093/humrep/deac065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
17
|
Artificial Oocyte: Development and Potential Application. Cells 2022; 11:cells11071135. [PMID: 35406698 PMCID: PMC8998074 DOI: 10.3390/cells11071135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/25/2022] [Accepted: 03/26/2022] [Indexed: 02/07/2023] Open
Abstract
Millions of people around the world suffer from infertility, with the number of infertile couples and individuals increasing every year. Assisted reproductive technologies (ART) have been widely developed in recent years; however, some patients are unable to benefit from these technologies due to their lack of functional germ cells. Therefore, the development of alternative methods seems necessary. One of these methods is to create artificial oocytes. Oocytes can be generated in vitro from the ovary, fetal gonad, germline stem cells (GSCs), ovarian stem cells, or pluripotent stem cells (PSCs). This approach has raised new hopes in both basic research and medical applications. In this article, we looked at the principle of oocyte development, the landmark studies that enhanced our understanding of the cellular and molecular mechanisms that govern oogenesis in vivo, as well as the mechanisms underlying in vitro generation of functional oocytes from different sources of mouse and human stem cells. In addition, we introduced next-generation ART using somatic cells with artificial oocytes. Finally, we provided an overview of the reproductive application of in vitro oogenesis and its use in human fertility.
Collapse
|
18
|
Wu M, Lu Z, Zhu Q, Ma L, Xue L, Li Y, Zhou S, Yan W, Ye W, Zhang J, Luo A, Wang S. DDX4 + stem cells in the ovaries of postmenopausal women: existence and differentiation potential. Stem Cells 2022; 40:88-101. [PMID: 35511860 DOI: 10.1093/stmcls/sxab002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 09/02/2021] [Indexed: 11/12/2022]
Abstract
Abstract
Ovarian aging is a pacemaker with multiple organ dysfunction. Recently, stem cells with the ability to generate new oocytes have been identified, which provides the possibility of stem cell therapy for ovarian aging. Several studies have revealed the existence of stem cells in human postmenopausal ovary. In this study, we describe a new method using magnetic activated cell sorting combined with differential adhesion to isolate DDX4 + stem cells from ovaries of postmenopausal women and show that the cells exhibit similar gene expression profiles and growth characteristics with primitive germ cells. Furthermore, the DDX4 + stem cells could enter meiosis stage and differentiation into oocytes. The RNA-seq data of the differentiated oocytes shows that mitochondrial metabolism may play an important role in the oogenesis process of the DDX4 + stem cells. Through using human ovarian cortical fragments transplantation model, we indicated that the GFP-DDX4 + stem cells differentiated into some GFP positive oocyte-like structure in vivo. Our study provided a new method for the isolation of DDX4 + stem cells from the ovaries of postmenopausal women and confirmed the ability of these stem cells to differentiate into oocytes.
Collapse
Affiliation(s)
- Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiyong Lu
- Hubei Key Laboratory of Embryonic Stem Cell Research, TaiHe Hospital, Hubei University of Medicine, Shiyan, China
| | - Qingqing Zhu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lingwei Ma
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liru Xue
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Su Zhou
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Yan
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenlei Ye
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Aiyue Luo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
19
|
Bhartiya D, Patel H, Kaushik A, Singh P, Sharma D. Endogenous, tissue-resident stem/progenitor cells in gonads and bone marrow express FSHR and respond to FSH via FSHR-3. J Ovarian Res 2021; 14:145. [PMID: 34717703 PMCID: PMC8556987 DOI: 10.1186/s13048-021-00883-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Follicle stimulating hormone (FSH) is secreted by the anterior pituitary and acts on the germ cells indirectly through Granulosa cells in ovaries and Sertoli cells in the testes. Extragonadal action of FSH has been reported but is still debated. Adult tissues harbor two populations of stem cells including a reserve population of primitive, small-sized, pluripotent very small embryonic-like stem cells (VSELs) and slightly bigger, tissue-specific progenitors which include ovarian stem cells (OSCs) in ovaries, spermatogonial stem cells (SSCs) in testes, endometrial stem cells (EnSCs) in uterus and hematopoietic stem cells (HSCs) in the bone marrow. Data has accumulated in animal models showing FSHR expression on both VSELs and progenitors in ovaries, testes, uterus and bone marrow and eventually gets lost as the cells differentiate further. FSH exerts a direct action on the stem/progenitor cells via alternatively spliced FSHR-3 rather than the canonical FSHR-1. FSH stimulates VSELs to undergo asymmetrical cell divisions to self-renew and give rise to the progenitors that in turn undergo symmetrical cell divisions and clonal expansions followed by differentiation into specific cell types. Excessive self-renewal of VSELs results in cancer and this explains ubiquitous expression of embryonic markers including nuclear OCT-4 along with FSHR in cancerous tissues. Focus of this review is to compile published data to support this concept. FSHR expression in stem/progenitor cells was confirmed by immuno-fluorescence, Western blotting, in situ hybridization and by quantitative RT-PCR. Two different commercially available antibodies (Abcam, Santacruz) were used to confirm specificity of FSHR expression along with omission of primary antibody and pre-incubation of antibody with immunizing peptide as negative controls. Western blotting allowed detection of alternatively spliced FSHR isoforms. Oligoprobes and primers specific for Fshr-1 and Fshr-3 were used to study these alternately-sliced isoforms by in situ hybridization and their differential expression upon FSH treatment by qRT-PCR. To conclude, stem/progenitor cells in adult tissues express FSHR and directly respond to FSH via FSHR-3. These findings change the field of FSH-FSHR biology, call for paradigm shift, explain FSHR expression on cancer cells in multiple organs and provide straightforward explanations for various existing conundrums including extragonadal expression of FSHR.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India.
| | - Hiren Patel
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
- Present address: Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ankita Kaushik
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| | - Pushpa Singh
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| | - Diksha Sharma
- Stem Cell Biology Department, ICMR- National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| |
Collapse
|
20
|
Ebrahim N, Al Saihati HA, Shaman A, Dessouky AA, Farid AS, Hussien NI, Mostafa O, Seleem Y, Sabry D, Saad AS, Emam HT, Hassouna A, Badr OAM, Saffaf BA, Forsyth NR, Salim RF. Bone marrow-derived mesenchymal stem cells combined with gonadotropin therapy restore postnatal oogenesis of chemo-ablated ovaries in rats via enhancing very small embryonic-like stem cells. Stem Cell Res Ther 2021; 12:517. [PMID: 34579781 PMCID: PMC8477571 DOI: 10.1186/s13287-021-02415-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/25/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Very small embryonic-like stem cells (VSELs) are a rare population within the ovarian epithelial surface. They contribute to postnatal oogenesis as they have the ability to generate immature oocytes and resist the chemotherapy. These cells express markers of pluripotent embryonic and primordial germ cells. OBJECTIVE We aimed to explore the capability of VSELs in restoring the postnatal oogenesis of chemo-ablated rat ovaries treated with bone marrow-derived mesenchymal stem cells (BM-MSCs) combined with pregnant mare serum gonadotropin (PMSG). METHODS Female albino rats were randomly assigned across five groups: I (control), II (chemo-ablation), III (chemo-ablation + PMSG), IV (chemo-ablation + MSCs), and V (chemo-ablation + PMSG + MSCs). Postnatal oogenesis was assessed through measurement of OCT4, OCT4A, Scp3, Mvh, Nobox, Dazl4, Nanog, Sca-1, FSHr, STRA8, Bax, miR143, and miR376a transcript levels using qRT-PCR. Expression of selected key proteins were established as further confirmation of transcript expression changes. Histopathological examination and ovarian hormonal assessment were determined. RESULTS Group V displayed significant upregulation of all measured genes when compared with group II, III or IV. Protein expression confirmed the changes in transcript levels as group V displayed the highest average density in all targeted proteins. These results were confirmed histologically by the presence of cuboidal germinal epithelium, numerous primordial, unilaminar, and mature Graafian follicles in group V. CONCLUSION VSELs can restore the postnatal oogenesis in chemo-ablated ovaries treated by BM-MSCs combined with PMSG.
Collapse
Affiliation(s)
- Nesrine Ebrahim
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha, Egypt
- Stem Cell Unit, Faculty of Medicine, Benha University, Benha, Egypt
| | - Hajir A Al Saihati
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, University of Hafr Albatin, Al-Batin, Saudi Arabia
| | - Amani Shaman
- Obstetrics and Gynecology Medical College, Tabuk University, Tabuk, Saudi Arabia
| | - Arigue A Dessouky
- Department of Medical Histology and Cell Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ayman Samir Farid
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Noha I Hussien
- Department of Medical Physiology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Ola Mostafa
- Department of Histology and Cell Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Yasmin Seleem
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Badr University, Cairo, Egypt
| | - Ahmed S Saad
- Department of Obstetrics & Gynecology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Hanan Tawfeek Emam
- Department of Clinical Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Amira Hassouna
- School of Interprofessional Health Studies, Faculty of Health and Environmental Sciences, AUT University, Auckland, New Zealand
| | - Omnia A M Badr
- Department of Genetics and Genetic Engineering, Faculty of Agriculture, Benha University, Benha, Egypt
| | - Bayan A Saffaf
- Department of pharmacology, Faculty of Pharmacy, Future University, Cairo, Egypt
| | - Nicholas R Forsyth
- Guy Hilton Research Laboratories, School of Pharmacy and Bioengineering, Faculty of Medicine and Health Sciences, Keele University, Keele, UK
| | - Rabab F Salim
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Qalyubia, 13512, Egypt.
| |
Collapse
|
21
|
Tocci A. The safety of VASA pos presumptive adult ovarian stem cells. Reprod Biomed Online 2021; 43:587-597. [PMID: 34474974 DOI: 10.1016/j.rbmo.2021.06.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 01/16/2023]
Abstract
Isolation and characterization of presumptive human adult ovarian stem cells (OSC) has broken the long standing dogma of the absence of postnatal neo-oogenesis. Human adult OSC have been immunosorted by antibodies reacting against the RNA helicase VASA and have been reported to engraft into appropriate stem cell niches to promote neo-oogenesis. Analysis of published research, however, questions some of the findings on isolation, characterization, in-vitro self-renewal and clinical safety of the presumptive human adult OSC. In the present study, human VASApos embryo-fetal primordial germ cells and presumptive adult OSC are shown to share several pluripotency and early germ cell markers not ascertained in the initial characterization of adult OSC. A new hypothesis is made that the restoration of fertility claimed to result from presumptive human adult OSC may be attributed instead to VASApos embryo-fetal primordial germ cell remnants in the adult ovary, or alternatively to earlier VASAneg germ cells generated by in-vitro de-differentiation of the presumptive OSC. The suggested hypotheses have extensive implications for the practice and safety of adult OSC in the development of new treatments aimed at rescuing the ovarian reserve.
Collapse
Affiliation(s)
- Angelo Tocci
- Gruppo Donnamed, Reproductive Medicine Unit Via Cassia 1110 00189, Rome, Italy.
| |
Collapse
|
22
|
Stem Cells in Adult Mice Ovaries Form Germ Cell Nests, Undergo Meiosis, Neo-oogenesis and Follicle Assembly on Regular Basis During Estrus Cycle. Stem Cell Rev Rep 2021; 17:1695-1711. [PMID: 34455541 DOI: 10.1007/s12015-021-10237-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2021] [Indexed: 12/21/2022]
Abstract
Very small embryonic-like (VSELs) and ovarian (OSCs) stem cells are located in adult mammalian ovary surface epithelium (OSE). OSCs can expand long-term and differentiate into oocyte-like structures in vitro and have resulted in birth of fertile pups. Lineage tracing studies have provided evidence to suggest OSCs differentiation into oocytes in vivo. But how these stem cells function under normal physiological conditions has not yet been well worked out. Besides studying STRA-8 and SCP-3 expression in enzymatically isolated OSE cells smears, mice were injected BrdU to track mitosis, meiosis and follicle assembly. H&E stained OSE cells during late diestrus and proestrus showed VSELs undergoing asymmetrical cell divisions to give rise to slightly bigger OSCs which in turn underwent symmetrical cell divisions followed by clonal expansion (rapid expansion with incomplete cytokinesis) during early estrus to form germ cell nests (GCN). OCT-4, SSEA-1, MVH and DAZL positive cells in GCN expressed Erα, Erβ and FSHR, were interconnected by ring canals (TEX-14), showed mitochondrial aggregation (Cytochrome C) and Balbiani Body (TRAL). Apoptosis in 'nurse' cells was marked by PARP and putative oocytes were clearly visualized. BrdU was detected in cells undergoing mitosis/meiosis and also in an oocyte of secondary follicle. FACS sorted, green fluorescent protein (GFP) positive VSELs upon transplantation resulted in GFP positive GCN suggesting crucial role for VSELs in adult ovaries. Results suggest that various events described during oogenesis and follicle assembly in fetal ovaries are recapitulated on regular basis in adult ovary and result in the formation of follicles.
Collapse
|
23
|
Kakar SS, Ratajczak MZ. Paper of October Issue of Stem Cell Reviews and Reports Presents a Novel View on Oogenesis in Adult Mammalian Ovaries. Stem Cell Rev Rep 2021; 17:1519-1520. [PMID: 34448117 DOI: 10.1007/s12015-021-10248-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Sham S Kakar
- Department of Physiology, University of Louisville, Louisville, KY, 40202, USA
| | - Mariusz Z Ratajczak
- Department of Medicine, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
24
|
Sequeira RC, Sittadjody S, Criswell T, Atala A, Jackson JD, Yoo JJ. Enhanced method to select human oogonial stem cells for fertility research. Cell Tissue Res 2021; 386:145-156. [PMID: 34415395 DOI: 10.1007/s00441-021-03464-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/13/2021] [Indexed: 11/25/2022]
Abstract
Alternative methods to obtain mature oocytes are still needed for women with premature ovarian failure (POF). Oogonial stem cells (OSCs), found in adult ovaries, have provided insight into potential paths to treating infertility. Previously, the DDX4 antibody marker alone was utilized to isolate OSCs; however, extensive debate over its location in OSCs versus resulting oocytes (transmembrane or intracytoplasmic) has raised doubt about the identity of these cells. Separate groups, however, have efficiently isolated OSCs using another antibody marker Ifitm3 which is consistently recognized to be transmembrane in location. We hypothesized that by using anti-DDX4 and anti-IFITM3 antibodies, in combination, with MACS, we would improve the yield of isolated OSCs versus using anti-DDX4 antibodies alone. Our study supports earlier findings of OSCs in ovaries during the entire female lifespan: from reproductive age through post-menopausal age. MACS sorting ovarian cells using a the two-marker combination yielded a ~ twofold higher percentage of OSCs from a given mass of ovarian tissue compared to existing single marker methods while minimizing the debate surrounding germline marker selection. During in vitro culture, isolated cells retained the germline phenotype expression of DDX4 and IFITM3 as confirmed by gene expression analysis, demonstrated characteristic germline stem cell self-assembly into embryoid bodies, and formed > 40 µm "oocyte-like" structures that expressed the early oocyte markers GDF9, DAZL, and ZP1. This enhanced and novel method is clinically significant as it could be utilized in the future to more efficiently produce mature, secondary oocytes, for use with IVF/ICSI to treat POF patients.
Collapse
Affiliation(s)
- Russel C Sequeira
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA.
| | - Sivanandane Sittadjody
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Tracy Criswell
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - John D Jackson
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| | - James J Yoo
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27101, USA
| |
Collapse
|
25
|
Bhartiya D, Singh P, Sharma D, Kaushik A. Very small embryonic-like stem cells (VSELs) regenerate whereas mesenchymal stromal cells (MSCs) rejuvenate diseased reproductive tissues. Stem Cell Rev Rep 2021; 18:1718-1727. [PMID: 34410593 DOI: 10.1007/s12015-021-10243-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 12/15/2022]
Abstract
Compared to embryonic and induced pluripotent stem cells, mesenchymal stem/stromal cells (MSCs) have made their presence felt with good therapeutic promise and safety profile. Transplanting MSCs has successfully helped to reverse infertility and resulted in live births in animal models and also in humans. But the underlying mechanism for their therapeutic potential is not yet clear. MSCs are not pluripotent and hence lack plasticity to differentiate into multiple adult cell types. They rather act as 'paracrine providers' to the tissue-resident stem cells since similar beneficial effects are also observed when their secretome (microvesicles or exosomes) is transplanted. Cytokines, growth factors, signaling lipids, mRNAs, and miRNAs secreted by MSCs enables tissue-resident stem cells to undergo differentiation into specific cell types. Tissue-resident stem cells include pluripotent, very small embryonic-like stem cells (VSELs) and progenitors [spermatogonial (SSCs), ovarian (OSCs) and endometrial (EnSCs) stem cells in testes, ovary and uterus respectively] which function in a subtle manner to maintain life-long tissue homeostasis and regenerate damaged (non-functional) reproductive tissues by differentiating into sperm, oocytes and endometrial epithelial cells respectively. Similar to restoring spermatogenesis, primordial follicles numbers are increased upon transplanting MSCs. Published literature suggests that MSCs do not differentiate into epithelial cells in the endometrium. Nuclear OCT-4 positive VSELs and cytoplasmic OCT-4, AXIN2 and KERATIN-19 positive epithelial progenitors have a greater role during endometrial regeneration. We propose, transplantation of MSCs simply provides growth factors/cytokines essential for the tissue-resident stem/progenitor cells to undergo differentiation into sperm, eggs and endometrial epithelial cells in the reproductive tissues.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India.
| | - Pushpa Singh
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | - Diksha Sharma
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| | - Ankita Kaushik
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400012, India
| |
Collapse
|
26
|
Llarena N, Hine C. Reproductive Longevity and Aging: Geroscience Approaches to Maintain Long-Term Ovarian Fitness. J Gerontol A Biol Sci Med Sci 2021; 76:1551-1560. [PMID: 32808646 PMCID: PMC8361335 DOI: 10.1093/gerona/glaa204] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Indexed: 11/12/2022] Open
Abstract
Increases in delayed childbearing worldwide have elicited the need for a better understanding of the biological underpinnings and implications of age-related infertility. In women 35 years and older the incidences of infertility, aneuploidy, and birth defects dramatically increase. These outcomes are a result of age-related declines in both ovarian reserve and oocyte quality. In addition to waning reproductive function, the decline in estrogen secretion at menopause contributes to multisystem aging and the initiation of frailty. Both reproductive and hormonal ovarian function are limited by the primordial follicle pool, which is established in utero and declines irreversibly until menopause. Because ovarian function is dependent on the primordial follicle pool, an understanding of the mechanisms that regulate follicular growth and maintenance of the primordial follicle pool is critical for the development of interventions to prolong the reproductive life span. Multiple pathways related to aging and nutrient-sensing converge in the mammalian ovary to regulate quiescence or activation of primordial follicles. The PI3K/PTEN/AKT/FOXO3 and associated TSC/mTOR pathways are central to the regulation of the primordial follicle pool; however, aging-associated systems such as the insulin-like growth factor-1/growth hormone pathway, and transsulfuration/hydrogen sulfide pathways may also play a role. Additionally, sirtuins aid in maintaining developmental metabolic competence and chromosomal integrity of the oocyte. Here we review the pathways that regulate ovarian reserve and oocyte quality, and discuss geroscience interventions that leverage our understanding of these pathways to promote reproductive longevity.
Collapse
Affiliation(s)
- Natalia Llarena
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Ohio
- Reproductive Endocrinology and Infertility, Cleveland Clinic Women’s Health Institute, Ohio
| | - Christopher Hine
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Ohio
| |
Collapse
|
27
|
Lunenfeld E. Specialty Grand Challenge—Assisted Reproduction. FRONTIERS IN REPRODUCTIVE HEALTH 2021; 3:551499. [PMID: 36304062 PMCID: PMC9580703 DOI: 10.3389/frph.2021.551499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/23/2021] [Indexed: 12/31/2022] Open
|
28
|
Atkinson L, Martin F, Sturmey RG. Intraovarian injection of platelet-rich plasma in assisted reproduction: too much too soon? Hum Reprod 2021; 36:1737-1750. [PMID: 33963408 PMCID: PMC8366566 DOI: 10.1093/humrep/deab106] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/09/2021] [Indexed: 12/13/2022] Open
Abstract
The prospect of ovarian rejuvenation offers the tantalising prospect of treating age-related declines in fertility or in pathological conditions such as premature ovarian failure. The concept of ovarian rejuvenation was invigorated by the indication of the existence of oogonial stem cells (OSCs), which have been shown experimentally to have the ability to differentiate into functional follicles and generate oocytes; however, their clinical potential remains unknown. Furthermore, there is now growing interest in performing ovarian rejuvenation in situ. One proposed approach involves injecting the ovary with platelet rich plasma (PRP). PRP is a component of blood that remains after the in vitro removal of red and white blood cells. It contains blood platelets, tiny anucleate cells of the blood, which are responsible for forming athrombus to prevent bleeding. In addition, PRP contains an array of cytokines and growth factors, as well as a number of small molecules.The utility ofPRP has been investigatedin a range of regenerative medicine approaches and has been shown to induce differentiation of a range of cell types, presumably through the action of cytokines. A handful ofcasereports have described the use of PRP injections into the ovaryin the human, and while these clinical data report promising results, knowledge on the mechanisms and safety of PRP injections into the ovary remain limited.In this article, we summarise some of the physiological detail of platelets and PRP, before reviewing the existing emerging literature in this area. We then propose potential mechanisms by which PRP may be eliciting any effects before reflecting on some considerations for future studies in the area. Importantly, on the basis of our existing knowledge, we suggest that immediate use of PRP in clinical applications is perhaps premature and further fundamental and clinical research on the nature of ovarian insufficiency, as well as the mechanism by which PRP may act on the ovary, is needed to fully understand this promising development.
Collapse
Affiliation(s)
- Lloyd Atkinson
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Francesca Martin
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Roger G Sturmey
- Centre for Atherothrombosis and Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK.,Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, St Mary's Hospital, Manchester, UK
| |
Collapse
|
29
|
Regenerative Medicine Approaches in Bioengineering Female Reproductive Tissues. Reprod Sci 2021; 28:1573-1595. [PMID: 33877644 DOI: 10.1007/s43032-021-00548-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/15/2021] [Indexed: 10/21/2022]
Abstract
Diseases, disorders, and dysfunctions of the female reproductive tract tissues can result in either infertility and/or hormonal imbalance. Current treatment options are limited and often do not result in tissue function restoration, requiring alternative therapeutic approaches. Regenerative medicine offers potential new therapies through the bioengineering of female reproductive tissues. This review focuses on some of the current technologies that could address the restoration of functional female reproductive tissues, including the use of stem cells, biomaterial scaffolds, bio-printing, and bio-fabrication of tissues or organoids. The use of these approaches could also be used to address issues in infertility. Strategies such as cell-based hormone replacement therapy could provide a more natural means of restoring normal ovarian physiology. Engineering of reproductive tissues and organs could serve as a powerful tool for correcting developmental anomalies. Organ-on-a-chip technologies could be used to perform drug screening for personalized medicine approaches and scientific investigations of the complex physiological interactions between the female reproductive tissues and other organ systems. While some of these technologies have already been developed, others have not been translated for clinical application. The continuous evolution of biomaterials and techniques, advances in bioprinting, along with emerging ideas for new approaches, shows a promising future for treating female reproductive tract-related disorders and dysfunctions.
Collapse
|
30
|
Advances in Female Germ Cell Induction from Pluripotent Stem Cells. Stem Cells Int 2021; 2021:8849230. [PMID: 33510796 PMCID: PMC7822693 DOI: 10.1155/2021/8849230] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/15/2020] [Accepted: 12/30/2020] [Indexed: 12/31/2022] Open
Abstract
Germ cells are capable of maintaining species continuity through passing genetic and epigenetic information across generations. Female germ cells mainly develop during the embryonic stage and pass through subsequent developmental stages including primordial germ cells, oogonia, and oocyte. However, due to the limitation of using early human embryos as in vivo research model, in vitro research models are needed to reveal the early developmental process and related mechanisms of female germ cells. After birth, the number of follicles gradually decreases with age. Various conditions which damage ovarian functions would cause premature ovarian failure. Alternative treatments to solve these problems need to be investigated. Germ cell differentiation from pluripotent stem cells in vitro can simulate early embryonic development of female germ cells and clarify unresolved issues during the development process. In addition, pluripotent stem cells could potentially provide promising applications for female fertility preservation after proper in vitro differentiation. Mouse female germ cells have been successfully reconstructed in vitro and delivered to live offspring. However, the derivation of functional human female germ cells has not been fully achieved due to technical limitations and ethical issues. To provide an updated and comprehensive information, this review centers on the major studies on the differentiation of mouse and human female germ cells from pluripotent stem cells and provides references to further studies of developmental mechanisms and potential therapeutic applications of female germ cells.
Collapse
|
31
|
Bhartiya D, Kaushik A. Testicular Stem Cell Dysfunction Due to Environmental Insults Could Be Responsible for Deteriorating Reproductive Health of Men. Reprod Sci 2021; 28:649-658. [PMID: 33409879 DOI: 10.1007/s43032-020-00411-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/22/2020] [Indexed: 02/06/2023]
Abstract
Reproductive health of men has declined over time including reduced semen quality specifically sperm count, increased incidence of infertility, and testicular cancers. Our recent findings suggest that these disease states possibly arise as a result of disruption of testicular stem cells biology by perinatal insults including exposure to endocrine disrupting chemicals. Testicular stem cells include relatively quiescent, very small embryonic-like stem cells (VSELs), and actively dividing spermatogonial stem cells (SSCs). Both VSELs and SSCs express estrogen receptors and are directly vulnerable to endocrine disruption. Exposing mice pups to estradiol (20 μg/pup/day on days 5-7) or diethylstilbestrol (2 μg/pup/day on days 1-5) affected spermatogenesis during adult life with reduced numbers of tubules in stage VIII, tetraploid cells and sperm. These mice were infertile and majority of diethylstilbestrol treated mice revealed testicular cancer-like changes. An increase in VSEL numbers, observed by both flow cytometry and qRT-PCR, was associated with marked reduction of c-KIT positive spermatogonial cells. VSELs undergo epigenetic changes due to endocrine disruption that results in blocked differentiation (impaired spermatogenesis) leading to reduced sperm count and infertility, and their excessive self-renewal initiates cancer-like changes in adult life. Thus, testicular dysgenesis syndrome (TDS) has a stem cell rather than a genetic basis.
Collapse
Affiliation(s)
- Deepa Bhartiya
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India.
| | - Ankita Kaushik
- Stem Cell Biology Department, ICMR-National Institute for Research in Reproductive Health, Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| |
Collapse
|
32
|
Zhang PY, Fan Y, Tan T, Yu Y. Generation of Artificial Gamete and Embryo From Stem Cells in Reproductive Medicine. Front Bioeng Biotechnol 2020; 8:781. [PMID: 32793569 PMCID: PMC7387433 DOI: 10.3389/fbioe.2020.00781] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022] Open
Abstract
In addition to the great growing need for assisted reproduction technologies (ART), additional solutions for patients without functional gametes are strongly needed. Due to ethical restrictions, limited studies can be performed on human gametes and embryos; however, artificial gametes and embryos represent a new hope for clinical application and basic research in the field of reproductive medicine. Here, we provide a review of the research progress and possible application of artificial gametes and embryos from different species, including mice, monkeys and humans. Gametes specification from adult stem cells and embryonic stem cells (ESCs) as well as propagation of stem cells from the reproductive system and from organized embryos, which are similar to blastocysts, have been realized in some nonhuman mammals, but not all achievements can be replicated in humans. This area of research remains noteworthy and requires further study and effort to achieve the reconstitution of the entire cycle of gametogenesis and embryo development in vitro.
Collapse
Affiliation(s)
- Pu-Yao Zhang
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yong Fan
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tao Tan
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.,Yunnan Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China
| | - Yang Yu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology and Key Laboratory of Assisted Reproduction, Ministry of Education, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| |
Collapse
|
33
|
Dompe C, Kranc W, Jopek K, Kowalska K, Ciesiółka S, Chermuła B, Bryja A, Jankowski M, Perek J, Józkowiak M, Moncrieff L, Hutchings G, Janowicz K, Pawelczyk L, Bruska M, Petitte J, Mozdziak P, Kulus M, Piotrowska-Kempisty H, Spaczyński RZ, Nowicki M, Kempisty B. Muscle Cell Morphogenesis, Structure, Development and Differentiation Processes Are Significantly Regulated during Human Ovarian Granulosa Cells In Vitro Cultivation. J Clin Med 2020; 9:jcm9062006. [PMID: 32604796 PMCID: PMC7355984 DOI: 10.3390/jcm9062006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 01/03/2023] Open
Abstract
Granulosa cells (GCs) have many functions and are fundamental for both folliculogenesis and oogenesis, releasing hormones and communicating directly with the oocyte. Long-term in vitro cultures of GCs show significant stem-like characteristics. In the current study, RNA of human ovarian granulosa cells was collected at 1, 7, 15 and 30 days of long-term in vitro culture. Understanding the process of differentiation of GCs towards different cell lineages, as well as the molecular pathways underlying these mechanisms, is fundamental to revealing other possible stemness markers of this type of cell. Identifying new markers of GC plasticity may help to understand the aetiology and recurrence of a wide variety of diseases and health conditions and reveal possible clinical applications of the ovarian tissue cells, affecting not only the reproductive ability but also sex hormone production. Granulosa cells were the subject of this study, as they are readily available as remnant material leftover after in vitro fertilisation procedures and exhibit significant stem-like characteristics in culture. The change in gene expression was investigated through a range of molecular and bioinformatic analyses. Expression microarrays were used, allowing the identification of groups of genes typical of specific cellular pathways. This candidate gene study focused on ontological groups associated with muscle cell morphogenesis, structure, development and differentiation, namely, “muscle cell development”, “muscle cell differentiation”, “muscle contraction”, “muscle organ development”, “muscle organ morphogenesis”, “muscle structure development”, “muscle system process” and “muscle tissue development”. The results showed that the 10 most upregulated genes were keratin 19, oxytocin receptor, connective tissue growth factor, nexilin, myosin light chain kinase, cysteine and glycine-rich protein 3, caveolin 1, actin, activating transcription factor 3 and tropomyosin, while the 10 most downregulated consisted of epiregulin, prostaglandin-endoperoxide synthase 2, transforming growth factor, interleukin, collagen, 5-hydroxytryptmine, interleukin 4, phosphodiesterase, wingless-type MMTV integration site family and SRY-box 9. Moreover, ultrastructural observations showing heterogeneity of granulosa cell population are presented in the study. At least two morphologically different subpopulations were identified: large, light coloured and small, darker cells. The expression of genes belonging to the mentioned ontological groups suggest the potential ability of GCs to differentiate and proliferate toward muscle lineage, showing possible application in muscle regeneration and the treatment of different diseases.
Collapse
Affiliation(s)
- Claudia Dompe
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.D.); (L.M.); (G.H.); (K.J.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (K.J.); (K.K.); (S.C.); (M.N.)
| | - Wiesława Kranc
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (W.K.); (A.B.); (M.J.); (J.P.); (M.B.)
| | - Karol Jopek
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (K.J.); (K.K.); (S.C.); (M.N.)
| | - Katarzyna Kowalska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (K.J.); (K.K.); (S.C.); (M.N.)
| | - Sylwia Ciesiółka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (K.J.); (K.K.); (S.C.); (M.N.)
| | - Błażej Chermuła
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, 33 Polna St., 60-535 Poznan, Poland; (B.C.); (L.P.); (R.Z.S.)
| | - Artur Bryja
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (W.K.); (A.B.); (M.J.); (J.P.); (M.B.)
| | - Maurycy Jankowski
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (W.K.); (A.B.); (M.J.); (J.P.); (M.B.)
| | - Joanna Perek
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (W.K.); (A.B.); (M.J.); (J.P.); (M.B.)
| | - Małgorzata Józkowiak
- Department of Toxicology, Poznan University of Medical Sciences, 30 Dojazd St., 60-631 Poznan, Poland; (M.J.); (H.P.-K.)
| | - Lisa Moncrieff
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.D.); (L.M.); (G.H.); (K.J.)
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (K.J.); (K.K.); (S.C.); (M.N.)
| | - Greg Hutchings
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.D.); (L.M.); (G.H.); (K.J.)
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (W.K.); (A.B.); (M.J.); (J.P.); (M.B.)
| | - Krzysztof Janowicz
- The School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen AB25 2ZD, UK; (C.D.); (L.M.); (G.H.); (K.J.)
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (W.K.); (A.B.); (M.J.); (J.P.); (M.B.)
| | - Leszek Pawelczyk
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, 33 Polna St., 60-535 Poznan, Poland; (B.C.); (L.P.); (R.Z.S.)
| | - Małgorzata Bruska
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (W.K.); (A.B.); (M.J.); (J.P.); (M.B.)
| | - James Petitte
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695, USA;
| | - Paul Mozdziak
- Physiology Graduate Program, North Carolina State University, Raleigh, NC 27695, USA;
| | - Magdalena Kulus
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 1 Lwowska St., 87-100 Toruń, Poland;
| | - Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 30 Dojazd St., 60-631 Poznan, Poland; (M.J.); (H.P.-K.)
| | - Robert Z. Spaczyński
- Division of Infertility and Reproductive Endocrinology, Department of Gynecology, Obstetrics and Gynecological Oncology, Poznan University of Medical Sciences, 33 Polna St., 60-535 Poznan, Poland; (B.C.); (L.P.); (R.Z.S.)
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (K.J.); (K.K.); (S.C.); (M.N.)
| | - Bartosz Kempisty
- Department of Histology and Embryology, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (K.J.); (K.K.); (S.C.); (M.N.)
- Department of Anatomy, Poznan University of Medical Sciences, 6 Święcickiego St., 60-781 Poznan, Poland; (W.K.); (A.B.); (M.J.); (J.P.); (M.B.)
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, 1 Lwowska St., 87-100 Toruń, Poland;
- Department of Obstetrics and Gynecology, University Hospital and Masaryk University, 20 Jihlavská St., 62500 Brno, Czech Republic
- Correspondence: ; Tel.: +48-61-854-6567; Fax: +48-61-854-6568
| |
Collapse
|
34
|
Wartalski K, Gorczyca G, Wiater J, Tabarowski Z, Palus-Chramiec K, Setkowicz Z, Duda M. Efficient generation of neural-like cells from porcine ovarian putative stem cells - morphological characterization and evaluation of their electrophysiological properties. Theriogenology 2020; 155:256-268. [PMID: 32810809 DOI: 10.1016/j.theriogenology.2020.05.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/20/2020] [Accepted: 05/23/2020] [Indexed: 12/20/2022]
Abstract
Until recently, the mammalian ovary was considered to consist of fully differentiated tissues, but evidence for the presence of adult stem cells in this organ appeared. The differentiation potential of these cells, referred to as putative stem cells, is not well defined. Porcine ovarian putative stem cells (poPSCs) were immunomagnetically isolated from postnatal pig ovaries based on the presence of the SSEA-4 surface marker protein. First, they were cultured in the undifferentiated state. After the third passage, a novel 7-day culture method inducing their differentiation into neural-like cells by the addition of forskolin (FSK), retinoic acid (RA) and basic fibroblast growth factor (bFGF) to the culture medium was applied. After 7 days, poPSCs successfully differentiated into neural-like cells, as evidenced by neural morphology and the presence of the neuronal markers nestin, NeuN, and GFAP, as confirmed by immunofluorescence, western blot, and real-time PCR. Electrophysiological analysis of potassium and sodium channel activity (patch clamp) confirmed that they indeed differentiated into neurons. The plasticity of poPSCs offers an excellent opportunity, especially in the field of neuroscience, since they can differentiate into neurons or glial cells. Although poPSCs might not be pluripotent cells, they also escape the rigid classification framework of adult stem cells.
Collapse
Affiliation(s)
- Kamil Wartalski
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland; Department of Histology, Jagiellonian University Medical College, Kopernika 7, 31-034, Krakow, Poland
| | - Gabriela Gorczyca
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Jerzy Wiater
- Department of Histology, Jagiellonian University Medical College, Kopernika 7, 31-034, Krakow, Poland; Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Zbigniew Tabarowski
- Department of Experimental Hematology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Katarzyna Palus-Chramiec
- Department of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Zuzanna Setkowicz
- Department of Neuroanatomy, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland
| | - Małgorzata Duda
- Department of Endocrinology, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University in Krakow, Gronostajowa 9, 30-387, Krakow, Poland.
| |
Collapse
|
35
|
Sabini C, Sorbi F, Cunnea P, Fotopoulou C. Ovarian cancer stem cells: ready for prime time? Arch Gynecol Obstet 2020; 301:895-899. [PMID: 32200419 DOI: 10.1007/s00404-020-05510-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/12/2020] [Indexed: 01/03/2023]
Abstract
INTRODUCTION The role of cancer stem cells (CSC) remains controversial and increasingly subject of investigation as a potential oncogenetic platform with promising therapeutic implications. Understanding the role of CSCs in a highly heterogeneous disease like epithelial ovarian cancer (EOC) may potentially lead to the better understanding of the oncogenetic and metastatic pathways of the disease, but also to develop novel strategies against its progression and platinum resistance. METHODS We have performed a review of all relevant literature that addresses the oncogenetic potential of stem cells in EOC, their mechanisms, and the associated therapeutic targets. RESULTS Cancer stem cells (CSCs) have been reported to be implicated not only in the development and pathways of intratumoral heterogeneity (ITH), but also potentially modulating the tumor microenvironment, leading to the selection of sub-clones resistant to chemotherapy. Furthermore, it appears that the enhanced DNA repair abilities of CSCs are connected with their endurance and resistance maintaining their genomic integrity during novel targeted treatments such as PARP inhibitors, allowing them to survive and causing disease relapse functioning as a tumor seeds. CONCLUSIONS It appears that CSCs play a major role in the underlying mechanisms of oncogenesis and development of relapse in EOC. Part of promising future plans would be to not only use them as therapeutic targets, but also extent their value on a preventative level through engineering mechanisms and prevention of EOC in its origin.
Collapse
Affiliation(s)
- Carlotta Sabini
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, 50134, Florence, Italy
| | - Flavia Sorbi
- Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and Gynecology, University of Florence, 50134, Florence, Italy
| | - Paula Cunnea
- West London Gynecological Cancer Centre, Imperial College NHS Trust, London, W12 OHS, UK.,Department of Surgery and Cancer, Imperial College London, Du Cane Road, London, W12 0HS, UK
| | - Christina Fotopoulou
- West London Gynecological Cancer Centre, Imperial College NHS Trust, London, W12 OHS, UK. .,Department of Surgery and Cancer, Imperial College London, Du Cane Road, London, W12 0HS, UK.
| |
Collapse
|
36
|
Zhang M, Liu L, Cao X, Liu Y, Di J, Huang X, Sun F, Huang W, Xu F. Efficiently accumulating germ-like stem cells from mouse postnatal ovary by in situ tissue culture. Dev Growth Differ 2020; 62:223-231. [PMID: 32189336 DOI: 10.1111/dgd.12656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/15/2020] [Accepted: 01/20/2020] [Indexed: 11/27/2022]
Abstract
Although recent studies have revealed that germline stem cells (GSCs) exist in the mouse postnatal ovary, how to efficiently obtain GSCs for regenerating neo-oogenesis is still a technical challenge. Here, we report that using in situ tissue culture we can efficiently accumulate large amounts of proliferating germ-like cells from mouse postnatal ovaries. Usually, more than 10,000 germ-like cells can be derived from one ovary by this method, and over 20% of these cells can grow into germ-like cells with self-renewal, which thus can serve as a good cell pool to isolate GSCs by other cell assorting methods such as FACS. This method is simple and time-saving, which should be useful for in future studies on mouse GSCs.
Collapse
Affiliation(s)
- Meizi Zhang
- Reproductive Medicine Center, Tianjin First Central Hospital, Tianjin, China
| | - Li Liu
- Reproductive Medicine Center, Tianjin First Central Hospital, Tianjin, China
| | - Xiaomin Cao
- Reproductive Medicine Center, Tianjin First Central Hospital, Tianjin, China
| | - Ye Liu
- Reproductive Medicine Center, Tianjin First Central Hospital, Tianjin, China
| | - Jianyong Di
- Reproductive Medicine Center, Tianjin First Central Hospital, Tianjin, China
| | - Xiuying Huang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Fangzhen Sun
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Weihong Huang
- Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - Fengqin Xu
- Reproductive Medicine Center, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
37
|
Ovarian Cancer, Cancer Stem Cells and Current Treatment Strategies: A Potential Role of Magmas in the Current Treatment Methods. Cells 2020; 9:cells9030719. [PMID: 32183385 PMCID: PMC7140629 DOI: 10.3390/cells9030719] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/01/2020] [Accepted: 03/06/2020] [Indexed: 12/18/2022] Open
Abstract
Epithelial ovarian cancer (EOC) constitutes 90% of ovarian cancers (OC) and is the eighth most common cause of cancer-related death in women. The cancer histologically and genetically is very complex having a high degree of tumour heterogeneity. The pathogenic variability in OC causes significant impediments in effectively treating patients, resulting in a dismal prognosis. Disease progression is predominantly influenced by the peritoneal tumour microenvironment rather than properties of the tumor and is the major contributor to prognosis. Standard treatment of OC patients consists of debulking surgery, followed by chemotherapy, which in most cases end in recurrent chemoresistant disease. This review discusses the different origins of high-grade serous ovarian cancer (HGSOC), the major sub-type of EOC. Tumour heterogeneity, genetic/epigenetic changes, and cancer stem cells (CSC) in facilitating HGSOC progression and their contribution in the circumvention of therapy treatments are included. Several new treatment strategies are discussed including our preliminary proof of concept study describing the role of mitochondria-associated granulocyte macrophage colony-stimulating factor signaling protein (Magmas) in HGSOC and its unique potential role in chemotherapy-resistant disease.
Collapse
|
38
|
Bharti D, Jang SJ, Lee SY, Lee SL, Rho GJ. In Vitro Generation of Oocyte Like Cells and Their In Vivo Efficacy: How Far We have been Succeeded. Cells 2020; 9:E557. [PMID: 32120836 PMCID: PMC7140496 DOI: 10.3390/cells9030557] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 12/15/2022] Open
Abstract
In the last few decades, stem cell therapy has grown as a boon for many pathological complications including female reproductive disorders. In this review, a brief description of available strategies that are related to stem cell-based in vitro oocyte-like cell (OLC) development are given. We have tried to cover all the aspects and latest updates of the in vitro OLC developmental methodologies, marker profiling, available disease models, and in vivo efficacies, with a special focus on mesenchymal stem cells (MSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs) usage. The differentiation abilities of both the ovarian and non-ovarian stem cell sources under various induction conditions have shown different effects on morphological alterations, proliferation- and size-associated developments, hormonal secretions under gonadotropic stimulations, and their neo-oogenesis or folliculogenesis abilities after in vivo transplantations. The attainment of characters like oocyte-like morphology, size expansion, and meiosis initiation have been found to be major obstacles during in vitro oogenesis. A number of reports have either lacked in vivo studies or have shown their functional incapability to produce viable and healthy offspring. Though researchers have gained many valuable insights regarding in vitro gametogenesis, still there are many things to do to make stem cell-derived OLCs fully functional.
Collapse
Affiliation(s)
- Dinesh Bharti
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (D.B.); (S.-J.J.); (S.-Y.L.); (S.-L.L.)
| | - Si-Jung Jang
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (D.B.); (S.-J.J.); (S.-Y.L.); (S.-L.L.)
| | - Sang-Yun Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (D.B.); (S.-J.J.); (S.-Y.L.); (S.-L.L.)
| | - Sung-Lim Lee
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (D.B.); (S.-J.J.); (S.-Y.L.); (S.-L.L.)
| | - Gyu-Jin Rho
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea; (D.B.); (S.-J.J.); (S.-Y.L.); (S.-L.L.)
- Research Institute of Life Sciences, Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|
39
|
Basal characterization and in vitro differentiation of putative stem cells derived from the adult mouse ovary. In Vitro Cell Dev Biol Anim 2020; 56:59-66. [PMID: 31900800 DOI: 10.1007/s11626-019-00411-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 10/14/2019] [Indexed: 10/25/2022]
Abstract
Lately, stem cell approaches have provided new information on reproductive organ function and additionally recommended novel treatment possibilities. The type(s) and differentiation potential of stem cells present in the mammalian ovary are largely unknown; while oogonial stem cells have been reported, we explored the possibility that multipotent stem cells may reside in the ovary and have wide differentiation potential. In this experimental study, homogenates of whole mouse ovaries were sorted using the stem cell surface markers stem cell antigen-1 and stage specific embryonic antigen-1/CD15. Viable double-positive cells 3-10 μm in diameter were evaluated immediately after sorting and after culture using differentiation conditions. Ovarian-derived stem cells were differentiated into the three main cell types: adipocytes, chondrocytes, or osteocytes. The subsequent culture was performed in media containing bone morphogenetic protein 4 (BMP-4) and/or retinoic acid (RA). RA, BMP-4 or the two agents in combination, consistently stimulated germ cell gene expression. RA treatment strongly stimulated germline gene expression and also the development of cells that were morphologically reminiscent of oocytes. The germ cell genes Dazl, Ddx4, Figla, Gdf-9, Nobox, Prdm9, and Sycp-1 were all detected at low levels. Remarkably, treatment with BMP-4 alone significantly increased protein expression of the granulosa cell product anti-Müllerian hormone (AMH). We have shown that an inclusive isolation protocol results in the consistent derivation of multipotent stem cells from the adult ovary; these cells can be differentiated towards the germ cell fate (RA alone), somatic ovarian cell fate as indicated by AMH production (BMP-4 alone), or classical mesenchymal cell types. Taken together, these data suggest the presence of multipotent mesenchymal stem cells in the murine ovary.
Collapse
|
40
|
Zhao F, Wang W. Gengnianchun Recipe Protects Ovarian Reserve of Rats Treated by 4-Vinylcyclohexene Diepoxide via the AKT Pathway. Int J Endocrinol 2020; 2020:9725898. [PMID: 33381174 PMCID: PMC7758144 DOI: 10.1155/2020/9725898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 11/16/2020] [Accepted: 12/01/2020] [Indexed: 12/22/2022] Open
Abstract
Diminished ovarian reserve (DOR) refers to a decrease in the number and quality of oocytes. Western treatment of DOR does not improve the ovarian reserve fundamentally, and the effect is limited. Gengnianchun recipe (GNC) is a traditional Chinese medicine formula originally applied to treat menopausal syndrome but is also found to be effective in treating clinical DOR patients. Here we aim to examine the effect of GNC in a DOR rat model induced by 4-vinylcyclohexene diepoxide (VCD), a chemical that selectively destroys ovarian small preantral follicles, and further investigate the possible mechanisms. Female SD rats were randomly divided into four groups: control group (C), model group (M), high-dose GNC group (H), and low-dose GNC group (L). Rats in M, H, and L were administered with VCD and normal saline, high-dose GNC, and low-dose GNC separately. Rat ovaries were harvested either to conduct HE staining for follicle count, immunohistochemistry, or western blot. We found that high dose of GNC significantly increased the ovarian index and sustained the number of primordial follicles and primary follicles in VCD treated rats. Moreover, high dose of GNC significantly increased the ovarian protein expression of mouse vasa homologue (MVH), anti-Müllerian hormone (AMH), follicle-stimulating hormone receptor (FSHR), and estrogen receptor β (ERβ) compared with that in the model group. Besides, high-dose GNC significantly increased ovarian AKT phosphorylation and the expression of downstream forkhead box O3 (FOXO3a). Proapoptosis proteins of Bax, cleaved caspase-3, and poly ADP-ribose polymerase (PARP) were significantly decreased after high-dose GNC treatment compared with those in the model group. Taken together, these findings suggest that high-dose GNC could protect ovarian reserve against VCD-induced toxicity via the activation of the AKT signaling pathway and reduced cell apoptosis in SD Rats. This effect could either be induced by the increased FSHR signaling or by the nontranscriptional activation of ERβ, which requires further investigation.
Collapse
Affiliation(s)
- Fangui Zhao
- Department of Ultrasound Diagnosis, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
| | - Wenjun Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, China
- Department of Integrated Traditional Chinese Medicine and Western Medicine, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| |
Collapse
|
41
|
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: 3.8] [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
|
42
|
Mariniello K, Ruiz-Babot G, McGaugh EC, Nicholson JG, Gualtieri A, Gaston-Massuet C, Nostro MC, Guasti L. Stem Cells, Self-Renewal, and Lineage Commitment in the Endocrine System. Front Endocrinol (Lausanne) 2019; 10:772. [PMID: 31781041 PMCID: PMC6856655 DOI: 10.3389/fendo.2019.00772] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
The endocrine system coordinates a wide array of body functions mainly through secretion of hormones and their actions on target tissues. Over the last decades, a collective effort between developmental biologists, geneticists, and stem cell biologists has generated a wealth of knowledge related to the contribution of stem/progenitor cells to both organogenesis and self-renewal of endocrine organs. This review provides an up-to-date and comprehensive overview of the role of tissue stem cells in the development and self-renewal of endocrine organs. Pathways governing crucial steps in both development and stemness maintenance, and that are known to be frequently altered in a wide array of endocrine disorders, including cancer, are also described. Crucially, this plethora of information is being channeled into the development of potential new cell-based treatment modalities for endocrine-related illnesses, some of which have made it through clinical trials.
Collapse
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Emily C. McGaugh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - James G. Nicholson
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Angelica Gualtieri
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
43
|
Kenda Suster N, Virant-Klun I. Presence and role of stem cells in ovarian cancer. World J Stem Cells 2019; 11:383-397. [PMID: 31396367 PMCID: PMC6682502 DOI: 10.4252/wjsc.v11.i7.383] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/23/2019] [Accepted: 06/13/2019] [Indexed: 02/06/2023] Open
Abstract
Ovarian cancer is the deadliest gynecological malignancy. It is typically diagnosed at advanced stages of the disease, with metastatic sites disseminated widely within the abdominal cavity. Ovarian cancer treatment is challenging due to high disease recurrence and further complicated pursuant to acquired chemoresistance. Cancer stem cell (CSC) theory proposes that both tumor development and progression are driven by undifferentiated stem cells capable of self-renewal and tumor-initiation. The most recent evidence revealed that CSCs in terms of ovarian cancer are not only responsible for primary tumor growth, metastasis and relapse of disease, but also for the development of chemoresistance. As the elimination of this cell population is critical for increasing treatment success, a deeper understanding of ovarian CSCs pathobiology, including epithelial-mesenchymal transition, signaling pathways and tumor microenvironment, is needed. Finally, before introducing new therapeutic agents for ovarian cancer, targeting CSCs, accurate identification of different ovarian stem cell subpopulations, including the very small embryonic-like stem cells suggested as progenitors, is necessary. To these ends, reliable markers of ovarian CSCs should be identified. In this review, we present the current knowledge and a critical discussion concerning ovarian CSCs and their clinical role.
Collapse
Affiliation(s)
- Natasa Kenda Suster
- Department of Gynecology, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Ljubljana 1000, Slovenia
| | - Irma Virant-Klun
- Department of Human Reproduction, Division of Obstetrics and Gynecology, University Medical Centre Ljubljana, Ljubljana 1000, Slovenia
| |
Collapse
|
44
|
Ai A, Tang Z, Liu Y, Yu S, Li B, Huang H, Wang X, Cao Y, Zhang W. Characterization and identification of human immortalized granulosa cells derived from ovarian follicular fluid. Exp Ther Med 2019; 18:2167-2177. [PMID: 31452708 PMCID: PMC6704934 DOI: 10.3892/etm.2019.7802] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 06/13/2019] [Indexed: 12/21/2022] Open
Abstract
Follicular fluid serves a crucial role in follicular development and oocyte maturation. Increasing evidence indicates that follicular fluid is rich in proteins and functional cells. In addition to oocyte cells, follicular fluid contains granulosa, thecal and ovarian surface epithelial cells. Granulosa cells (GCs) represent the predominant somatic cell type of the ovarian follicle and are involved in steroidogenesis and folliculogenesis. However, the long-term culture of GCs in vitro remains challenging. The present study aimed to extend the culture of GCs in vitro. Human GCs were collected from the follicular fluid of patients included in an in vitro fertilization program and cultured in the presence of conditioned medium obtained from mouse embryonic fibroblasts. GCs were cultured for over a year and 130 passages, and the population doubling time was ~22 h. Cells presented epithelial-like morphology and a cobblestone-like appearance when they reached confluence. Flow cytometric analysis demonstrated that cells expressed CD29, CD166 and CD49f but not CD31, CD34, CD45, CD90, CD105 or CD13. Immunofluorescence staining revealed that cells expressed follicle stimulating hormone receptor, luteinizing hormone receptor and cytochrome P450 aromatase, which was confirmed by reverse transcription-quantitative polymerase chain reaction. In the presence of androstenedione, cells secreted estradiol. In addition, estradiol level was further stimulated by dibutyryl cAMP treatment. In addition, intracellular cAMP and progesterone expression levels were upregulated by follicle stimulating hormone and/or human chorionic gonadotropin. Furthermore, cells survived in severe combined immunodeficiency mice following intra-ovarian injection. Histological analysis revealed that certain cells formed follicle-like structures. The results from the present study suggested that immortalized GCs may be a useful tool for further research on GC and improve the clinical application of drugs such as follicle-stimulating hormone or human chorionic gonadotropin.
Collapse
Affiliation(s)
- Ai Ai
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China.,Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai 200011, P.R. China
| | - Zhengya Tang
- Shanghai Key Laboratory of Tissue Engineering, Shanghai 200011, P.R. China
| | - Yali Liu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai 200011, P.R. China
| | - Sha Yu
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai 200011, P.R. China
| | - Bin Li
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai 200011, P.R. China
| | - He Huang
- Shanghai Key Laboratory of Tissue Engineering, Shanghai 200011, P.R. China
| | - Xiangsheng Wang
- Shanghai Key Laboratory of Tissue Engineering, Shanghai 200011, P.R. China
| | - Yilin Cao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China.,Shanghai Key Laboratory of Tissue Engineering, Shanghai 200011, P.R. China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China.,Shanghai Key Laboratory of Tissue Engineering, Shanghai 200011, P.R. China
| |
Collapse
|
45
|
Similar Population of CD133+ and DDX4+ VSEL-Like Stem Cells Sorted from Human Embryonic Stem Cell, Ovarian, and Ovarian Cancer Ascites Cell Cultures: The Real Embryonic Stem Cells? Cells 2019; 8:cells8070706. [PMID: 31336813 PMCID: PMC6678667 DOI: 10.3390/cells8070706] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022] Open
Abstract
A population of small stem cells with diameters of up to 5 μm resembling very small embryonic-like stem cells (VSELs) were sorted from human embryonic stem cell (hESC) cultures using magnetic-activated cell sorting (MACS) based on the expression of a stem-cell-related marker prominin-1 (CD133). These VSEL-like stem cells had nuclei that almost filled the whole cell volume and expressed stem-cell-related markers (CD133, SSEA-4) and markers of germinal lineage (DDX4/VASA, PRDM14). They were comparable to similar populations of small stem cells sorted from cell cultures of normal ovaries and were the predominant cells in ascites of recurrent ovarian cancer. The sorted populations of CD133+ VSEL-like stem cells were quiescent in vitro, except for ascites, and were highly activated after exposure to valproic acid and follicle-stimulating hormone (FSH), indicating a new tool to study these cells in vitro. These VSEL-like stem cells spontaneously formed clusters resembling tumour-like structures or grew into larger, oocyte-like cells and were differentiated in vitro into adipogenic, osteogenic and neural lineages after sorting. We propose the population of VSEL-like stem cells from hESC cultures as potential original embryonic stem cells, which are present in the human embryo, persist in adult human ovaries from the embryonic period of life and are involved in cancer manifestation.
Collapse
|
46
|
Jiang Y, Zhu D, Liu W, Qin Q, Fang Z, Pan Z. Hedgehog pathway inhibition causes primary follicle atresia and decreases female germline stem cell proliferation capacity or stemness. Stem Cell Res Ther 2019; 10:198. [PMID: 31277696 PMCID: PMC6612207 DOI: 10.1186/s13287-019-1299-5] [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: 03/04/2019] [Revised: 05/23/2019] [Accepted: 06/07/2019] [Indexed: 02/07/2023] Open
Abstract
Background Follicle depletion is one of the causes of premature ovarian failure (POF) and primary ovarian insufficiency (POI). Hence, maintenance of a certain number of female germline stem cells (FGSCs) is optimal to produce oocytes and replenish the primordial follicle pool. The mechanism that regulates proliferation or stemness of FGSCs could contribute to restoring ovarian function, but it remains uncharacterized in postnatal mammalian ovaries. This study aims to investigate the mechanism by which inhibiting the activity of the hedgehog (Hh) signaling pathway regulates follicle development and FGSC proliferation. Methods and results To understand the role of the Hh pathway in ovarian aging, we measured Hh signaling activity at different reproductive ages and the correlation between them in physiological and pathological mice. Furthermore, we evaluated the follicle number and development and the changes in FGSC proliferation or stemness after blocking the Hh pathway in vitro and in vivo. In addition, we aimed to explain one of the mechanisms for the FGSC phenotype changes induced by treatment with the Hh pathway-specific inhibitor GANT61 via oxidative stress and apoptosis. The results show that the activity of Hh signaling is decreased in the ovaries in physiological aging and POF models, which is consistent with the trend of expression levels of the germline stem cell markers Mvh and Oct4. In vitro, blocking the Hh pathway causes follicular developmental disorders and depletes ovarian germ cells and FGSCs after treating ovaries with GANT61. The proliferation or stemness of cultured primary FGSCs is reduced when Hh activity is blocked. Our results show that the antioxidative enzyme level and the ratio of Bcl-2/Bax decrease, the expression level of caspase 3 increases, the mitochondrial membrane potential is abnormal, and ROS accumulate in this system. Conclusions We observed that the inhibition of the Hh signaling pathway with GANT61 could reduce primordial follicle number and decrease FGSC reproductive capacity or stemness through oxidative damage and apoptosis. Electronic supplementary material The online version of this article (10.1186/s13287-019-1299-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu Jiang
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Dantian Zhu
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Wenfeng Liu
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Qiushi Qin
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhi Fang
- Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Zezheng Pan
- Faculty of Basic Medical Science, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China. .,Medical College, Nanchang University, Nanchang, Jiangxi Province, China.
| |
Collapse
|
47
|
Ddx4 + Oogonial Stem Cells in Postmenopausal Women's Ovaries: A Controversial, Undefined Role. Cells 2019; 8:cells8070650. [PMID: 31261822 PMCID: PMC6678385 DOI: 10.3390/cells8070650] [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: 05/09/2019] [Revised: 06/13/2019] [Accepted: 06/20/2019] [Indexed: 12/18/2022] Open
Abstract
Recent studies support the existence of oogonial stem cells (OSCs) in the ovarian cortex of different mammals, including women.These cells are characterized by small size, membrane expression of DEAD(Asp-Glu-Ala-Asp)-box polypeptide-4 (Ddx4), and stemness properties (such as self-renewal and clonal expansion) as well as the ability to differentiate in vitro into oocyte-like cells. However, the discovery of OSCs contrasts with the popular theory that there is a numerically defined oocyte pool for female fertility which undergoes exhaustion with menopause. Indeed, in the ovarian cortex of postmenopausal women OSCs have been detected that possess both viability and capability to differentiate into oocytes, which is similar to those observed in younger patients. The pathophysiological role of this cell population in aged women is still debated since OSCs, under appropriate stimuli, differentiate into somatic cells, and the occurrence of Ddx4+ cells in ovarian tumor samples also suggests their potential involvement in carcinogenesis. Although further investigation into these observations is needed to clarify OSC function in ovary physiology, clinical investigators and researchers studying female infertility are presently focusing on OSCs as a novel opportunity to restore ovarian reserve in both young women undergoing early ovarian failure and cancer survivors experiencing iatrogenic menopause.
Collapse
|
48
|
Sills ES, Wood SH. Autologous activated platelet-rich plasma injection into adult human ovary tissue: molecular mechanism, analysis, and discussion of reproductive response. Biosci Rep 2019; 39:BSR20190805. [PMID: 31092698 PMCID: PMC6549090 DOI: 10.1042/bsr20190805] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/09/2019] [Accepted: 05/14/2019] [Indexed: 01/19/2023] Open
Abstract
In clinical infertility practice, one intractable problem is low (or absent) ovarian reserve which in turn reflects the natural oocyte depletion associated with advancing maternal age. The number of available eggs has been generally thought to be finite and strictly limited, an entrenched and largely unchallenged tenet dating back more than 50 years. In the past decade, it has been suggested that renewable ovarian germline stem cells (GSCs) exist in adults, and that such cells may be utilized as an oocyte source for women seeking to extend fertility. Currently, the issue of whether mammalian females possess such a population of renewable GSCs remains unsettled. The topic is complex and even agreement on a definitive approach to verify the process of 'ovarian rescue' or 're-potentiation' has been elusive. Similarities have been noted between wound healing and ovarian tissue repair following capsule rupture at ovulation. In addition, molecular signaling events which might be necessary to reverse the effects of reproductive ageing seem congruent with changes occurring in tissue injury responses elsewhere. Recently, clinical experience with such a technique based on autologous activated platelet-rich plasma (PRP) treatment of the adult human ovary has been reported. This review summarizes the present state of understanding of the interaction of platelet-derived growth factors with adult ovarian tissue, and the outcome of human reproductive potential following PRP treatment.
Collapse
Affiliation(s)
- E Scott Sills
- Gen 5 Fertility Center, Office for Reproductive Research, Center for Advanced Genetics; San Diego, CA, U.S.A.
- Applied Biotechnology Research Group, University of Westminster; London W1B 2HW, U.K
| | - Samuel H Wood
- Gen 5 Fertility Center, Office for Reproductive Research, Center for Advanced Genetics; San Diego, CA, U.S.A
| |
Collapse
|
49
|
Clarkson YL, Weatherall E, Waterfall M, McLaughlin M, Lu H, Skehel PA, Anderson RA, Telfer EE. Extracellular Localisation of the C-Terminus of DDX4 Confirmed by Immunocytochemistry and Fluorescence-Activated Cell Sorting. Cells 2019; 8:cells8060578. [PMID: 31212843 PMCID: PMC6627596 DOI: 10.3390/cells8060578] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/06/2019] [Accepted: 06/09/2019] [Indexed: 02/03/2023] Open
Abstract
Putative oogonial stem cells (OSCs) have been isolated by fluorescence-activated cell sorting (FACS) from adult human ovarian tissue using an antibody against DEAD-box helicase 4 (DDX4). DDX4 has been reported to be germ cell specific within the gonads and localised intracellularly. White et al. (2012) hypothesised that the C-terminus of DDX4 is localised on the surface of putative OSCs but is internalised during the process of oogenesis. This hypothesis is controversial since it is assumed that RNA helicases function intracellularly with no extracellular expression. To determine whether the C-terminus of DDX4 could be expressed on the cell surface, we generated a novel expression construct to express full-length DDX4 as a DsRed2 fusion protein with unique C- and N-terminal epitope tags. DDX4 and the C-terminal myc tag were detected at the cell surface by immunocytochemistry and FACS of non-permeabilised human embryonic kidney HEK 293T cells transfected with the DDX4 construct. DDX4 mRNA expression was detected in the DDX4-positive sorted cells by RT-PCR. This study clearly demonstrates that the C-terminus of DDX4 can be expressed on the cell surface despite its lack of a conventional membrane-targeting or secretory sequence. These results validate the use of antibody-based FACS to isolate DDX4-positive putative OSCs.
Collapse
Affiliation(s)
- Yvonne L Clarkson
- Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3FF, UK.
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.
- Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, UK.
| | - Emma Weatherall
- Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3FF, UK.
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.
- Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, UK.
| | - Martin Waterfall
- Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, UK.
| | - Marie McLaughlin
- Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3FF, UK.
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.
- Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, UK.
| | - Haojiang Lu
- Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3FF, UK.
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.
- Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, UK.
| | - Paul A Skehel
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| | - Evelyn E Telfer
- Institute of Cell Biology, University of Edinburgh, Edinburgh EH9 3FF, UK.
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK.
- Ashworth Laboratories, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3JR, UK.
| |
Collapse
|
50
|
Virant-Klun I. Functional Testing of Primitive Oocyte-like Cells Developed in Ovarian Surface Epithelium Cell Culture from Small VSEL-like Stem Cells: Can They Be Fertilized One Day? Stem Cell Rev Rep 2019; 14:715-721. [PMID: 29876729 DOI: 10.1007/s12015-018-9832-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Data from the literature show that there are different populations of stem cells present in human adult ovaries, including small stem cells resembling very small embryonic-like stem cells (VSELs). These small ovarian stem cells with diameters of up to 5 μm are present in the ovarian surface epithelium and can grow into bigger, primitive oocyte-like cells that express several markers of a germinal lineage and exhibit pluripotency but not the zona pellucida structure when cultured in vitro. In this report, we present the results of the functional testing of such primitive oocyte-like cells from one patient with premature ovarian failure after insemination with her partners' sperm. Knowing that even immature oocytes collected in an in vitro fertilization program cannot be fertilized naturally, we were only interested in determining whether and how these cells react to added sperm and whether spermatozoa somehow "recognize" them. Interestingly, the primitive oocyte-like cells quickly released a zona pellucida-like structure in the presence of sperm. Two different populations of cells were distinguished, those with a thick and those with a thin zona pellucida-like structure. The primitive oocyte-like cells with a released zona pellucida-like structure expressed the pluripotency-related gene OCT4A (POU5F1) and zona pellucida-related gene ZP3, similar to oocytes obtained from in vitro fertilization but not somatic chondrocytes. In a small proportion of these cells, a single-spermatozoon was observed inside the cytoplasm, but no signs of fertilization were found. These observations may suggest a primitive "cortical reaction". Our data further confirm the presence of germinal stem cells in the ovarian surface epithelium cell culture.
Collapse
Affiliation(s)
- Irma Virant-Klun
- Department of Obstetrics and Gynaecology, University Medical Centre Ljubljana, Slajmerjeva 3, 1000, Ljubljana, Slovenia.
| |
Collapse
|