101
|
Elkouby YM, Mullins MC. Coordination of cellular differentiation, polarity, mitosis and meiosis - New findings from early vertebrate oogenesis. Dev Biol 2017; 430:275-287. [PMID: 28666956 DOI: 10.1016/j.ydbio.2017.06.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 06/23/2017] [Accepted: 06/26/2017] [Indexed: 12/21/2022]
Abstract
A mechanistic dissection of early oocyte differentiation in vertebrates is key to advancing our knowledge of germline development, reproductive biology, the regulation of meiosis, and all of their associated disorders. Recent advances in the field include breakthroughs in the identification of germline stem cells in Medaka, in the cellular architecture of the germline cyst in mice, in a mechanistic dissection of chromosomal pairing and bouquet formation in meiosis in mice, in tracing oocyte symmetry breaking to the chromosomal bouquet of meiosis in zebrafish, and in the biology of the Balbiani body, a universal oocyte granule. Many of the major events in early oogenesis are universally conserved, and some are co-opted for species-specific needs. The chromosomal events of meiosis are of tremendous consequence to gamete formation and have been extensively studied. New light is now being shed on other aspects of early oocyte differentiation, which were traditionally considered outside the scope of meiosis, and their coordination with meiotic events. The emerging theme is of meiosis as a common groundwork for coordinating multifaceted processes of oocyte differentiation. In an accompanying manuscript we describe methods that allowed for investigations in the zebrafish ovary to contribute to these breakthroughs. Here, we review these advances mostly from the zebrafish and mouse. We discuss oogenesis concepts across established model organisms, and construct an inclusive paradigm for early oocyte differentiation in vertebrates.
Collapse
Affiliation(s)
- Yaniv M Elkouby
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mary C Mullins
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
102
|
Richardson SR, Gerdes P, Gerhardt DJ, Sanchez-Luque FJ, Bodea GO, Muñoz-Lopez M, Jesuadian JS, Kempen MJHC, Carreira PE, Jeddeloh JA, Garcia-Perez JL, Kazazian HH, Ewing AD, Faulkner GJ. Heritable L1 retrotransposition in the mouse primordial germline and early embryo. Genome Res 2017; 27:1395-1405. [PMID: 28483779 PMCID: PMC5538555 DOI: 10.1101/gr.219022.116] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 05/02/2017] [Indexed: 12/31/2022]
Abstract
LINE-1 (L1) retrotransposons are a noted source of genetic diversity and disease in mammals. To expand its genomic footprint, L1 must mobilize in cells that will contribute their genetic material to subsequent generations. Heritable L1 insertions may therefore arise in germ cells and in pluripotent embryonic cells, prior to germline specification, yet the frequency and predominant developmental timing of such events remain unclear. Here, we applied mouse retrotransposon capture sequencing (mRC-seq) and whole-genome sequencing (WGS) to pedigrees of C57BL/6J animals, and uncovered an L1 insertion rate of ≥1 event per eight births. We traced heritable L1 insertions to pluripotent embryonic cells and, strikingly, to early primordial germ cells (PGCs). New L1 insertions bore structural hallmarks of target-site primed reverse transcription (TPRT) and mobilized efficiently in a cultured cell retrotransposition assay. Together, our results highlight the rate and evolutionary impact of heritable L1 retrotransposition and reveal retrotransposition-mediated genomic diversification as a fundamental property of pluripotent embryonic cells in vivo.
Collapse
Affiliation(s)
- Sandra R Richardson
- Mater Research Institute-University of Queensland, Woolloongabba QLD 4102, Australia
| | - Patricia Gerdes
- Mater Research Institute-University of Queensland, Woolloongabba QLD 4102, Australia
| | - Daniel J Gerhardt
- Mater Research Institute-University of Queensland, Woolloongabba QLD 4102, Australia.,Invenra, Incorporated, Madison, Wisconsin 53719, USA
| | - Francisco J Sanchez-Luque
- Mater Research Institute-University of Queensland, Woolloongabba QLD 4102, Australia.,Department of Genomic Medicine, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, PTS Granada, 18016 Granada, Spain
| | - Gabriela-Oana Bodea
- Mater Research Institute-University of Queensland, Woolloongabba QLD 4102, Australia
| | - Martin Muñoz-Lopez
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, PTS Granada, 18016 Granada, Spain
| | - J Samuel Jesuadian
- Mater Research Institute-University of Queensland, Woolloongabba QLD 4102, Australia
| | | | - Patricia E Carreira
- Mater Research Institute-University of Queensland, Woolloongabba QLD 4102, Australia
| | | | - Jose L Garcia-Perez
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada-Andalusian Regional Government, PTS Granada, 18016 Granada, Spain.,Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| | - Haig H Kazazian
- Institute of Genetic Medicine and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Adam D Ewing
- Mater Research Institute-University of Queensland, Woolloongabba QLD 4102, Australia
| | - Geoffrey J Faulkner
- Mater Research Institute-University of Queensland, Woolloongabba QLD 4102, Australia.,School of Biomedical Sciences.,Queensland Brain Institute, University of Queensland, Brisbane QLD 4072, Australia
| |
Collapse
|
103
|
Sun YC, Sun XF, Dyce PW, Shen W, Chen H. The role of germ cell loss during primordial follicle assembly: a review of current advances. Int J Biol Sci 2017; 13:449-457. [PMID: 28529453 PMCID: PMC5436565 DOI: 10.7150/ijbs.18836] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/25/2017] [Indexed: 12/27/2022] Open
Abstract
In most female mammals, early germline development begins with the appearance of primordial germ cells (PGCs), and develops to form mature oocytes following several vital processes. It remains well accepted that significant germ cell apoptosis and oocyte loss takes place around the time of birth. The transition of the ovarian environment from fetal to neonatal, coincides with the loss of germ cells and the timing of follicle formation. All told it is common to lose approximately two thirds of germ cells during this transition period. The current consensus is that germ cell loss can be attributed, at least in part, to programmed cell death (PCD). Recently, autophagy has been implicated as playing a part in germ cell loss during the time of parturition. In this review, we discuss the major opinions and mechanisms of mammalian ovarian PCD during the process of germ cell loss. We also pay close attention to the function of autophagy in germ cell loss, and speculate that autophagy may also serve as a critical and necessary process during the establishment of primordial follicle pool.
Collapse
Affiliation(s)
- Yuan-Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling Shaanxi 712100, China
| | - Xiao-Feng Sun
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Paul W Dyce
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA
| | - Wei Shen
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Hong Chen
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling Shaanxi 712100, China
| |
Collapse
|
104
|
Wang YY, Sun YC, Sun XF, Cheng SF, Li B, Zhang XF, De Felici M, Shen W. Starvation at birth impairs germ cell cyst breakdown and increases autophagy and apoptosis in mouse oocytes. Cell Death Dis 2017; 8:e2613. [PMID: 28182014 PMCID: PMC5386484 DOI: 10.1038/cddis.2017.3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 01/02/2017] [Accepted: 01/03/2017] [Indexed: 01/12/2023]
Abstract
The female reproductive lifespan is largely determined by the size of primordial follicle pool, which is established following germ cell cyst breakdown around birth. Almost two-third of oocytes are lost during germ cell cysts breakdown, following autophagic and apoptosis mechanisms. To investigate a possible relationship between germ cell cyst breakdown and nutrition supply, we established a starvation model in mouse pups at birth and evaluated the dynamics of cyst breakdown during nutrient deprivation. Our results showed that after 36 h of starvation between 1.5 and 3 d.p.p., indicators of metabolism both at systemic and ovarian level were significantly altered and the germ cell cyst breakdown markedly decreased. We also found that markers of oxidative stress, autophagy and apoptosis were increased and higher number of oocytes in cyst showing autophagic markers and of TUNEL-positive oocytes and somatic cells were present in the ovaries of starved pups. Moreover, the proliferation of pre-granulosa cells and the expression of the oocyte-specific transcription factor Nobox were decreased in such ovaries. Finally, we observed that the ovaries of the starved pups could recover a normal number of follicles after about 3 weeks from re-feeding. In conclusion, these data indicate that nutrient deficiency at birth can generate a number of adaptive metabolic and oxidative responses in the ovaries causing increased apoptosis both in the somatic cells and oocyte and autophagy mainly in these latter and leading to a delay of germ cell cyst breakdown and follicle assembly.
Collapse
Affiliation(s)
- Yong-Yong Wang
- College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China.,College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Yuan-Chao Sun
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiao-Feng Sun
- College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China.,College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Shun-Feng Cheng
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Bo Li
- Chengguo Station of Animal Husbandry and Veterinary, Laizhou 261437, China
| | - Xi-Feng Zhang
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome 00133, Italy
| | - Wei Shen
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| |
Collapse
|
105
|
Ikami K, Nuzhat N, Lei L. Organelle transport during mouse oocyte differentiation in germline cysts. Curr Opin Cell Biol 2017; 44:14-19. [PMID: 28038435 DOI: 10.1016/j.ceb.2016.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/25/2016] [Accepted: 12/05/2016] [Indexed: 10/20/2022]
Abstract
During mammalian oogenesis, germ cells undergo oocyte differentiation and oocyte development to form mature oocytes that contain essential components for supporting early embryogenesis. However, only a small fraction of germ cells become mature oocytes and the mechanism of this massive germ cell loss has been unclear. Our recent studies suggested that the formation of functional oocytes and germ cell loss are interlinked by a 'nursing' process in germline cysts during oocyte differentiation in mouse fetal ovaries. 80% of the fetal germ cells sacrifice themselves by donating their cytoplasmic contents to the remaining sister germ cells that differentiate into primary oocytes with augmented developmental potential. In this review, we will summarize the process of mouse oocyte differentiation with a particular focus on organelle transport in germline cysts.
Collapse
Affiliation(s)
- Kanako Ikami
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3045, Ann Arbor, MI 48109, United States
| | - Nafisa Nuzhat
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3045, Ann Arbor, MI 48109, United States
| | - Lei Lei
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3045, Ann Arbor, MI 48109, United States.
| |
Collapse
|
106
|
Abstract
The ovary, the female gonad, serves as the source for the germ cells as well as the major supplier of steroid sex hormones. During embryonic development, the primordial germ cells (PGCs) are specified, migrate to the site of the future gonad, and proliferate, forming structures of germ cells nests, which will eventually break down to generate the primordial follicles (PMFs). Each PMF contains an oocyte arrested at the first prophase of meiosis, surrounded by a flattened layer of somatic pre-granulosa cells. Most of the PMFs are kept dormant and only a selected population is activated to join the growing pool of follicles in a process regulated by both intra- and extra-oocyte factors. The PMFs will further develop into secondary pre-antral follicles, a stage which depends on bidirectional communication between the oocyte and the surrounding somatic cells. Many of the signaling molecules involved in this dialog belong to the transforming growth factor β (TGF-β) superfamily. As the follicle continues to develop, a cavity called antrum is formed. The resulting antral follicles relay on the pituitary gonadotropins, follicle-stimulating hormone (FSH), and luteinizing hormone (LH) for their development. Most of the follicles undergo atretic degeneration and only a subset of the antral follicles, known as the dominant follicles, will reach the preovulatory stage at each reproductive cycle, respond to LH, and subsequently ovulate, releasing a fertilizable oocyte. The remaining somatic cells in the raptured follicle will undergo terminal differentiation and form the corpus luteum, which secretes progesterone necessary to maintain pregnancy.
Collapse
|
107
|
Sun MH, Yang M, Xie FY, Wang W, Zhang L, Shen W, Yin S, Ma JY. DNA Double-Strand Breaks Induce the Nuclear Actin Filaments Formation in Cumulus-Enclosed Oocytes but Not in Denuded Oocytes. PLoS One 2017; 12:e0170308. [PMID: 28099474 PMCID: PMC5242499 DOI: 10.1371/journal.pone.0170308] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/02/2017] [Indexed: 01/21/2023] Open
Abstract
As a gamete, oocyte needs to maintain its genomic integrity and passes this haploid genome to the next generation. However, fully-grown mouse oocyte cannot respond to DNA double-strand breaks (DSBs) effectively and it is also unable to repair them before the meiosis resumption. To compensate for this disadvantage and control the DNA repair events, oocyte needs the cooperation with its surrounding cumulus cells. Recently, evidences have shown that nuclear actin filament formation plays roles in cellular DNA DSB repair. To explore whether these nuclear actin filaments are formed in the DNA-damaged oocytes, here, we labeled the filament actins in denuded oocytes (DOs) and cumulus-enclosed oocytes (CEOs). We observed that the nuclear actin filaments were formed only in the DNA-damaged CEOs, but not in DOs. Formation of actin filaments in the nucleus was an event downstream to the DNA damage response. Our data also showed that the removal of cumulus cells led to a reduction in the nuclear actin filaments in oocytes. Knocking down of the Adcy1 gene in cumulus cells did not affect the formation of nuclear actin filaments in oocytes. Notably, we also observed that the nuclear actin filaments in CEOs could be induced by inhibition of gap junctions. From our results, it was confirmed that DNA DSBs induce the nuclear actin filament formation in oocyte and which is controlled by the cumulus cells.
Collapse
Affiliation(s)
- Ming-Hong Sun
- College of Animal Science and Technology, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
| | - Mo Yang
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Feng-Yun Xie
- College of Animal Science and Technology, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
| | - Wei Wang
- College of Animal Science and Technology, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
| | - Lili Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
| | - Wei Shen
- College of Animal Science and Technology, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
- Institute of Reproductive Science, Qingdao Agricultural University, Qingdao, China
| | - Shen Yin
- College of Animal Science and Technology, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
- Institute of Reproductive Science, Qingdao Agricultural University, Qingdao, China
| | - Jun-Yu Ma
- College of Animal Science and Technology, Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
- Institute of Reproductive Science, Qingdao Agricultural University, Qingdao, China
- * E-mail:
| |
Collapse
|
108
|
Abstract
Lineage analysis is widely used because it provides a very powerful tool for characterizing the developmental behavior of the cells in vivo. In this chapter, we describe a particularly informative variant of lineage analysis that we term "single-cell lineage analysis". As in traditional lineage analysis, the method employs a Tamoxifen (Tmx)-inducible CAGCreER mouse line, which is crossed to an R26R reporter line that can be activated by Cre-mediated DNA recombination. However, instead of driving CreER at a high level within a subset of cells defined by a particular promoter, CreER is driven with a generic promoter that is active in essentially all cells throughout the lifespan of the mouse. Specificity comes from using only a very low dose of Tmx so that just a few random, widely separated individual cells undergo recombination and become labeled. The growth and behavior of most such initially marked cells can subsequently be followed over time because each one forms a growing clone of marked cells that does not overlap with other clones due to their rarity. Following individual cell growth patterns provides much more information than can be derived from traditional lineage analysis, which relies on promoter specificity and uses high doses of Tmx that cannot resolve the behavior of single cells. We illustrate the value of single-cell lineage analysis using a recent study of fetal germ cell development and a recent search for female germ-line stem cells in adult mouse ovaries.
Collapse
Affiliation(s)
- Lei Lei
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, 3045 BSRB, Ann Arbor, MI, 48109, USA.
| | - Allan C Spradling
- Department of Embryology, Howard Hughes Medical Institute, Carnegie Institution for Science, 3520 San Martin Dr., Baltimore, MD, 21218, USA.
| |
Collapse
|
109
|
Abstract
Egg or sperm? The mechanism of sexual fate decision in germ cells has been a long‐standing issue in biology. A recent analysis identified foxl3 as a gene that determines the sexual fate decision of germ cells in the teleost fish, medaka. foxl3/Foxl3 acts in female germline stem cells to repress commitment into male fate (spermatogenesis), indicating that the presence of mitotic germ cells in the female is critical for continuous sexual fate decision of germ cells in medaka gonads. Interestingly, foxl3 is found in most vertebrate genomes except for mammals. This provides the interesting possibility that the sexual fate of germ cells in mammals is determined in a different way compared to foxl3‐possessing vertebrates. Considering the fact that germline stem cells are the cells where foxl3 begins to express and sexual fate decision initiates and mammalian ovary does not have typical germline stem cells, the mechanism in mammals may have been co‐evolved with germline stem cell loss in mammalian ovary.
Collapse
Affiliation(s)
- Minoru Tanaka
- Laboratory of Molecular Genetics of Reproduction, National Institute for Basic Biology, Okazaki, Japan
| |
Collapse
|
110
|
Nursing the Follicles. Dev Cell 2016; 37:7-8. [PMID: 27046826 DOI: 10.1016/j.devcel.2016.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In a recent issue of Science, Lei and Spradling (2016) uncover how germ cells differentiate into oocytes in mouse embryos. Mouse germ cells form cysts, in which sister cells nurse the developing oocyte by donating their organelles and cytoplasmic materials.
Collapse
|
111
|
Zhang J, Liu W, Sun X, Kong F, Zhu Y, Lei Y, Su Y, Su Y, Li J. Inhibition of mTOR Signaling Pathway Delays Follicle Formation in Mice. J Cell Physiol 2016; 232:585-595. [PMID: 27301841 DOI: 10.1002/jcp.25456] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/13/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| | - Wenwen Liu
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| | - Xinhui Sun
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| | - Feifei Kong
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| | - Ye Zhu
- Department of Gynecology; Nanjing Maternal and Child Health Hospital; Nanjing Medical University; Nanjing China
| | - Yue Lei
- Department of Gynecology; Nanjing Maternal and Child Health Hospital; Nanjing Medical University; Nanjing China
| | - Youqiang Su
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| | - Yiping Su
- Department of Gynecology; Nanjing Maternal and Child Health Hospital; Nanjing Medical University; Nanjing China
| | - Jing Li
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| |
Collapse
|
112
|
Wear HM, McPike MJ, Watanabe KH. From primordial germ cells to primordial follicles: a review and visual representation of early ovarian development in mice. J Ovarian Res 2016; 9:36. [PMID: 27329176 PMCID: PMC4915180 DOI: 10.1186/s13048-016-0246-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/13/2016] [Indexed: 01/08/2023] Open
Abstract
Background Normal development of reproductive organs is crucial for successful reproduction. In mice the early ovarian developmental process occurs during the embryonic and postnatal period and is regulated through a series of molecular signaling events. Early ovarian development in mice is a seventeen-day process that begins with the rise of six primordial germ cells on embryonic day five (E5) and ends with the formation of primordial follicles on postnatal day two (P2). Results We reviewed the current literature and created a visual representation of early ovarian development that depicts the important molecular events and associated phenotypic outcomes based on primary data. The visual representation shows the timeline of key signaling interactions and regulation of protein expression in different cells involved in ovarian development. The major developmental events were divided into five phases: 1) origin of germ cells and maintenance of pluripotency; 2) primordial germ cell migration; 3) sex differentiation; 4) formation of germ cell nests; and 5) germ cell nest breakdown and primordial follicle formation. Conclusions This review and visual representation provide a summary of the current scientific understanding of the key regulation and signaling during ovarian development and highlights areas needing further study. The visual representation can be used as an educational resource to link molecular events with phenotypic outcomes; serves as a tool to generate new hypotheses and predictions of adverse reproductive outcomes due to perturbations at the molecular and cellular levels; and provides a comprehendible foundation for computational model development and hypothesis testing.
Collapse
Affiliation(s)
- Hannah M Wear
- Institute of Environmental Health, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd. Mail code HRC3, Portland, OR, 97239, USA
| | - Matthew J McPike
- Institute of Environmental Health, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd. Mail code HRC3, Portland, OR, 97239, USA
| | - Karen H Watanabe
- School of Public Health, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd. Mail code GH230, Portland, OR, 97239, USA.
| |
Collapse
|
113
|
Vaithiyanathan K, Liew SH, Zerafa N, Gamage T, Cook M, O’Reilly LA, Bouillet P, Scott CL, Strasser A, Findlay JK, Hutt KJ. BCL2-modifying factor promotes germ cell loss during murine oogenesis. Reproduction 2016; 151:553-62. [DOI: 10.1530/rep-15-0561] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/25/2016] [Indexed: 11/08/2022]
Abstract
Abstract
Apoptosis plays a prominent role during ovarian development by eliminating large numbers of germ cells from the female germ line. However, the precise mechanisms and regulatory proteins involved in germ cell death are yet to be determined. In this study, we characterised the role of the pro-apoptotic BH3-only protein, BCL2-modifying factor (BMF), in germ cell apoptosis in embryonic and neonatal mouse ovaries. BMF protein was immunohistochemically localised to germ cells at embryonic days 15.5 (E15.5) and E17.5 and postnatal day 1 (PN1), coincident with entry into the meiotic prophase, but was undetectable at E13.5, and only present at low levels at PN3 and PN5. Consistent with this expression pattern, loss of BMF in female mice was associated with a decrease in apoptosis at E15.5 and E17.5. Furthermore, increased numbers of germ cells were found in ovaries from Bmf−/− mice compared with WT animals at E15.5 and PN1. However, germ cell numbers were comparable between Bmf−/− and WT ovaries at PN3, PN5 and PN10. Collectively, these data indicate that BMF mediates foetal oocyte loss and its action limits the maximal number of germ cells attained in the developing ovary, but does not influence the number of primordial follicles initially established in ovarian reserve.
Collapse
|
114
|
|
115
|
Lei L, Spradling AC. Mouse oocytes differentiate through organelle enrichment from sister cyst germ cells. Science 2016; 352:95-9. [PMID: 26917595 PMCID: PMC6910648 DOI: 10.1126/science.aad2156] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 02/11/2016] [Indexed: 12/29/2022]
Abstract
Oocytes differentiate in diverse species by receiving organelles and cytoplasm from sister germ cells while joined in germline cysts or syncytia. Mouse primordial germ cells form germline cysts, but the role of cysts in oogenesis is unknown. We find that mouse germ cells receive organelles from neighboring cyst cells and build a Balbiani body to become oocytes, whereas nurselike germ cells die. Organelle movement, Balbiani body formation, and oocyte fate determination are selectively blocked by low levels of microtubule-dependent transport inhibitors. Membrane breakdown within the cyst and an apoptosis-like process are associated with organelle transfer into the oocyte, events reminiscent of nurse cell dumping in Drosophila We propose that cytoplasmic and organelle transport plays an evolutionarily conserved and functionally important role in mammalian oocyte differentiation.
Collapse
Affiliation(s)
- Lei Lei
- Howard Hughes Medical Institute Research Laboratories, Department of Embryology, Carnegie Institution for Science, 3520 San Martin Drive, Baltimore, MD 21218, USA.
| | - Allan C Spradling
- Howard Hughes Medical Institute Research Laboratories, Department of Embryology, Carnegie Institution for Science, 3520 San Martin Drive, Baltimore, MD 21218, USA.
| |
Collapse
|
116
|
Guo K, Li CH, Wang XY, He DJ, Zheng P. Germ stem cells are active in postnatal mouse ovary under physiological conditions. Mol Hum Reprod 2016; 22:316-28. [PMID: 26916381 DOI: 10.1093/molehr/gaw015] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/19/2016] [Indexed: 11/12/2022] Open
Abstract
STUDY HYPOTHESIS Are active ovarian germ stem cells present in postnatal mouse ovaries under physiological conditions? STUDY FINDING Active ovarian germ stem cells exist and function in adult mouse ovaries under physiological conditions. WHAT IS KNOWN ALREADY In vitro studies suggested the existence of germ stem cells in postnatal ovaries of mouse, pig and human. However, in vivo studies provided evidence against the existence of active germ stem cells in postnatal mouse ovaries. Thus, it remains controversial whether such germ stem cells really exist and function in vivo in postnatal mammalian ovaries. STUDY DESIGN, SAMPLES/MATERIALS, METHODS Octamer-binding transcription factor 4 (Oct4)-MerCreMer transgenic mice were crossed with R26R-enhanced yellow fluorescent protein (EYFP) mice to establish a tamoxifen-inducible tracing system so that Oct4-expressing potential ovarian germ stem cells in young adult mice (5-6 weeks old) can be labeled with EYFP. The germ cell activities of DNA replication, mitotic division, entry into meiosis and progression to primordial follicle stage were investigated by means of immunofluorescent staining of ovarian tissues collected at different time points post-tamoxifen injection (1 day, 3 days, 2 months and 4 months). Meiosis entry and primordial follicle formation were also measured by EYFP-labeled single-cell RT-PCR. Germ cell proliferation and mitotic division were examined through 5-bromodeoxyuridine triphosphate incorporation assay. At each time point, ovaries from two to three animals were used for each set of experiment. MAIN RESULTS AND THE ROLE OF CHANCE By labeling the Oct4-expressing small germ cells and tracing their fates for up to 4 months, we observed persistent meiosis entry and primordial follicle replenishment. Furthermore, we captured the transient processes of mitotic DNA replication as well as mitotic division of the marked germ cells at various time periods after tracing. These lines of evidence unambiguously support the presence of active germ stem cells in postnatal ovaries and their function in replenishing primordial follicle pool under physiological conditions. Moreover, we pointed out that Oct4(+) deleted in azoospermia-like (Dazl)(-) but not Oct4(+)Dazl(+) or Oct4(+) DEAD (Asp-Glu-Ala-Asp) Box Polypeptide 4 (Ddx4)(+) cells contain a population of germ stem cells in mouse ovary. LIMITATIONS, REASONS FOR CAUTION This study was conducted in mice. Whether or not the results are applicable to human remain unclear. The future work should aim at identifying the specific ovarian germ stem cell marker and evaluating the significance of these stem cells to normal ovarian function. WIDER IMPLICATIONS OF THE FINDINGS Clarifying the existence of active germ stem cells and their functional significance in postnatal mammalian ovaries could provide new insights in understanding the mechanism of ovarian aging and failure. LARGE SCALE DATA Not applicable. STUDY FUNDING/COMPETING INTERESTS This work was supported by the National Key Basic Research Program of China (grant number 2012CBA01300) and the National Natural Science Foundation of China to P.Z. (31571484). No competing interests are reported.
Collapse
Affiliation(s)
- Kun Guo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiao Chang Dong Lu, Kunming 650223, Yunnan, China Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiao Chang Dong Lu, Kunming 650223, Yunnan, China Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, Yunnan, China
| | - Chao-Hui Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiao Chang Dong Lu, Kunming 650223, Yunnan, China Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiao Chang Dong Lu, Kunming 650223, Yunnan, China Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, Yunnan, China
| | - Xin-Yi Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiao Chang Dong Lu, Kunming 650223, Yunnan, China Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiao Chang Dong Lu, Kunming 650223, Yunnan, China Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, Yunnan, China
| | - Da-Jian He
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiao Chang Dong Lu, Kunming 650223, Yunnan, China Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiao Chang Dong Lu, Kunming 650223, Yunnan, China Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming 650204, Yunnan, China
| | - Ping Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiao Chang Dong Lu, Kunming 650223, Yunnan, China Yunnan Key Laboratory of Animal Reproduction, Kunming Institute of Zoology, Chinese Academy of Sciences, 32 Jiao Chang Dong Lu, Kunming 650223, Yunnan, China
| |
Collapse
|
117
|
Yoshida S. From cyst to tubule: innovations in vertebrate spermatogenesis. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2016; 5:119-31. [PMID: 26305780 PMCID: PMC5049625 DOI: 10.1002/wdev.204] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 06/20/2015] [Accepted: 07/05/2015] [Indexed: 12/15/2022]
Abstract
Although vertebrates share many common traits, their germline development and function exhibit significant divergence. In particular, this article focuses on their spermatogenesis. The fundamental elements that constitute vertebrate spermatogenesis and the evolutionary changes that occurred upon transition from water to land will be discussed. The life-long continuity of spermatogenesis is supported by the function of stem cells. Series of mitotic and meiotic germ cell divisions are 'incomplete' due to incomplete cytokinesis, forming syncytia interconnected via intercellular bridges (ICBs). Throughout this process, germ cells are supported by appropriate microenvironments established primarily by somatic Sertoli cells. In anamniotes (fish and amphibians) spermatogenesis progresses in cysts, in which developing germ cell syncytia are individually encapsulated by Sertoli cells. Accordingly, Sertoli cells undergo turnover with germ cells that they nourish. This mode of cystic spermatogenesis is also observed in nonvertebrates as insects. In amniotes (reptiles, birds, and mammals), however, Sertoli cells do not turn over but comprise a persistent structure of seminiferous tubules. Sertoli cells nourish different stages of germ cells simultaneously in distinct regions of their surface. This function of Sertoli cells is spatiotemporally orchestrated, and the seminiferous epithelial cycle and spermatogenic wave make the seminiferous tubules a high-throughput factory for sperm production. Furthermore, contrary to the organized differentiating cells, undifferentiated spermatogonia that comprise the stem cell compartment exhibit active motion over the basal layer of seminiferous tubules and the frequent breakdown of ICBs. Thus, amniote seminiferous tubules represent a typical facultative (or open) niche environment without a stem cell tethering anatomically defined niche. WIREs Dev Biol 2016, 5:119-131. doi: 10.1002/wdev.204 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Shosei Yoshida
- Division of Germ Cell Biology, National Institute for Basic Biology, National Institutes of Natural Sciences, Okazaki, Japan
| |
Collapse
|
118
|
Sex Specification and Heterogeneity of Primordial Germ Cells in Mice. PLoS One 2015; 10:e0144836. [PMID: 26700643 PMCID: PMC4689518 DOI: 10.1371/journal.pone.0144836] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/23/2015] [Indexed: 11/19/2022] Open
Abstract
In mice, primordial germ cells migrate into the genital ridges by embryonic day 13.5 (E13.5), where they are then subjected to a sex-specific fate with female and male primordial germ cells undergoing mitotic arrest and meiosis, respectively. However, the sex-specific basis of primordial germ cell differentiation is poorly understood. The aim of this study was to investigate the sex-specific features of mouse primordial germ cells. We performed RNA-sequencing (seq) of E13.5 female and male mouse primordial germ cells using next-generation sequencing. We identified 651 and 428 differentially expressed transcripts (>2-fold, P < 0.05) in female and male primordial germ cells, respectively. Of these, many transcription factors were identified. Gene ontology and network analysis revealed differing functions of the identified female- and male-specific genes that were associated with primordial germ cell acquisition of sex-specific properties required for differentiation into germ cells. Furthermore, DNA methylation and ChIP-seq analysis of histone modifications showed that hypomethylated gene promoter regions were bound with H3K4me3 and H3K27me3. Our global transcriptome data showed that in mice, primordial germ cells are decisively assigned to a sex-specific differentiation program by E13.5, which is necessary for the development of vital germ cells.
Collapse
|
119
|
Mu X, Liao X, Chen X, Li Y, Wang M, Shen C, Zhang X, Wang Y, Liu X, He J. DEHP exposure impairs mouse oocyte cyst breakdown and primordial follicle assembly through estrogen receptor-dependent and independent mechanisms. JOURNAL OF HAZARDOUS MATERIALS 2015; 298:232-240. [PMID: 26073378 DOI: 10.1016/j.jhazmat.2015.05.052] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 06/04/2023]
Abstract
Estrogen plays an essential role in the development of mammalian oocytes, and recent studies suggest that it also regulates primordial follicle assembly in the neonatal ovaries. During the last decade, potential exposure of humans and animals to estrogen-like endocrine disrupting chemicals has become a growing concern. In the present study, we focused on the effect of diethylhexyl phthalate (DEHP), a widespread plasticizer with estrogen-like activity, on germ-cell cyst breakdown and primordial follicle assembly in the early ovarian development of mouse. Neonatal mice injected with DEHP displayed impaired cyst breakdown. Using ovary organ cultures, we revealed that impairment was mediated through estrogen receptors (ERs), as ICI 182,780, an efficient antagonist of ER, reversed this DEHP-mediated effect. DEHP exposure reduced the expression of ERβ, progesterone receptor (PR), and Notch2 signaling components. Finally, DEHP reduced proliferation of pregranulosa precursor cells during the process of primordial folliculogenesis. Together, our results indicate that DEHP influences oocyte cyst breakdown and primordial follicle formation through several mechanisms. Therefore, exposure to estrogen-like chemicals during fetal or neonatal development may adversely influence early ovarian development.
Collapse
Affiliation(s)
- Xinyi Mu
- Laboratory of Reproductive Biology, Chongqing Medical University, Chongqing 400016, PR China; Department of Histology and Embryology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, PR China
| | - Xinggui Liao
- Laboratory of Reproductive Biology, Chongqing Medical University, Chongqing 400016, PR China
| | - Xuemei Chen
- Laboratory of Reproductive Biology, Chongqing Medical University, Chongqing 400016, PR China
| | - Yanli Li
- Laboratory of Reproductive Biology, Chongqing Medical University, Chongqing 400016, PR China
| | - Meirong Wang
- Laboratory of Reproductive Biology, Chongqing Medical University, Chongqing 400016, PR China
| | - Cha Shen
- Laboratory of Reproductive Biology, Chongqing Medical University, Chongqing 400016, PR China
| | - Xue Zhang
- Laboratory of Reproductive Biology, Chongqing Medical University, Chongqing 400016, PR China
| | - Yingxiong Wang
- Laboratory of Reproductive Biology, Chongqing Medical University, Chongqing 400016, PR China
| | - Xueqing Liu
- Laboratory of Reproductive Biology, Chongqing Medical University, Chongqing 400016, PR China
| | - Junlin He
- Laboratory of Reproductive Biology, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
120
|
Abstract
The adult mammalian ovary is devoid of definitive germline stem cells. As such, female reproductive senescence largely results from the depletion of a finite ovarian follicle pool that is produced during embryonic development. Remarkably, the crucial nature and regulation of follicle assembly and survival during embryogenesis is just coming into focus. This developmental pathway involves the coordination of meiotic progression and the breakdown of germ cell cysts into individual oocytes housed within primordial follicles. Recent evidence also indicates that genetic and environmental factors can specifically perturb primordial follicle assembly. Here, we review the cellular and molecular mechanisms by which the mammalian ovarian reserve is established, highlighting the presence of a crucial checkpoint that allows survival of only the highest-quality oocytes.
Collapse
Affiliation(s)
- Kathryn J Grive
- Brown University, MCB Graduate Program, Providence, RI 02912, USA
| | | |
Collapse
|
121
|
Luo Y, Schimenti JC. MCM9 deficiency delays primordial germ cell proliferation independent of the ATM pathway. Genesis 2015; 53:678-84. [DOI: 10.1002/dvg.22901] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/14/2015] [Accepted: 09/17/2015] [Indexed: 02/02/2023]
Affiliation(s)
- Yunhai Luo
- Department of Biomedical Sciences; Cornell University; Ithaca New York
| | - John C. Schimenti
- Department of Biomedical Sciences; Cornell University; Ithaca New York
| |
Collapse
|
122
|
Malki S, Tharp ME, Bortvin A. A Whole-Mount Approach for Accurate Quantitative and Spatial Assessment of Fetal Oocyte Dynamics in Mice. Biol Reprod 2015; 93:113. [PMID: 26423126 DOI: 10.1095/biolreprod.115.132118] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/23/2015] [Indexed: 11/01/2022] Open
Abstract
Depletion of oocytes from the embryonic ovary is a key feature of mammalian oogenesis; however, the rational and molecular bases for this phenomenon remain poorly understood. Presently in the field, the most systematic analysis used to understand the effect of a given molecular pathway on fetal oocyte attrition is to count the number of oocytes in ovaries at different stages of development. This analysis is commonly done using a sampling method based on sectioning of the ovary, a technique that includes many laborious steps culminating in an inaccurate estimate of oocyte number contained within that ovary. This inability to generate data that are directly comparable between labs hinders the field and raises questions about the timing and rate of oocyte depletion. Therefore, we set out to implement a robust method that can be easily used by most research laboratories to study the dynamics of oogenesis during fetal mouse ovary development in both normal and experimental conditions. Here we describe an approach to accurately count the total number of oocytes in embryonic ovaries. This method is based on whole-mount immunofluorescence, tissue clearing with sucrose and ScaleA2 reagent, and automatic detection and counting of germ cells in intact ovaries using confocal microscopy and three-dimensional software analyses. We demonstrate the power of the method by assessing variation of fetal oocyte numbers between left and right ovaries and between litters of mice. Finally, we anticipate that the method could be adopted to the analysis of substages of meiotic prophase I and ovarian somatic cells.
Collapse
Affiliation(s)
- Safia Malki
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland
| | - Marla E Tharp
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | - Alex Bortvin
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland
| |
Collapse
|
123
|
Findlay JK, Hutt KJ, Hickey M, Anderson RA. How Is the Number of Primordial Follicles in the Ovarian Reserve Established? Biol Reprod 2015; 93:111. [PMID: 26423124 DOI: 10.1095/biolreprod.115.133652] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/22/2015] [Indexed: 12/15/2022] Open
Abstract
The number of primordial follicles in the ovarian reserve is an important determinant of the length of the ovarian lifespan, and therefore the fertility of an individual. This reserve contains all of the oocytes potentially available for fertilization throughout the fertile lifespan. The maximum number is set during pregnancy or just after birth in most mammalian species; current evidence does not support neofolliculogenesis after the ovarian reserve is established, although this is increasingly being reexamined. Under physiological circumstances, this number will be influenced by the number of primordial germ cells initially specified in the epiblast of the developing embryo, their proliferation during and after migration to the developing gonads, and their death during oogenesis and formation of primordial follicles at nest breakdown. Death of germ cells during the establishment of the ovarian reserve occurs principally by autophagy or apoptosis, although the triggers that initiate these remain elusive. This review outlines the regulatory steps that determine the number of primordial follicles and thus the number of oocytes in the ovarian reserve at birth, using the mouse as the model, interspersed with human data where available. This information has application for understanding the variability in duration of fertility that occurs between normal individuals and with age, in premature ovarian insufficiency, and after chemotherapy or radiotherapy.
Collapse
Affiliation(s)
- John K Findlay
- Centre for Reproductive Biology, Hudson Institute of Medical Research, Clayton, Victoria, Australia Department of Obstetrics & Gynaecology, Monash University, Clayton, Victoria, Australia Department of Obstetrics & Gynaecology, University of Melbourne, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Karla J Hutt
- Centre for Reproductive Biology, Hudson Institute of Medical Research, Clayton, Victoria, Australia Department of Anatomy & Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Martha Hickey
- Department of Obstetrics & Gynaecology, University of Melbourne, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Richard A Anderson
- Medical Research Council Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
124
|
Suzuki H, Kanai-Azuma M, Kanai Y. From Sex Determination to Initial Folliculogenesis in Mammalian Ovaries: Morphogenetic Waves along the Anteroposterior and Dorsoventral Axes. Sex Dev 2015; 9:190-204. [DOI: 10.1159/000440689] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2015] [Indexed: 11/19/2022] Open
|
125
|
Hermann BP, Mutoji KN, Velte EK, Ko D, Oatley JM, Geyer CB, McCarrey JR. Transcriptional and translational heterogeneity among neonatal mouse spermatogonia. Biol Reprod 2015; 92:54. [PMID: 25568304 DOI: 10.1095/biolreprod.114.125757] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Spermatogonial stem cells (SSCs) are a subset of undifferentiated spermatogonia responsible for ongoing spermatogenesis in mammalian testes. Spermatogonial stem cells arise from morphologically homogeneous prospermatogonia, but growing evidence suggests that only a subset of prospermatogonia develops into the foundational SSC pool. This predicts that subtypes of undifferentiated spermatogonia with discrete mRNA and protein signatures should be distinguishable in neonatal testes. We used single-cell quantitative RT-PCR to examine mRNA levels of 172 genes in individual spermatogonia from 6-day postnatal (P6) mouse testes. Cells enriched from P6 testes using the StaPut or THY1(+) magnetic cell sorting methods exhibited considerable heterogeneity in the abundance of specific germ cell and stem cell mRNAs, segregating into one somatic and three distinct spermatogonial clusters. However, P6 Id4-eGFP(+) transgenic spermatogonia, which are known to be enriched for SSCs, were more homogeneous in their mRNA levels, exhibiting uniform levels for the majority of genes examined (122 of 172). Interestingly, these cells displayed nonuniform (50 of 172) expression of a smaller cohort of these genes, suggesting there is substantial heterogeneity even within the Id4-eGFP(+) population. Further, although immunofluorescence staining largely demonstrated conformity between mRNA and protein levels, some proteins were observed in patterns that were disparate from those detected for the corresponding mRNAs in Id4-eGFP(+) spermatogonia (e.g., Kit, Sohlh2, Stra8), suggesting additional heterogeneity is introduced at the posttranscriptional level. Taken together, these data demonstrate the existence of multiple spermatogonial subtypes in P6 mouse testes and raise the intriguing possibility that these subpopulations may correlate with the development of functionally distinct spermatogenic cell types.
Collapse
Affiliation(s)
- Brian P Hermann
- Department of Biology, The University of Texas at San Antonio, San Antonio, Texas
| | - Kazadi N Mutoji
- Department of Biology, The University of Texas at San Antonio, San Antonio, Texas
| | - Ellen K Velte
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Daijin Ko
- Department of Management Science and Statistics, The University of Texas at San Antonio, San Antonio, Texas
| | - Jon M Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, Washington State University, Pullman, Washington
| | - Christopher B Geyer
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, North Carolina East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina
| | - John R McCarrey
- Department of Biology, The University of Texas at San Antonio, San Antonio, Texas
| |
Collapse
|
126
|
Parte S, Patel H, Sriraman K, Bhartiya D. Isolation and characterization of stem cells in the adult mammalian ovary. Methods Mol Biol 2015; 1235:203-29. [PMID: 25388396 DOI: 10.1007/978-1-4939-1785-3_16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Female mammals are born with a fixed pool of germ cells, which does not replenish during adult life. However, this has been recently challenged and adult ovaries produce oocytes throughout adult life just like sperm in the testes. Evidence is accumulating on the presence of ovarian stem cells, but the need for robust protocols to isolate, identify, further characterize, and subject them to various functionality tests is essential. Knowledge about the function and potential of ovarian stem cells is well demonstrated by various groups, but their true identity remains elusive because of the variability in the approaches used to identify them by different groups. In order to address this we have made attempts to compile our protocols to isolate, identify, characterize, and culture the stem cells using different animal models including human. Two distinct populations of stem cells exist in adult mammalian ovary, including very small embryonic-like stem cells (VSELs) and the progenitors termed ovarian germ stem cells (OGSCs). VSELs are relatively quiescent and undergo asymmetric cell division to give rise to OGSCs, which divide rapidly, occasionally form germ cell nests and undergo meiosis and differentiation into oocytes, which are surrounded by granulosa cells to assemble as primordial follicles.
Collapse
Affiliation(s)
- Seema Parte
- Stem Cell Biology Department, National Institute for Research and Reproductive Health (ICMR), Jehangir Merwanji Street, Parel, Mumbai, 400 012, India
| | | | | | | |
Collapse
|
127
|
Makker A, Goel MM, Mahdi AA. PI3K/PTEN/Akt and TSC/mTOR signaling pathways, ovarian dysfunction, and infertility: an update. J Mol Endocrinol 2014; 53:R103-18. [PMID: 25312969 DOI: 10.1530/jme-14-0220] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Abnormalities in ovarian function, including defective oogenesis and folliculogenesis, represent a key female reproductive deficiency. Accumulating evidence in the literature has shown that the PI3K/PTEN/Akt and TSC/mTOR signaling pathways are critical regulators of ovarian function including quiescence, activation, and survival of primordial follicles, granulosa cell proliferation and differentiation, and meiotic maturation of oocytes. Dysregulation of these signaling pathways may contribute to infertility caused by impaired follicular development, intrafollicular oocyte development, and ovulation. This article reviews the current state of knowledge of the functional role of the PI3K/PTEN/Akt and TSC/mTOR pathways during mammalian oogenesis and folliculogenesis and their association with female infertility.
Collapse
Affiliation(s)
- Annu Makker
- Post-Graduate Department of PathologyDepartment of BiochemistryKing George's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Madhu Mati Goel
- Post-Graduate Department of PathologyDepartment of BiochemistryKing George's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Abbas Ali Mahdi
- Post-Graduate Department of PathologyDepartment of BiochemistryKing George's Medical University, Lucknow 226003, Uttar Pradesh, India
| |
Collapse
|
128
|
Bhartiya D, Hinduja I, Patel H, Bhilawadikar R. Making gametes from pluripotent stem cells--a promising role for very small embryonic-like stem cells. Reprod Biol Endocrinol 2014; 12:114. [PMID: 25421462 PMCID: PMC4255929 DOI: 10.1186/1477-7827-12-114] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/01/2014] [Indexed: 01/15/2023] Open
Abstract
The urge to have one's own biological child supersedes any desire in life. Several options have been used to obtain gametes including pluripotent stem cells (embryonic ES and induced pluripotent iPS stem cells); gonadal stem cells (spermatogonial SSCs, ovarian OSCs stem cells), bone marrow, mesenchymal cells and fetal skin. However, the field poses a huge challenge including inefficient existing protocols for differentiation, epigenetic and genetic changes associated with extensive in vitro manipulation and also ethical/regulatory constraints. A tremendous leap in the field occurred using mouse ES and iPS cells wherein they were first differentiated into epiblast-like cells and then primordial germ cell-like cells. These on further development produced sperm, oocytes and live offspring (had associated genetic problems). Evidently differentiating pluripotent stem cells into primordial germ cells (PGCs) remains a major bottleneck. Against this backdrop, we propose that a novel population of pluripotent stem cells termed very small embryonic-like stem cells (VSELs) may serve as an alternative, potential source of autologus gametes, keeping in mind that they are indeed PGCs surviving in adult mammalian ovaries and testes. Both VSELs and PGCs are pluripotent, relatively quiescent because of epigenetic modifications of parentally imprinted genes loci like Igf2-H19 and KCNQ1p57, share several markers like Stella, Fragilis, Mvh, Dppa2, Dppa4, Sall4, Blimp1 and functional receptors. VSELs are localized in the basement membrane of seminiferous tubules in testis and in the ovary surface epithelium. Ovarian stem cells from mouse, rabbit, sheep, marmoset and humans (menopausal women and those with premature ovarian failure) spontaneously differentiate into oocyte-like structures in vitro with no additional requirement of growth factors. Thus a more pragmatic option to obtain autologus gametes may be the pluripotent VSELs and if we could manipulate them in vivo - existing ethical and epigenetic/genetic concerns associated with in vitro culture may also be minimized. The field of oncofertility may undergo a sea-change and existing strategies of cryopreservation of gametes and gonadal tissue for fertility preservation in cancer patients will necessitate a revision. However, first the scientific community needs to arrive at a consensus about VSELs in the gonads and then work towards exploiting their potential.
Collapse
Affiliation(s)
- Deepa Bhartiya
- />Stem Cell Biology Department, National Institute for Research in Reproductive Health (ICMR), Mumbai, 400 012 India
| | - Indira Hinduja
- />Hinduja IVF Centre, PD Hinduja Hospital and Medical Research Centre, Veer Savarkar Marg, Mumbai, 400 016 India
| | - Hiren Patel
- />Stem Cell Biology Department, National Institute for Research in Reproductive Health (ICMR), Mumbai, 400 012 India
| | - Rashmi Bhilawadikar
- />Hinduja IVF Centre, PD Hinduja Hospital and Medical Research Centre, Veer Savarkar Marg, Mumbai, 400 016 India
| |
Collapse
|
129
|
Dutta S, Mark-Kappeler CJ, Hoyer PB, Pepling ME. The Steroid Hormone Environment During Primordial Follicle Formation in
Perinatal Mouse Ovaries1. Biol Reprod 2014; 91:68. [DOI: 10.1095/biolreprod.114.119214] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Affiliation(s)
- Sudipta Dutta
- Department of Biology, Syracuse University, Syracuse, New York
| | | | - Patricia B. Hoyer
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona
| | | |
Collapse
|
130
|
Myers M, Morgan FH, Liew SH, Zerafa N, Gamage TU, Sarraj M, Cook M, Kapic I, Sutherland A, Scott CL, Strasser A, Findlay JK, Kerr JB, Hutt KJ. PUMA regulates germ cell loss and primordial follicle endowment in mice. Reproduction 2014; 148:211-9. [DOI: 10.1530/rep-13-0666] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The number of primordial follicles initially established within the ovary is influenced by the extent of germ cell death during foetal ovarian development, but the mechanisms that mediate this death have not been fully uncovered. In this study, we identified BBC3 (PUMA) (p53 upregulated modulator of apoptosis, also known as BCL2-binding component 3), a pro-apoptotic BH3-only protein belonging to the BCL2 family, as a critical determinant of the number of germ cells during ovarian development. Targeted disruption of the Bbc3 gene revealed a significant increase in the number of germ cells as early as embryonic day 13.5. The number of germ cells remained elevated in Bbc3−/− female mice compared with WT female mice throughout the remainder of embryonic and early postnatal life, resulting in a 1.9-fold increase in the number of primordial follicles in the ovary on postnatal day 10. The increase in the number of germ cells observed in the ovaries of Bbc3−/− mice could not be attributed to the altered proliferative activity of germ cells within the ovaries. Furthermore, BBC3 was found to be not required for the massive germ cell loss that occurs during germ cell nest breakdown. Our data indicate that BBC3 is a critical regulator of germ cell death that acts during the migratory phase of oogenesis or very soon after the arrival of germ cells in the gonad and that BBC3-mediated cell death limits the number of primordial follicles established in the initial ovarian reserve.
Collapse
|
131
|
Malki S, van der Heijden GW, O'Donnell KA, Martin SL, Bortvin A. A role for retrotransposon LINE-1 in fetal oocyte attrition in mice. Dev Cell 2014; 29:521-533. [PMID: 24882376 DOI: 10.1016/j.devcel.2014.04.027] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 03/31/2014] [Accepted: 04/23/2014] [Indexed: 11/24/2022]
Abstract
Fetal oocyte attrition (FOA) is a conserved but poorly understood process of elimination of more than two-thirds of meiotic prophase I (MPI) oocytes before birth. We now implicate retrotransposons LINE-1 (L1), activated during epigenetic reprogramming of the embryonic germline, in FOA in mice. We show that wild-type fetal oocytes possess differential nuclear levels of L1ORF1p, an L1-encoded protein essential for L1 ribonucleoprotein particle (L1RNP) formation and L1 retrotransposition. We demonstrate that experimental elevation of L1 expression correlates with increased MPI defects, FOA, oocyte aneuploidy, and embryonic lethality. Conversely, reverse transcriptase (RT) inhibitor AZT has a profound effect on the FOA dynamics and meiotic recombination, and it implicates an RT-dependent trigger in oocyte elimination in early MPI. We propose that FOA serves to select oocytes with limited L1 activity that are therefore best suited for the next generation.
Collapse
Affiliation(s)
- Safia Malki
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
| | | | - Kathryn A O'Donnell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sandra L Martin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Alex Bortvin
- Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA.
| |
Collapse
|
132
|
Vanorny DA, Prasasya RD, Chalpe AJ, Kilen SM, Mayo KE. Notch signaling regulates ovarian follicle formation and coordinates follicular growth. Mol Endocrinol 2014; 28:499-511. [PMID: 24552588 DOI: 10.1210/me.2013-1288] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Ovarian follicles form through a process in which somatic pregranulosa cells encapsulate individual germ cells from germ cell syncytia. Complementary expression of the Notch ligand, Jagged1, in germ cells and the Notch receptor, Notch2, in pregranulosa cells suggests a role for Notch signaling in mediating cellular interactions during follicle assembly. Using a Notch reporter mouse, we demonstrate that Notch signaling is active within somatic cells of the embryonic ovary, and these cells undergo dramatic reorganization during follicle histogenesis. This coincides with a significant increase in the expression of the ligands, Jagged1 and Jagged2; the receptor, Notch2; and the target genes, Hes1 and Hey2. Histological examination of ovaries from mice with conditional deletion of Jagged1 within germ cells (J1 knockout [J1KO]) or Notch2 within granulosa cells (N2 knockout [N2KO]) reveals changes in follicle dynamics, including perturbations in the primordial follicle pool and antral follicle development. J1KO and N2KO ovaries also contain multi-oocytic follicles, which represent a failure to resolve germ cell syncytia, and follicles with enlarged oocytes but lacking somatic cell growth, signifying a potential role of Notch signaling in follicle activation and the coordination of follicle development. We also observed decreased cell proliferation and increased apoptosis in the somatic cells of both conditional knockout lines. As a consequence of these defects, J1KO female mice are subfertile; however, N2KO female mice remain fertile. This study demonstrates important functions for Jagged1 and Notch2 in the resolution of germ cell syncytia and the coordination of somatic and germ cell growth within follicles of the mouse ovary.
Collapse
Affiliation(s)
- Dallas A Vanorny
- Department of Molecular Biosciences and Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208
| | | | | | | | | |
Collapse
|
133
|
Yang QE, Oatley JM. Spermatogonial stem cell functions in physiological and pathological conditions. Curr Top Dev Biol 2014; 107:235-67. [PMID: 24439809 DOI: 10.1016/b978-0-12-416022-4.00009-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sperm have a vital role in the continuity of a species by contributing genetic information to the next generation. Production of these specialized gametes in numbers sufficient to confer normal fertility occurs via cycling of the spermatogenic lineage, a process referred to as spermatogenesis. Continuity relies on the activities of a self-renewing reservoir of spermatogonial stem cells (SSCs) from which progenitors will arise that transiently amplify in number before committing to a pathway of terminal differentiation. A primary population of SSCs is established during neonatal development from a pool of quiescent gonocyte precursors that forms in embryogenesis. Disruption of this process has dire consequences on maintenance of a cycling spermatogenic lineage in adulthood. At present, the molecular mechanisms underlying initial formation of the SSC pool are largely undefined. However, several transcription factors and posttranscriptional regulators have been identified as important regulators of SSC self-renewal from studies with mutant mouse models and experimental manipulation within primary cultures of mouse SSCs. Importantly, loss of function of these self-renewal factors may be underlying causes of infertility. Furthermore, disruption in the establishment of the SSC state within gonocytes or misregulation of self-renewal may manifest as testicular germ cell tumors in postnatal life.
Collapse
Affiliation(s)
- Qi-En Yang
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Jon M Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA.
| |
Collapse
|
134
|
Jorgensen JS. Defining the neighborhoods that escort the oocyte through its early life events and into a functional follicle. Mol Reprod Dev 2013; 80:960-76. [PMID: 24105719 PMCID: PMC3980676 DOI: 10.1002/mrd.22232] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 08/15/2013] [Indexed: 01/19/2023]
Abstract
The ovary functions to chaperone the most precious cargo for female individuals, the oocyte, thereby allowing the passage of genetic material to subsequent generations. Within the ovary, single oocytes are surrounded by a legion of granulosa cells inside each follicle. These two cell types depend upon one another to support follicle formation and oocyte survival. The infrastructure and events that work together to ultimately form these functional follicles within the ovary are unprecedented, given that the oocyte originates as a cell like all other neighboring cells within the embryo prior to gastrulation. This review discusses the journey of the germ cell in the context of the developing female mouse embryo, with a focus on specific signaling events and cell-cell interactions that escort the primordial germ cell as it is specified into the germ cell fate, migrates through the hindgut into the gonad, differentiates into an oocyte, and culminates upon formation of the primordial and then primary follicle.
Collapse
Affiliation(s)
- Joan S Jorgensen
- Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
135
|
De Felici M, Barrios F. Seeking the origin of female germline stem cells in the mammalian ovary. Reproduction 2013; 146:R125-30. [PMID: 23801781 DOI: 10.1530/rep-13-0069] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The function of female germline stem cells (FGSCs, also called oogonial stem cells) in the adult mammalian ovary is currently debated in the scientific community. As the evidence to support or discard the possible crucial role of this new class of germ cells in mammals has been extensively discussed, in this review, we wonder which could be their origin. We will assume that FGSCs are present in the post-natal ovaries and speculate as to what origin and characteristics such cells could have. We believe that the definition of these features might shed light on future experimental approaches that could clarify the ongoing debate.
Collapse
Affiliation(s)
- Massimo De Felici
- Section of Histology and Embryology, Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy.
| | | |
Collapse
|
136
|
Lim AK, Lorthongpanich C, Chew TG, Tan CWG, Shue YT, Balu S, Gounko N, Kuramochi-Miyagawa S, Matzuk MM, Chuma S, Messerschmidt DM, Solter D, Knowles BB. The nuage mediates retrotransposon silencing in mouse primordial ovarian follicles. Development 2013; 140:3819-25. [PMID: 23924633 DOI: 10.1242/dev.099184] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Mobilization of endogenous retrotransposons can destabilize the genome, an imminent danger during epigenetic reprogramming of cells in the germline. The P-element-induced wimpy testis (PIWI)-interacting RNA (piRNA) pathway is known to silence retrotransposons in the mouse testes. Several piRNA pathway components localize to the unique, germline structure known as the nuage. In this study, we surveyed mouse ovaries and found, for the first time, transient appearance of nuage-like structures in oocytes of primordial follicles. Mouse vasa homolog (MVH), Piwi-like 2 (PIWIL2/MILI) and tudor domain-containing 9 (TDRD9) are present in these structures, whereas aggregates of germ cell protein with ankyrin repeats, sterile alpha motif and leucine zipper (GASZ) localize separately in the cytoplasm. Retrotransposons are silenced in primordial ovarian follicles, and de-repressed upon reduction of piRNA expression in Mvh, Mili or Gasz mutants. However, these null-mutant females, unlike their male counterparts, are fertile, uncoupling retrotransposon activation from sterility.
Collapse
Affiliation(s)
- Ai Khim Lim
- Institute of Medical Biology, A*STAR, 8A Biomedical Grove, Immunos, 138648 Singapore.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
137
|
Female mice lack adult germ-line stem cells but sustain oogenesis using stable primordial follicles. Proc Natl Acad Sci U S A 2013; 110:8585-90. [PMID: 23630252 DOI: 10.1073/pnas.1306189110] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Whether or not mammalian females generate new oocytes during adulthood from germ-line stem cells to sustain the ovarian follicle pool has recently generated controversy. We used a sensitive lineage-labeling system to determine whether stem cells are needed in female adult mice to compensate for follicular losses and to directly identify active germ-line stem cells. Primordial follicles generated during fetal life are highly stable, with a half-life during adulthood of 10 mo, and thus are sufficient to sustain adult oogenesis without a source of renewal. Moreover, in normal mice or following germ-cell depletion with Busulfan, only stable, single oocytes are lineage-labeled, rather than cell clusters indicative of new oocyte formation. Even one germ-line stem cell division per 2 wk would have been detected by our method, based on the kinetics of fetal follicle formation. Thus, adult female mice neither require nor contain active germ-line stem cells or produce new oocytes in vivo.
Collapse
|