51
|
Ikami K, Nuzhat N, Abbott H, Pandoy R, Haky L, Spradling AC, Tanner H, Lei L. Altered germline cyst formation and oogenesis in Tex14 mutant mice. Biol Open 2021; 10:269245. [PMID: 34156079 PMCID: PMC8249907 DOI: 10.1242/bio.058807] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/24/2022] Open
Abstract
During oocyte differentiation in mouse fetal ovaries, sister germ cells are connected by intercellular bridges, forming germline cysts. Within the cyst, primary oocytes form via gaining cytoplasm and organelles from sister germ cells through germ cell connectivity. To uncover the role of intercellular bridges in oocyte differentiation, we analyzed mutant female mice lacking testis-expressed 14 (TEX14), a protein involved in intercellular bridge formation and stabilization. In Tex14 homozygous mutant fetal ovaries, germ cells divide to form a reduced number of cysts in which germ cells remained connected via syncytia or fragmented cell membranes, rather than normal intercellular bridges. Compared with wild-type cysts, homozygous mutant cysts fragmented at a higher frequency and produced a greatly reduced number of primary oocytes with precocious cytoplasmic enrichment and enlarged volume. By contrast, Tex14 heterozygous mutant germline cysts were less fragmented and generate primary oocytes at a reduced size. Moreover, enlarged primary oocytes in homozygous mutants were used more efficiently to sustain folliculogenesis than undersized heterozygous mutant primary oocytes. Our observations directly link the nature of fetal germline cysts to oocyte differentiation and development. Summary: Altered germline cyst formation and fragmentation due to defective germ cell connectivity leads to changes in oocyte differentiation and development in Tex14 mutant mice.
Collapse
Affiliation(s)
- Kanako Ikami
- Buck Institute for Research on Aging, 94949, Novato, CA, USA
| | - Nafisa Nuzhat
- Department of Cell and Developmental Biology, University of Michigan Medical School, 48109, Ann Arbor, MI, USA
| | - Haley Abbott
- Department of Cell and Developmental Biology, University of Michigan Medical School, 48109, Ann Arbor, MI, USA
| | - Ronald Pandoy
- Buck Institute for Research on Aging, 94949, Novato, CA, USA
| | - Lauren Haky
- Buck Institute for Research on Aging, 94949, Novato, CA, USA
| | - Allan C Spradling
- Department of Embryology, Carnegie Institution for Science, 21218, Baltimore, MD, USA
| | - Heather Tanner
- Buck Institute for Research on Aging, 94949, Novato, CA, USA
| | - Lei Lei
- Buck Institute for Research on Aging, 94949, Novato, CA, USA
| |
Collapse
|
52
|
Watanabe R, Sasaki S, Kimura N. Activation of autophagy in early neonatal mice increases primordial follicle number and improves lifelong fertility†. Biol Reprod 2021; 102:399-411. [PMID: 31566206 DOI: 10.1093/biolre/ioz179] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 08/21/2019] [Accepted: 09/14/2019] [Indexed: 12/17/2022] Open
Abstract
The number of stockpiled primordial follicles is thought to be responsible for the fate of female fertility and reproductive lifetime. We previously reported that starvation in nonsuckling early neonatal mice increases the number of primordial follicles with concomitant autophagy activation, suggesting that autophagy may accelerate the formation of primordial follicles. In this study, we attempted to upregulate the numbers of primordial follicles by administering an autophagy inducer and evaluated the progress of primordial follicle formation and their fertility during the life of the mice. To induce autophagy, mice were intraperitoneally injected with the Tat-beclin1 D-11 peptide (0.02 mg/g body weight) at 6-54 h or 60-84 h after birth. In animals that received Tat-beclin 1 D-11 by 54 h after birth, the primordial follicle numbers were significantly increased compared with the control group at 60 h. The ratio of expressed LC3-II/LC3-I proteins was also significantly greater. The numbers of littermates from pregnant females that had been treated with Tat-beclin 1 D-11 were maintained at remarkably greater levels until 10 months old. These results were supported by an abundance of primordial follicles at even 13-15 months old.
Collapse
Affiliation(s)
- Ren Watanabe
- Laboratory of Animal Reproduction, Graduate School of Agricultural Science, Yamagata University, Tsuruoka, Japan.,Japan Society for the Promotion of Science (JSPS) Research Fellowships for Young Scientists, Tokyo, Japan
| | - Sho Sasaki
- Laboratory of Animal Reproduction, Graduate School of Agricultural Science, Yamagata University, Tsuruoka, Japan
| | - Naoko Kimura
- Laboratory of Animal Reproduction, Graduate School of Agricultural Science, Yamagata University, Tsuruoka, Japan
| |
Collapse
|
53
|
Jeedigunta SP, Minenkova AV, Palozzi JM, Hurd TR. Avoiding Extinction: Recent Advances in Understanding Mechanisms of Mitochondrial DNA Purifying Selection in the Germline. Annu Rev Genomics Hum Genet 2021; 22:55-80. [PMID: 34038145 DOI: 10.1146/annurev-genom-121420-081805] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Mitochondria are unusual organelles in that they contain their own genomes, which are kept apart from the rest of the DNA in the cell. While mitochondrial DNA (mtDNA) is essential for respiration and most multicellular life, maintaining a genome outside the nucleus brings with it a number of challenges. Chief among these is preserving mtDNA genomic integrity from one generation to the next. In this review, we discuss what is known about negative (purifying) selection mechanisms that prevent deleterious mutations from accumulating in mtDNA in the germline. Throughout, we focus on the female germline, as it is the tissue through which mtDNA is inherited in most organisms and, therefore, the tissue that most profoundly shapes the genome. We discuss recent progress in uncovering the mechanisms of germline mtDNA selection, from humans to invertebrates.
Collapse
Affiliation(s)
- Swathi P Jeedigunta
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada;
| | - Anastasia V Minenkova
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada;
| | - Jonathan M Palozzi
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada;
| | - Thomas R Hurd
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1M1, Canada;
| |
Collapse
|
54
|
Wang X, Pepling ME. Regulation of Meiotic Prophase One in Mammalian Oocytes. Front Cell Dev Biol 2021; 9:667306. [PMID: 34095134 PMCID: PMC8172968 DOI: 10.3389/fcell.2021.667306] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 04/28/2021] [Indexed: 11/23/2022] Open
Abstract
In female mammals, meiotic prophase one begins during fetal development. Oocytes transition through the prophase one substages consisting of leptotene, zygotene, and pachytene, and are finally arrested at the diplotene substage, for months in mice and years in humans. After puberty, luteinizing hormone induces ovulation and meiotic resumption in a cohort of oocytes, driving the progression from meiotic prophase one to metaphase two. If fertilization occurs, the oocyte completes meiosis two followed by fusion with the sperm nucleus and preparation for zygotic divisions; otherwise, it is passed into the uterus and degenerates. Specifically in the mouse, oocytes enter meiosis at 13.5 days post coitum. As meiotic prophase one proceeds, chromosomes find their homologous partner, synapse, exchange genetic material between homologs and then begin to separate, remaining connected at recombination sites. At postnatal day 5, most of the oocytes have reached the late diplotene (or dictyate) substage of prophase one where they remain arrested until ovulation. This review focuses on events and mechanisms controlling the progression through meiotic prophase one, which include recombination, synapsis and control by signaling pathways. These events are prerequisites for proper chromosome segregation in meiotic divisions; and if they go awry, chromosomes mis-segregate resulting in aneuploidy. Therefore, elucidating the mechanisms regulating meiotic progression is important to provide a foundation for developing improved treatments of female infertility.
Collapse
|
55
|
Frost ER, Taylor G, Baker MA, Lovell-Badge R, Sutherland JM. Establishing and maintaining fertility: the importance of cell cycle arrest. Genes Dev 2021; 35:619-634. [PMID: 33888561 PMCID: PMC8091977 DOI: 10.1101/gad.348151.120] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In this review, Frost et al. summarize the current knowledge on the Cip/Kip family of cyclin-dependent kinase inhibitors in mouse gonad development and highlight new roles for cell cycle inhibitors in controlling and maintaining female fertility. Development of the ovary or testis is required to establish reproductive competence. Gonad development relies on key cell fate decisions that occur early in embryonic development and are actively maintained. During gonad development, both germ cells and somatic cells proliferate extensively, a process facilitated by cell cycle regulation. This review focuses on the Cip/Kip family of cyclin-dependent kinase inhibitors (CKIs) in mouse gonad development. We particularly highlight recent single-cell RNA sequencing studies that show the heterogeneity of cyclin-dependent kinase inhibitors. This diversity highlights new roles for cell cycle inhibitors in controlling and maintaining female fertility.
Collapse
Affiliation(s)
- Emily R Frost
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia.,Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Güneş Taylor
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Mark A Baker
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia
| | - Robin Lovell-Badge
- Stem Cell Biology and Developmental Genetics Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Jessie M Sutherland
- Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, School of Environmental and Life Sciences, University of Newcastle, Callaghan, New South Wales 2308, Australia.,Hunter Medical Research Institute, New Lambton Heights, New South Wales 2305, Australia
| |
Collapse
|
56
|
Bertho S, Clapp M, Banisch TU, Bandemer J, Raz E, Marlow FL. Zebrafish dazl regulates cystogenesis and germline stem cell specification during the primordial germ cell to germline stem cell transition. Development 2021; 148:dev187773. [PMID: 33722898 PMCID: PMC8077517 DOI: 10.1242/dev.187773] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/04/2021] [Indexed: 01/14/2023]
Abstract
Fertility and gamete reserves are maintained by asymmetric divisions of the germline stem cells to produce new stem cells or daughters that differentiate as gametes. Before entering meiosis, differentiating germ cells (GCs) of sexual animals typically undergo cystogenesis. This evolutionarily conserved process involves synchronous and incomplete mitotic divisions of a GC daughter (cystoblast) to generate sister cells connected by intercellular bridges that facilitate the exchange of materials to support rapid expansion of the gamete progenitor population. Here, we investigated cystogenesis in zebrafish and found that early GCs are connected by ring canals, and show that Deleted in azoospermia-like (Dazl), a conserved vertebrate RNA-binding protein (Rbp), is a regulator of this process. Analysis of dazl mutants revealed the essential role of Dazl in regulating incomplete cytokinesis, germline cyst formation and germline stem cell specification before the meiotic transition. Accordingly, dazl mutant GCs form defective ring canals, and ultimately remain as individual cells that fail to differentiate as meiocytes. In addition to promoting cystoblast divisions and meiotic entry, dazl is required for germline stem cell establishment and fertility.
Collapse
Affiliation(s)
- Sylvain Bertho
- Department of Cell, Developmental and Regenerative Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1020 New York, NY 10029-6574, USA
| | - Mara Clapp
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Torsten U. Banisch
- Institute of Cell Biology Center for Molecular Biology of Inflammation, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
- New York University School of Medicine, Department of Cell Biology, New York, NY 10012, USA
| | - Jan Bandemer
- Institute of Cell Biology Center for Molecular Biology of Inflammation, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
| | - Erez Raz
- Institute of Cell Biology Center for Molecular Biology of Inflammation, Von-Esmarch-Str. 56, D-48149 Muenster, Germany
| | - Florence L. Marlow
- Department of Cell, Developmental and Regenerative Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place Box 1020 New York, NY 10029-6574, USA
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
57
|
Soygur B, Jaszczak RG, Fries A, Nguyen DH, Malki S, Hu G, Demir N, Arora R, Laird DJ. Intercellular bridges coordinate the transition from pluripotency to meiosis in mouse fetal oocytes. SCIENCE ADVANCES 2021; 7:7/15/eabc6747. [PMID: 33827806 PMCID: PMC8026130 DOI: 10.1126/sciadv.abc6747] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 02/18/2021] [Indexed: 05/14/2023]
Abstract
Meiosis is critical to generating oocytes and ensuring female fertility; however, the mechanisms regulating the switch from mitotic primordial germ cells to meiotic germ cells are poorly understood. Here, we implicate intercellular bridges (ICBs) in this state transition. We used three-dimensional in toto imaging to map meiotic initiation in the mouse fetal ovary and revealed a radial geometry of this transition that precedes the established anterior-posterior wave. Our studies reveal that appropriate timing of meiotic entry across the ovary and coordination of mitotic-meiotic transition within a cyst depend on the ICB component Tex14, which we show is required for functional cytoplasmic sharing. We find that Tex14 mutants more rapidly attenuate the pluripotency transcript Dppa3 upon meiotic initiation, and Dppa3 mutants undergo premature meiosis similar to Tex14 Together, these results lead to a model that ICBs coordinate and buffer the transition from pluripotency to meiosis through dilution of regulatory factors.
Collapse
Affiliation(s)
- B Soygur
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - R G Jaszczak
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - A Fries
- Biological Imaging Development Center, University of California, San Francisco, San Francisco, CA, USA
| | - D H Nguyen
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
| | - S Malki
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - G Hu
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - N Demir
- Department of Histology and Embryology, Akdeniz University School of Medicine, Antalya, Turkey
| | - R Arora
- Department of Obstetrics, Gynecology and Reproductive Biology, The Institute for Quantitative Health Science and Engineering, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - D J Laird
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Sciences, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
58
|
Abstract
Formation of primordial follicles occurs when germ cell nests break apart and individual oocytes become surrounded by pregranulosa cells. Why mammalian germ cells develop in germ cell nests is not fully understood but recent work has provided evidence that some oocytes serve as nurse cells supporting other oocytes in the cyst. Headway has also been made in understanding interactions that occur between cyst cells that must change as individual oocytes separate to associate with pregranulosa cells. As germ cell nests undergo breakdown some oocytes are lost by programmed cell death that has been attributed to apoptosis, but newer studies have implicated autophagy in counteracting apoptosis to promote cell survival and maintain the ovarian reserve. Work in the past few years has added to already known pathways regulating primordial follicle formation and has identified new players including signaling molecules, transcription factors and RNA binding proteins.
Collapse
|
59
|
Arias Padilla LF, Castañeda-Cortés DC, Rosa IF, Moreno Acosta OD, Hattori RS, Nóbrega RH, Fernandino JI. Cystic proliferation of germline stem cells is necessary to reproductive success and normal mating behavior in medaka. eLife 2021; 10:62757. [PMID: 33646121 PMCID: PMC7946426 DOI: 10.7554/elife.62757] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 02/28/2021] [Indexed: 12/11/2022] Open
Abstract
The production of an adequate number of gametes is necessary for normal reproduction, for which the regulation of proliferation from early gonadal development to adulthood is key in both sexes. Cystic proliferation of germline stem cells is an especially important step prior to the beginning of meiosis; however, the molecular regulators of this proliferation remain elusive in vertebrates. Here, we report that ndrg1b is an important regulator of cystic proliferation in medaka. We generated mutants of ndrg1b that led to a disruption of cystic proliferation of germ cells. This loss of cystic proliferation was observed from embryogenic to adult stages, impacting the success of gamete production and reproductive parameters such as spawning and fertilization. Interestingly, the depletion of cystic proliferation also impacted male sexual behavior, with a decrease of mating vigor. These data illustrate why it is also necessary to consider gamete production capacity in order to analyze reproductive behavior.
Collapse
Affiliation(s)
| | - Diana C Castañeda-Cortés
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Ivana F Rosa
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Omar D Moreno Acosta
- Salmonid Experimental Station at Campos do Jordão, UPD-CJ, Sao Paulo Fisheries Institute (APTA/SAA), Campos do Jordao, Brazil
| | - Ricardo S Hattori
- Reproductive and Molecular Biology Group, Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Rafael H Nóbrega
- Instituto Tecnológico de Chascomús, INTECH (CONICET-UNSAM), Chascomús, Argentina
| | - Juan I Fernandino
- Instituto Tecnológico de Chascomús, INTECH (CONICET-UNSAM), Chascomús, Argentina
| |
Collapse
|
60
|
Meinsohn MC, Hughes CHK, Estienne A, Saatcioglu HD, Pépin D, Duggavathi R, Murphy BD. A role for orphan nuclear receptor liver receptor homolog-1 (LRH-1, NR5A2) in primordial follicle activation. Sci Rep 2021; 11:1079. [PMID: 33441767 PMCID: PMC7807074 DOI: 10.1038/s41598-020-80178-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/17/2020] [Indexed: 02/07/2023] Open
Abstract
Liver receptor homolog-1 (NR5A2) is expressed specifically in granulosa cells of developing ovarian follicles where it regulates the late stages of follicle development and ovulation. To establish its effects earlier in the trajectory of follicular development, NR5A2 was depleted from granulosa cells of murine primordial and primary follicles. Follicle populations were enumerated in neonates at postnatal day 4 (PND4) coinciding with the end of the formation of the primordial follicle pool. The frequency of primordial follicles in PND4 conditional knockout (cKO) ovaries was greater and primary follicles were substantially fewer relative to control (CON) counterparts. Ten-day in vitro culture of PND4 ovaries recapitulated in vivo findings and indicated that CON mice developed primary follicles in the ovarian medulla to a greater extent than did cKO animals. Two subsets of primordial follicles were observed in wildtype ovaries: one that expressed NR5A2 and the second in which the transcript was absent. Neither expressed the mitotic marker. KI-67, indicating their developmental quiescence. RNA sequencing on PND4 demonstrated that loss of NR5A2 induced changes in 432 transcripts, including quiescence markers, inhibitors of follicle activation, and regulators of cellular migration and epithelial-to-mesenchymal transition. These experiments suggest that NR5A2 expression poises primordial follicles for entry into the developing pool.
Collapse
Affiliation(s)
- Marie-Charlotte Meinsohn
- Centre de recherche en reproduction et fertilité, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 7C6, Canada
| | - Camilla H K Hughes
- Centre de recherche en reproduction et fertilité, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 7C6, Canada
| | - Anthony Estienne
- Centre de recherche en reproduction et fertilité, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 7C6, Canada
| | - Hatice D Saatcioglu
- Pediatric Surgical Research Laboratories, Simches Research Center, Massachusetts General Hospital, 185 Cambridge St., Boston, MA, 02114, USA
| | - David Pépin
- Pediatric Surgical Research Laboratories, Simches Research Center, Massachusetts General Hospital, 185 Cambridge St., Boston, MA, 02114, USA
| | - Raj Duggavathi
- Department of Animal Science, McGill University, 21111 Lakeshore Rd., MS1085, Ste-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Bruce D Murphy
- Centre de recherche en reproduction et fertilité, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 7C6, Canada.
| |
Collapse
|
61
|
Communal living: the role of polyploidy and syncytia in tissue biology. Chromosome Res 2021; 29:245-260. [PMID: 34075512 PMCID: PMC8169410 DOI: 10.1007/s10577-021-09664-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/10/2021] [Accepted: 05/16/2021] [Indexed: 01/22/2023]
Abstract
Multicellular organisms are composed of tissues with diverse cell sizes. Whether a tissue primarily consists of numerous, small cells as opposed to fewer, large cells can impact tissue development and function. The addition of nuclear genome copies within a common cytoplasm is a recurring strategy to manipulate cellular size within a tissue. Cells with more than two genomes can exist transiently, such as in developing germlines or embryos, or can be part of mature somatic tissues. Such nuclear collectives span multiple levels of organization, from mononuclear or binuclear polyploid cells to highly multinucleate structures known as syncytia. Here, we review the diversity of polyploid and syncytial tissues found throughout nature. We summarize current literature concerning tissue construction through syncytia and/or polyploidy and speculate why one or both strategies are advantageous.
Collapse
|
62
|
Place NJ, Prado AM, Faykoo-Martinez M, Brieño-Enriquez MA, Albertini DF, Holmes MM. Germ cell nests in adult ovaries and an unusually large ovarian reserve in the naked mole-rat. Reproduction 2021; 161:89-98. [PMID: 33151901 DOI: 10.1530/rep-20-0304] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/05/2020] [Indexed: 01/25/2023]
Abstract
The naked mole-rat (NMR, Heterocephalus glaber) is renowned for its eusociality and exceptionally long lifespan (> 30 y) relative to its small body size (35-40 g). A NMR phenomenon that has received far less attention is that females show no decline in fertility or fecundity into their third decade of life. The age of onset of reproductive decline in many mammalian species is closely associated with the number of germ cells remaining at the age of sexual maturity. We quantified ovarian reserve size in NMRs at the youngest age (6 months) when subordinate females can begin to ovulate after removal from the queen's suppression. We then compared the NMR ovarian reserve size to values for 19 other mammalian species that were previously reported. The NMR ovarian reserve at 6 months of age is exceptionally large at 108,588 ± 69,890 primordial follicles, which is more than 10-fold larger than in mammals of a comparable size. We also observed germ cell nests in ovaries from 6-month-old NMRs, which is highly unusual since breakdown of germ cell nests and the formation of primordial follicles is generally complete by early postnatal life in other mammals. Additionally, we found germ cell nests in young adult NMRs between 1.25 and 3.75 years of age, in both reproductively activated and suppressed females. The unusually large NMR ovarian reserve provides one mechanism to account for this species' protracted fertility. Whether germ cell nests in adult ovaries contribute to the NMR's long reproductive lifespan remains to be determined.
Collapse
Affiliation(s)
- Ned J Place
- Department of Population Medicine & Diagnostic Sciences, Cornell University, Ithaca, New York, USA
| | - Alexandra M Prado
- Department of Population Medicine & Diagnostic Sciences, Cornell University, Ithaca, New York, USA
| | | | - Miguel Angel Brieño-Enriquez
- Department of Obstetrics, Gynecology & Reproductive Medicine, Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David F Albertini
- Department of Reproductive Biology, Bedford Research Foundation, Bedford, Massachusetts, USA
| | - Melissa M Holmes
- Department of Cell & Systems Biology, University of Toronto, Toronto, Ontario, Canada.,Department of Psychology, University of Toronto Mississauga, Mississauga, Ontario, Canada
| |
Collapse
|
63
|
Deshmukh S, Saini S. Phenotypic Heterogeneity in Tumor Progression, and Its Possible Role in the Onset of Cancer. Front Genet 2020; 11:604528. [PMID: 33329751 PMCID: PMC7734151 DOI: 10.3389/fgene.2020.604528] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/10/2020] [Indexed: 12/20/2022] Open
Abstract
Heterogeneity among isogenic cells/individuals has been known for at least 150 years. Even Mendel, working on pea plants, realized that not all tall plants were identical. However, Mendel was more interested in the discontinuous variation between genetically distinct individuals. The concept of environment dictating distinct phenotypes among isogenic individuals has since been shown to impact the evolution of populations in numerous examples at different scales of life. In this review, we discuss how phenotypic heterogeneity and its evolutionary implications exist at all levels of life, from viruses to mammals. In particular, we discuss how a particular disease condition (cancer) is impacted by heterogeneity among isogenic cells, and propose a potential role that phenotypic heterogeneity might play toward the onset of the disease.
Collapse
Affiliation(s)
- Saniya Deshmukh
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Supreet Saini
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
64
|
Martínez-Marchal A, Huang Y, Guillot-Ferriols MT, Ferrer-Roda M, Guixé A, Garcia-Caldés M, Roig I. The DNA damage response is required for oocyte cyst breakdown and follicle formation in mice. PLoS Genet 2020; 16:e1009067. [PMID: 33206637 PMCID: PMC7710113 DOI: 10.1371/journal.pgen.1009067] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 12/02/2020] [Accepted: 08/20/2020] [Indexed: 01/03/2023] Open
Abstract
Mammalian oogonia proliferate without completing cytokinesis, forming cysts. Within these, oocytes differentiate and initiate meiosis, promoting double-strand break (DSBs) formation, which are repaired by homologous recombination (HR) causing the pairing and synapsis of the homologs. Errors in these processes activate checkpoint mechanisms, leading to apoptosis. At the end of prophase I, in contrast with what is observed in spermatocytes, oocytes accumulate unrepaired DSBs. Simultaneously to the cyst breakdown, there is a massive oocyte death, which has been proposed to be necessary to enable the individualization of the oocytes to form follicles. Based upon all the above-mentioned information, we hypothesize that the apparently inefficient HR occurring in the oocytes may be a requirement to first eliminate most of the oocytes and enable cyst breakdown and follicle formation. To test this idea, we compared perinatal ovaries from control and mutant mice for the effector kinase of the DNA Damage Response (DDR), CHK2. We found that CHK2 is required to eliminate ~50% of the fetal oocyte population. Nevertheless, the number of oocytes and follicles found in Chk2-mutant ovaries three days after birth was equivalent to that of the controls. These data revealed the existence of another mechanism capable of eliminating oocytes. In vitro inhibition of CHK1 rescued the oocyte number in Chk2-/- mice, implying that CHK1 regulates postnatal oocyte death. Moreover, we found that CHK1 and CHK2 functions are required for the timely breakdown of the cyst and to form follicles. Thus, we uncovered a novel CHK1 function in regulating the oocyte population in mice. Based upon these data, we propose that the CHK1- and CHK2-dependent DDR controls the number of oocytes and is required to properly break down oocyte cysts and form follicles in mammals.
Collapse
Affiliation(s)
- Ana Martínez-Marchal
- Unitat de Citologia i Histologia, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Grup d'Inestabilitat i Integritat del genoma, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Yan Huang
- Unitat de Citologia i Histologia, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Grup d'Inestabilitat i Integritat del genoma, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Maria Teresa Guillot-Ferriols
- Unitat de Citologia i Histologia, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Grup d'Inestabilitat i Integritat del genoma, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Mònica Ferrer-Roda
- Unitat de Citologia i Histologia, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Grup d'Inestabilitat i Integritat del genoma, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Anna Guixé
- Unitat de Citologia i Histologia, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Grup d'Inestabilitat i Integritat del genoma, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Montserrat Garcia-Caldés
- Grup d'Inestabilitat i Integritat del genoma, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Unitat de Biologia Cel·lular i Genètica Mèdica, Facultat de Medicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Ignasi Roig
- Unitat de Citologia i Histologia, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain.,Grup d'Inestabilitat i Integritat del genoma, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
65
|
Apoptosis in the fetal testis eliminates developmentally defective germ cell clones. Nat Cell Biol 2020; 22:1423-1435. [DOI: 10.1038/s41556-020-00603-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 10/12/2020] [Indexed: 01/22/2023]
|
66
|
Del Pino EM. From egg to embryo in marsupial frogs. Curr Top Dev Biol 2020; 145:91-109. [PMID: 34074537 DOI: 10.1016/bs.ctdb.2020.10.008] [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: 01/04/2023]
Abstract
Marsupial frogs (Hemiphractidae) evolved exceptional mechanisms for the conquest of terrestrial life. These adaptations include very large eggs. In some species eggs reach 10mm in diameter, and are considered to be the largest in frogs. Females have reproductive modifications for the incubation of embryos in their bodies. Modifications of embryos include adaptations for development inside the body of the mother, and changes in the developmental pattern. Moreover, in some species, oocytes are multinucleated instead of having a single germinal vesicle as in most vertebrates. This chapter provides an overview of the adaptations of marsupial frogs associated with terrestrial life, with a discussion of gastrulation and multinucleated oogenesis.
Collapse
Affiliation(s)
- Eugenia M Del Pino
- Escuela de Ciencias Biológicas, Pontificia Universidad Católica del Ecuador, Quito, Ecuador.
| |
Collapse
|
67
|
Abstract
A new study explores the mechanical basis of germline encapsulation in Drosophila gametogenesis, reporting that it is not driven solely by somatic tissue, as previously assumed, but instead relies on actomyosin-generated force in the germline cells.
Collapse
Affiliation(s)
- Tara M Finegan
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Dan T Bergstralh
- Department of Biology, University of Rochester, Rochester, NY 14627, USA; Department of Physics and Astronomy, University of Rochester, Rochester, NY 14627, USA; Department of Biomedical Genetics at the University of Rochester Medical Center, University of Rochester, Rochester, NY 14627, USA.
| |
Collapse
|
68
|
Zhai QY, Wang JJ, Tian Y, Liu X, Song Z. Review of psychological stress on oocyte and early embryonic development in female mice. Reprod Biol Endocrinol 2020; 18:101. [PMID: 33050936 PMCID: PMC7552561 DOI: 10.1186/s12958-020-00657-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/07/2020] [Indexed: 12/22/2022] Open
Abstract
Psychological stress can cause adverse health effects in animals and humans. Accumulating evidence suggests that psychological stress in female mice is associated with ovarian developmental abnormalities accompanied by follicle and oocyte defects. Oocyte and early embryonic development are impaired in mice facing psychological stress, likely resulting from hormone signalling disorders, reactive oxygen species (ROS) accumulation and alterations in epigenetic modifications, which are primarily mediated by the hypothalamic-pituitary-adrenal (HPA) and hypothalamic-pituitary-ovarian (HPO) axes. The present evidence suggests that psychological stress is increasingly becoming the most common causative factor for female subfertility. Here, we review recent progress on the impact of psychological stress on female reproduction, particularly for oocyte and early embryonic development in female mice. This review highlights the connection between psychological stress and reproductive health and provides novel insight on human subfertility.
Collapse
Affiliation(s)
- Qiu-Yue Zhai
- grid.410645.20000 0001 0455 0905School of Basic Medicine, Qingdao University, Qingdao, 266071 China
- grid.410645.20000 0001 0455 0905Qingdao Medical College, Qingdao University, Qingdao, 266071 China
| | - Jun-Jie Wang
- grid.412608.90000 0000 9526 6338College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109 China
| | - Yu Tian
- grid.412608.90000 0000 9526 6338College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, 266109 China
| | - Xiaofang Liu
- grid.43308.3c0000 0000 9413 3760Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, 266071 China
| | - Zhenhua Song
- grid.410645.20000 0001 0455 0905School of Basic Medicine, Qingdao University, Qingdao, 266071 China
- grid.410645.20000 0001 0455 0905Qingdao Medical College, Qingdao University, Qingdao, 266071 China
| |
Collapse
|
69
|
Chanet S, Huynh JR. Collective Cell Sorting Requires Contractile Cortical Waves in Germline Cells. Curr Biol 2020; 30:4213-4226.e4. [PMID: 32916115 DOI: 10.1016/j.cub.2020.08.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 07/01/2020] [Accepted: 08/12/2020] [Indexed: 12/11/2022]
Abstract
Encapsulation of germline cells by layers of somatic cells forms the basic unit of female reproduction called primordial follicles in mammals and egg chambers in Drosophila. How germline and somatic tissues are coordinated for the morphogenesis of each separated unit remains poorly understood. Here, using improved live imaging of Drosophila ovaries, we uncovered periodic actomyosin waves at the cortex of germ cells. These contractile waves are associated with pressure release blebs, which project from germ cells into somatic cells. We demonstrate that these cortical activities, together with cadherin-based adhesion, are required to sort each germline cyst as one collective unit. Genetic perturbations of cortical contractility, bleb protrusion, or adhesion between germline and somatic cells induced encapsulation defects resulting from failures to encapsulate any germ cells, or the inclusion of too many germ cells per egg chamber, or even the mechanical split of germline cysts. Live-imaging experiments revealed that reducing contractility or adhesion in the germline reduced the stiffness of germline cysts and their proper anchoring to the somatic cells. Germline cysts can then be squeezed and passively pushed by constricting surrounding somatic cells, resulting in cyst splitting and cyst collisions during encapsulation. Increasing germline cysts activity or blocking somatic cell constriction movements can reveal active forward migration of germline cysts. Our results show that germ cells play an active role in physical coupling with somatic cells to produce the female gamete.
Collapse
Affiliation(s)
- Soline Chanet
- Center for Interdisciplinary Research in Biology, Collège de France, PSL Research University, CNRS/UMR 7241 - INSERM U1050, 11 Place Marcelin Berthelot, 75005 Paris, France
| | - Jean-René Huynh
- Center for Interdisciplinary Research in Biology, Collège de France, PSL Research University, CNRS/UMR 7241 - INSERM U1050, 11 Place Marcelin Berthelot, 75005 Paris, France.
| |
Collapse
|
70
|
Proteome landscape and spatial map of mouse primordial germ cells. SCIENCE CHINA-LIFE SCIENCES 2020; 64:966-981. [PMID: 32860578 DOI: 10.1007/s11427-020-1762-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/19/2020] [Indexed: 10/23/2022]
Abstract
Primordial germ cells (PGCs) are precursors of both male and female gametes as fundamental materials for organism development. The transcriptome, methylome, and chromatin accessibility profiles of PGCs in both mice and humans have been recently reported. However, little is known about the characteristics of PGCs at the protein levels, which directly exert cellular functions. Here, we construct landscapes of both proteome and 3D spatial distribution of mouse PGCs at E11.5, E13.5 and E16.5 days, the three critical developmental windows for PGCs' sex differentiation, female meiosis initiation and male mitotic arrest. In each developmental stage of PGCs, nearly 2,000-3,000 proteins are identified, among which specific functional pathways such as oxidative phosphorylation, DNA damage repair, and meiotic cell cycle are involved for different events during PGCs development. Interestingly, by 3D modeling we find that PGCs spatially cluster into around 1,300 nests in genital ridge at E11.5 and the nest number is not increased by the exponential proliferation of PGCs. Comparative analysis of our proteomic data with published transcriptomic data does not show a close correlation, meaning that the practically executive factors are beyond the transcriptome. Thus, our work offers a valuable resource for the systematic investigations of PGC development at protein level and spatial map.
Collapse
|
71
|
Two distinct pathways of pregranulosa cell differentiation support follicle formation in the mouse ovary. Proc Natl Acad Sci U S A 2020; 117:20015-20026. [PMID: 32759216 PMCID: PMC7443898 DOI: 10.1073/pnas.2005570117] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This paper improves knowledge of the somatic and germ cells of the developing mouse ovary that assemble into ovarian follicles, by determining cellular gene expression, and tracing lineage relationships. The study covers the last week of fetal development through the first five days of postnatal development. During this time, many critically important processes take place, including sex determination, follicle assembly, and the initial events of meiosis. We report expression differences between pregranulosa cells of wave 1 follicles that function at puberty and wave 2 follicles that sustain fertility. These studies illuminate ovarian somatic cells and provide a resource to study the development, physiology, and evolutionary conservation of mammalian ovarian follicle formation. We sequenced more than 52,500 single cells from embryonic day 11.5 (E11.5) postembryonic day 5 (P5) gonads and performed lineage tracing to analyze primordial follicles and wave 1 medullar follicles during mouse fetal and perinatal oogenesis. Germ cells clustered into six meiotic substages, as well as dying/nurse cells. Wnt-expressing bipotential precursors already present at E11.5 are followed at each developmental stage by two groups of ovarian pregranulosa (PG) cells. One PG group, bipotential pregranulosa (BPG) cells, derives directly from bipotential precursors, expresses Foxl2 early, and associates with cysts throughout the ovary by E12.5. A second PG group, epithelial pregranulosa (EPG) cells, arises in the ovarian surface epithelium, ingresses cortically by E12.5 or earlier, expresses Lgr5, but delays robust Foxl2 expression until after birth. By E19.5, EPG cells predominate in the cortex and differentiate into granulosa cells of quiescent primordial follicles. In contrast, medullar BPG cells differentiate along a distinct pathway to become wave 1 granulosa cells. Reflecting their separate somatic cellular lineages, second wave follicles were ablated by diptheria toxin treatment of Lgr5-DTR-EGFP mice at E16.5 while first wave follicles developed normally and supported fertility. These studies provide insights into ovarian somatic cells and a resource to study the development, physiology, and evolutionary conservation of mammalian ovarian follicles.
Collapse
|
72
|
Grive KJ. Pathways coordinating oocyte attrition and abundance during mammalian ovarian reserve establishment. Mol Reprod Dev 2020; 87:843-856. [PMID: 32720428 DOI: 10.1002/mrd.23401] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022]
Abstract
The mammalian ovarian reserve is comprised of a finite pool of primordial follicles, representing the lifetime reproductive capacity of females. In most mammals, the reserve is produced during embryonic and early postnatal development with oocyte numbers peaking during mid-to-late gestation, and then experiencing a dramatic decline continuing until shortly after birth. Oocytes remaining after the bulk of this attrition are subsequently surrounded by a layer of somatic pre-granulosa cells with these units then referred to as "primordial follicles." The complex and varied cell death mechanisms intrinsic to this process are not only characteristic of, but also essential for, the proper formation of this pool of follicles, and as a result must be immaculately balanced to ensure long-term fertility and reproductive health. Too few follicles can lead to Primary Ovarian Insufficiency, resulting in fertility loss and other features of aging, such as an overall shorter lifespan. On the other hand, whereas an excess of follicles might extend reproductive lifespan, this might also be the underlying etiology of other ovarian pathologies. The last decade, in particular, has vastly expanded our understanding of oocyte attrition and determinants of ovarian reserve abundance. By continuing to decipher the intricacies underlying the cell death processes and development of the initial primordial follicle pool, we may be in a much better position to understand idiopathic cases of premature follicle depletion and improve ovarian health in reproductive-age women.
Collapse
Affiliation(s)
- Kathryn J Grive
- Department of Obstetrics and Gynecology, Program in Women's Oncology, Women and Infants Hospital of Rhode Island, Providence, Rhode Island.,Department of Obstetrics and Gynecology, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
73
|
Hayashi Y, Mori M, Igarashi K, Tanaka K, Takehara A, Ito-Matsuoka Y, Kanai A, Yaegashi N, Soga T, Matsui Y. Proteomic and metabolomic analyses uncover sex-specific regulatory pathways in mouse fetal germline differentiation†. Biol Reprod 2020; 103:717-735. [PMID: 32627815 DOI: 10.1093/biolre/ioaa115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/01/2020] [Accepted: 07/01/2020] [Indexed: 11/13/2022] Open
Abstract
Regulatory mechanisms of germline differentiation have generally been explained via the function of signaling pathways, transcription factors, and epigenetic regulation; however, little is known regarding proteomic and metabolomic regulation and their contribution to germ cell development. Here, we conducted integrated proteomic and metabolomic analyses of fetal germ cells in mice on embryonic day (E)13.5 and E18.5 and demonstrate sex- and developmental stage-dependent changes in these processes. In male germ cells, RNA processing, translation, oxidative phosphorylation, and nucleotide synthesis are dominant in E13.5 and then decline until E18.5, which corresponds to the prolonged cell division and more enhanced hyper-transcription/translation in male primordial germ cells and their subsequent repression. Tricarboxylic acid cycle and one-carbon pathway are consistently upregulated in fetal male germ cells, suggesting their involvement in epigenetic changes preceding in males. Increased protein stability and oxidative phosphorylation during female germ cell differentiation suggests an upregulation of aerobic energy metabolism, which likely contributes to the proteostasis required for oocyte maturation in subsequent stages. The features elucidated in this study shed light on the unrevealed mechanisms of germ cell development.
Collapse
Affiliation(s)
- Yohei Hayashi
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, Miyagi, Japan.,Laboratory of Germ Cell Development, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan.,Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Masaru Mori
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Kaori Igarashi
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Keiko Tanaka
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Asuka Takehara
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, Miyagi, Japan
| | - Yumi Ito-Matsuoka
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, Miyagi, Japan
| | - Akio Kanai
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Yasuhisa Matsui
- Cell Resource Center for Biomedical Research, Institute of Development, Aging and Cancer (IDAC), Tohoku University, Sendai, Miyagi, Japan.,Laboratory of Germ Cell Development, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan.,Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|
74
|
Newly Identified Regulators of Ovarian Folliculogenesis and Ovulation. Int J Mol Sci 2020; 21:ijms21124565. [PMID: 32604954 PMCID: PMC7349727 DOI: 10.3390/ijms21124565] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
Each follicle represents the basic functional unit of the ovary. From its very initial stage of development, the follicle consists of an oocyte surrounded by somatic cells. The oocyte grows and matures to become fertilizable and the somatic cells proliferate and differentiate into the major suppliers of steroid sex hormones as well as generators of other local regulators. The process by which a follicle forms, proceeds through several growing stages, develops to eventually release the mature oocyte, and turns into a corpus luteum (CL) is known as “folliculogenesis”. The task of this review is to define the different stages of folliculogenesis culminating at ovulation and CL formation, and to summarize the most recent information regarding the newly identified factors that regulate the specific stages of this highly intricated process. This information comprises of either novel regulators involved in ovarian biology, such as Ube2i, Phoenixin/GPR73, C1QTNF, and α-SNAP, or recently identified members of signaling pathways previously reported in this context, namely PKB/Akt, HIPPO, and Notch.
Collapse
|
75
|
Mixing and Matching Chromosomes during Female Meiosis. Cells 2020; 9:cells9030696. [PMID: 32178277 PMCID: PMC7140621 DOI: 10.3390/cells9030696] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/08/2020] [Accepted: 03/11/2020] [Indexed: 01/17/2023] Open
Abstract
Meiosis is a key event in the manufacturing of an oocyte. During this process, the oocyte creates a set of unique chromosomes by recombining paternal and maternal copies of homologous chromosomes, and by eliminating one set of chromosomes to become haploid. While meiosis is conserved among sexually reproducing eukaryotes, there is a bewildering diversity of strategies among species, and sometimes within sexes of the same species, to achieve proper segregation of chromosomes. Here, we review the very first steps of meiosis in females, when the maternal and paternal copies of each homologous chromosomes have to move, find each other and pair. We explore the similarities and differences observed in C. elegans, Drosophila, zebrafish and mouse females.
Collapse
|
76
|
Burks DM, McCoy MR, Dutta S, Mark-Kappeler CJ, Hoyer PB, Pepling ME. Molecular analysis of the effects of steroid hormones on mouse meiotic prophase I progression. Reprod Biol Endocrinol 2019; 17:105. [PMID: 31791345 PMCID: PMC6886186 DOI: 10.1186/s12958-019-0548-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/21/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Infertility is linked to depletion of the primordial follicle pool consisting of individual oocytes arrested at the diplotene stage of meiotic prophase I surrounded by granulosa cells. Primordial germ cells, the oocyte precursors, begin to differentiate during embryonic development. These cells migrate to the genital ridge and begin mitotic divisions, remaining connected, through incomplete cytokinesis, in clusters of synchronously dividing oogonia known as germ cell cysts. Subsequently, they enter meiosis, become oocytes and progress through prophase I to the diplotene stage. The cysts break apart, allowing individual oocytes to be surrounded by a layer of granulosa cells, forming primordial follicles each containing a diplotene arrested oocyte. A large number of oocytes are lost coincident with cyst breakdown, and may be important for quality control of primordial follicle formation. Exposure of developing ovaries to exogenous hormones can disrupt cyst breakdown and follicle formation, but it is unclear if hormones affect progression of oocytes through prophase I of meiosis. METHODS Fetal ovaries were treated in organ culture with estradiol, progesterone, or both hormones, labeled for MSY2 or Synaptonemal complex protein 3 (SYCP3) using whole mount immunocytochemistry and examined by confocal microscopy. Meiotic prophase I progression was also followed using the meiotic surface spread technique. RESULTS MSY2 expression in oocytes was reduced by progesterone but not estradiol or the hormone combination. However, while MSY2 expression was upregulated during development it was not a precise marker for the diplotene stage. We also followed meiotic prophase I progression using antibodies against SYCP3 using two different methods, and found that the percent of oocytes at the pachytene stage peaked at postnatal day 1. Finally, estradiol and progesterone treatment together but not either alone in organ culture increased the percent of oocytes at the pachytene stage. CONCLUSIONS We set out to examine the effects of hormones on prophase I progression and found that while MSY2 expression was reduced by progesterone, MSY2 was not a precise diplotene stage marker. Using antibodies against SYCP3 to identify pachytene stage oocytes we found that progesterone and estradiol together delayed progression of oocytes through prophase I.
Collapse
Affiliation(s)
- Deion M Burks
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Margaret R McCoy
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
| | - Sudipta Dutta
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA
- Present address: University of Nebraska Medical Center, Omaha, NE, USA
| | - Connie J Mark-Kappeler
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, AZ, USA
- Present address: PRA Health Sciences Lenexa, Lenexa, KS, USA
| | - Patricia B Hoyer
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Melissa E Pepling
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY, 13244, USA.
| |
Collapse
|
77
|
Capel B. WOMEN IN REPRODUCTIVE SCIENCE: To be or not to be a testis. Reproduction 2019; 158:F101-F111. [PMID: 31265995 PMCID: PMC9945370 DOI: 10.1530/rep-19-0151] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/25/2019] [Indexed: 11/08/2022]
Abstract
Work that established the testis as the driver of male development, and the Y chromosome as the bearer of the male-determining gene, established a working model, and set the stage for the molecular age of mammalian sex determination. The discovery and characterization of Sry/SRY at the top of the hierarchy in mammals launched the field in two major directions. The first was to identify the downstream transcription factors and other molecular players that drive the bifurcation of Sertoli and granulosa cell differentiation. The second major direction was to understand organogenesis of the early bipotential gonad, and how divergence of its two distinct morphogenetic pathways (testis and ovary) is regulated at the cellular level. This review will summarize the early discoveries soon after Sry was identified and focus on my study of the gonad as a model of organogenesis.
Collapse
Affiliation(s)
- Blanche Capel
- 1Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
78
|
Rossi LF, Nottola S, Miglietta S, Macchiarelli G, Luaces JP, Merico V, Merani S, Garagna S, Zuccotti M. Germ cell cysts, a fetal feature in mammals, are constitutively present in the adult armadillo. Mol Reprod Dev 2019; 87:91-101. [DOI: 10.1002/mrd.23296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/30/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Luis Francisco Rossi
- Laboratorio de Biología Cromosómica, Facultad de MedicinaUniversidad de Buenos AiresBuenos Aires Argentina
- Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires Argentina
| | - Stefania Nottola
- Department of Anatomy, Histology, Forensic Medicine and OrthopedicsUniversity of Rome La SapienzaRome Italy
| | - Selenia Miglietta
- Department of Anatomy, Histology, Forensic Medicine and OrthopedicsUniversity of Rome La SapienzaRome Italy
| | - Guido Macchiarelli
- Department of Life, Health and Environmental SciencesUniversity of L'aquilaL'aquila Italy
| | - Juan Pablo Luaces
- Laboratorio de Biología Cromosómica, Facultad de MedicinaUniversidad de Buenos AiresBuenos Aires Argentina
- Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires Argentina
| | - Valeria Merico
- Department of Biology and Biotechnology “Lazzaro Spallanzani,”University of PaviaPavia Italy
- Centre for Health TechnologyUniversity of PaviaPavia Italy
| | - Susana Merani
- Laboratorio de Biología Cromosómica, Facultad de MedicinaUniversidad de Buenos AiresBuenos Aires Argentina
- Consejo Nacional de Investigaciones Científicas y TécnicasBuenos Aires Argentina
| | - Silvia Garagna
- Department of Biology and Biotechnology “Lazzaro Spallanzani,”University of PaviaPavia Italy
- Centre for Health TechnologyUniversity of PaviaPavia Italy
| | - Maurizio Zuccotti
- Department of Biology and Biotechnology “Lazzaro Spallanzani,”University of PaviaPavia Italy
- Centre for Health TechnologyUniversity of PaviaPavia Italy
| |
Collapse
|
79
|
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: 2.8] [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
|
80
|
Rosario R, Crichton JH, Stewart HL, Childs AJ, Adams IR, Anderson RA. Dazl determines primordial follicle formation through the translational regulation of Tex14. FASEB J 2019; 33:14221-14233. [PMID: 31659914 DOI: 10.1096/fj.201901247r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Deleted in azoospermia-like (DAZL) is a germ cell RNA-binding protein that is essential for entry and progression through meiosis. The phenotype of the Dazl knockout mouse has extensive germ cell loss because of incomplete meiosis. We have created a Dazl hypomorph model using short interfering RNA knockdown in mouse fetal ovary cultures, allowing investigation of Dazl function in germ cell maturation. Dazl hypomorph ovaries had a phenotype of impaired germ cell nest breakdown with a 66% reduction in total follicle number and an increase in the proportion of primordial follicles (PMFs), with smaller oocytes within these follicles. There was no significant early germ cell loss or meiotic delay. Immunostaining of intercellular bridge component testis-expressed protein (Tex)14 showed ∼59% reduction in foci number and size, without any change in Tex14 mRNA levels. TEX14 expression was also confirmed in the human fetal ovary across gestation. Using 3'UTR-luciferase reporter assays, translational regulation of TEX14 was demonstrated to be DAZL-dependant. Dazl is therefore essential for normal intercellular bridges within germ cell nests and their timely breakdown, with a major impact on subsequent assembly of PMFs.-Rosario, R., Crichton, J. H., Stewart, H. L., Childs, A. J., Adams, I. R., Anderson, R. A. Dazl determines primordial follicle formation through the translational regulation of Tex14.
Collapse
Affiliation(s)
- Roseanne Rosario
- Medical Research Council (MRC) Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - James H Crichton
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Hazel L Stewart
- Medical Research Council (MRC) Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Andrew J Childs
- Department of Surgery and Cancer, Institute of Reproductive and Developmental Biology, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ian R Adams
- Medical Research Council (MRC) Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, Western General Hospital, Edinburgh, United Kingdom
| | - Richard A Anderson
- Medical Research Council (MRC) Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
81
|
Rossitto M, Ollivier M, Déjardin S, Pruvost A, Brun C, Marchive C, Nguyen AL, Ghettas A, Keime C, de Massy B, Poulat F, Philibert P, Boizet-Bonhoure B. In utero exposure to acetaminophen and ibuprofen leads to intergenerational accelerated reproductive aging in female mice. Commun Biol 2019; 2:310. [PMID: 31428698 PMCID: PMC6692356 DOI: 10.1038/s42003-019-0552-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/16/2019] [Indexed: 02/06/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) and analgesic drugs, such as acetaminophen (APAP), are frequently taken during pregnancy, even in combination. However, they can favour genital malformations in newborn boys and reproductive disorders in adults. Conversely, the consequences on postnatal ovarian development and female reproductive health after in utero exposure are unknown. Here, we found that in mice, in utero exposure to therapeutic doses of the APAP-ibuprofen combination during sex determination led to delayed meiosis entry and progression in female F1 embryonic germ cells. Consequently, follicular activation was reduced in postnatal ovaries through the AKT/FOXO3 pathway, leading in F2 animals to subfertility, accelerated ovarian aging with abnormal corpus luteum persistence, due to decreased apoptosis and increased AKT-mediated luteal cell survival. Our study suggests that administration of these drugs during the critical period of sex determination could lead in humans to adverse effects that might be passed to the offspring.
Collapse
Affiliation(s)
- Moïra Rossitto
- Development and Pathology of the Gonad, IGH, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Margot Ollivier
- Development and Pathology of the Gonad, IGH, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
- Service de Chirurgie et Urologie Pédiatrique, Hôpital Lapeyronie CHU Montpellier, Centre de Référence Maladies Rares Développement Génital, Montpellier, France
| | - Stéphanie Déjardin
- Development and Pathology of the Gonad, IGH, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Alain Pruvost
- Service de Pharmacologie et d’Immunoanalyse (SPI), plateforme SMArt-MS, CEA, INRA, Université Paris-Saclay, Gif sur Yvette, France
| | - Christine Brun
- Meiosis and Recombination, IGH, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Candice Marchive
- Development and Pathology of the Gonad, IGH, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Anvi Laetitia Nguyen
- Service de Pharmacologie et d’Immunoanalyse (SPI), plateforme SMArt-MS, CEA, INRA, Université Paris-Saclay, Gif sur Yvette, France
| | - Aurélie Ghettas
- Service de Pharmacologie et d’Immunoanalyse (SPI), plateforme SMArt-MS, CEA, INRA, Université Paris-Saclay, Gif sur Yvette, France
| | - Céline Keime
- IGBMC, Centre National de la Recherche Scientifique, Université de Strasbourg/INSERM, Illkirch, France
| | - Bernard de Massy
- Meiosis and Recombination, IGH, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Francis Poulat
- Development and Pathology of the Gonad, IGH, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| | - Pascal Philibert
- Development and Pathology of the Gonad, IGH, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
- Département de Biochimie et Hormonologie, Hôpital Lapeyronie, CHU de Montpellier, Montpellier, France
| | - Brigitte Boizet-Bonhoure
- Development and Pathology of the Gonad, IGH, Centre National de la Recherche Scientifique, Université de Montpellier, Montpellier, France
| |
Collapse
|
82
|
Law NC, Oatley MJ, Oatley JM. Developmental kinetics and transcriptome dynamics of stem cell specification in the spermatogenic lineage. Nat Commun 2019; 10:2787. [PMID: 31243281 PMCID: PMC6594958 DOI: 10.1038/s41467-019-10596-0] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 05/17/2019] [Indexed: 12/15/2022] Open
Abstract
Continuity, robustness, and regeneration of cell lineages relies on stem cell pools that are established during development. For the mammalian spermatogenic lineage, a foundational spermatogonial stem cell (SSC) pool arises from prospermatogonial precursors during neonatal life via mechanisms that remain undefined. Here, we mapped the kinetics of this process in vivo using a multi-transgenic reporter mouse model, in silico with single-cell RNA sequencing, and functionally with transplantation analyses to define the SSC trajectory from prospermatogonia. Outcomes revealed that a heterogeneous prospermatogonial population undergoes dynamic changes during late fetal and neonatal development. Differential transcriptome profiles predicted divergent developmental trajectories from fetal prospermatogonia to descendant postnatal spermatogonia. Furthermore, transplantation analyses demonstrated that a defined subset of fetal prospermatogonia is fated to function as SSCs. Collectively, these findings suggest that SSC fate is preprogrammed within a subset of fetal prospermatogonia prior to building of the foundational pool during early neonatal development. In neonatal testes, prospermatogonia generate both spermatogonia for the first wave of spermatogenesis and spermatogonial stem cells (SSCs) for maintenance of spermatogenesis in males. Here the authors characterize the development of mouse SSCs from prospermatogonia using single-cell RNA-seq and transplantation assays.
Collapse
Affiliation(s)
- Nathan C Law
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA
| | - Melissa J Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA
| | - Jon M Oatley
- Center for Reproductive Biology, School of Molecular Biosciences, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA.
| |
Collapse
|
83
|
Abstract
Primordial germ cells (PGCs) must complete a complex and dynamic developmental program during embryogenesis to establish the germline. This process is highly conserved and involves a diverse array of tasks required of PGCs, including migration, survival, sex differentiation, and extensive epigenetic reprogramming. A common theme across many organisms is that PGC success is heterogeneous: only a portion of all PGCs complete all these steps while many other PGCs are eliminated from further germline contribution. The differences that distinguish successful PGCs as a population are not well understood. Here, we examine variation that exists in PGCs as they navigate the many stages of this developmental journey. We explore potential sources of PGC heterogeneity and their potential implications in affecting germ cell behaviors. Lastly, we discuss the potential for PGC development to function as a multistage selection process that assesses heterogeneity in PGCs to refine germline quality.
Collapse
Affiliation(s)
- Daniel H Nguyen
- Department of Obstetrics, Gynecology and Reproductive Science, Center for Reproductive Sciences, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, United States
| | - Rebecca G Jaszczak
- Department of Obstetrics, Gynecology and Reproductive Science, Center for Reproductive Sciences, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, United States
| | - Diana J Laird
- Department of Obstetrics, Gynecology and Reproductive Science, Center for Reproductive Sciences, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, United States.
| |
Collapse
|
84
|
Wen J, Yan H, He M, Zhang T, Mu X, Wang H, Zhang H, Xia G, Wang C. GSK-3β protects fetal oocytes from premature death via modulating TAp63 expression in mice. BMC Biol 2019; 17:23. [PMID: 30866939 PMCID: PMC6417224 DOI: 10.1186/s12915-019-0641-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/26/2019] [Indexed: 01/24/2023] Open
Abstract
Background Female mammals have a limited reproductive lifespan determined by the size of the primordial follicle pool established perinatally. Over two thirds of fetal oocytes are abolished via programmed cell death during early folliculogenesis. However, the underlying mechanisms governing fetal oocyte attrition remain largely elusive. Results Here, we demonstrate that glycogen synthase kinase-3 beta (GSK-3β) is indispensable for fetal oocyte maintenance during meiotic prophase I in mice. In vitro inhibition of GSK-3β activity or in vivo conditional deletion of Gsk-3β in the germline led to a dramatic loss of fetal oocytes via apoptosis, which subsequently resulted in a reduced capacity of the primordial follicle pool. Inhibition of GSK-3β also impeded meiotic progression in fetal oocytes and led to a deficiency in DNA double-strand break (DSB) repair associated with premature upregulation of Tap63, the major genome guardian of the female germline, following GSK-3β inhibition in fetal ovaries. Mechanistically, we demonstrated that aberrant nuclear translocation of β-catenin was responsible for the abnormal expression of TAp63 and global fetal oocyte attrition following GSK-3β inhibition. Conclusions In summary, GSK-3β was essential for sustaining fetal oocyte survival and folliculogenesis via fine-tuning the cytoplasmic-nuclear translocation of β-catenin, which in turn modulates timely TAp63 expression during meiotic prophase I in mice. Our study provides a perspective on the physiological regulatory role of DNA damage checkpoint signaling in fetal oocyte guardianship and female fertility. Electronic supplementary material The online version of this article (10.1186/s12915-019-0641-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jia Wen
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Hao Yan
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Meina He
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Tuo Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Xinyi Mu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Medical College of Xiamen University, Xiamen, 361005, Fujian, China
| | - Hua Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.,Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, 539 W Helanshan Road, Xixia District, Yinchuan, 750021, Ningxia, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China. .,Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, 539 W Helanshan Road, Xixia District, Yinchuan, 750021, Ningxia, China.
| |
Collapse
|
85
|
Piprek RP, Kloc M, Kubiak JZ. Matrix metalloproteinase-dependent regulation of extracellular matrix shapes the structure of sexually differentiating mouse gonads. Differentiation 2019; 106:23-34. [PMID: 30852470 DOI: 10.1016/j.diff.2019.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 12/24/2018] [Accepted: 01/18/2019] [Indexed: 11/19/2022]
Abstract
The extracellular matrix (ECM) proteins play an important role in the establishment of the sex-dependent structure of developing gonads. The matrix metalloproteinases (MMPs) are the major players in the regulation of ECM. Our hypothesis was that the MMPs-dependent regulation of EMC is crucial for the establishment of the correct, either testis or ovary, structure of developing gonad. We cultured developing mouse gonads in vitro in the presence of the MMPs inhibitors (α-2-macroglobulin, leupeptin, phosphoramidon) or the MMPs activator, APMA (4-aminophenylmercuric acetate). These inhibitors and activator inhibit/activate, to a different degree, matrix metalloproteinases, but the exact mechanism of inhibition/activation remains unknown. We found that the MMP inhibitors increased accumulation of ECM in the developing gonads. The α-2-macroglobulin had the weakest, and the phosphoramidon the strongest effect on the ECM and the structure of the gonads. The α-2-macroglobulin caused a slight increase of ECM and did not disrupt the gonad structure. Leupeptin led to the strong accumulation of ECM, resulted in the formation of the structures resembling testis cords in both testes and ovaries, and caused increase of apoptosis and complete loss of germ cells. Phosphoramidon caused the strongest accumulation of ECM, which separated individual cells and completely prevented intercellular adhesion both in the testes and in the ovaries. As a result of aberrant morphology, the sex of the phosphoramidon-treated gonads was morphologically unrecognizable. The APMA - the activator of MMP caused ECM loss, which led to the loss of cell adhesion, cell dispersion and an aberrant morphology of the gonads. These results indicate that the ECM accumulation is MMPs-dependent and that the correct amount and distribution of ECM during gonad development plays a key role in the formation of the gonad structure.
Collapse
Affiliation(s)
- Rafal P Piprek
- Department of Comparative Anatomy, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland.
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA; Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA; University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Jacek Z Kubiak
- Univ Rennes, CNRS, Institute of Genetics and Development of Rennes, UMR 6290, Cell Cycle Group, Faculty of Medicine, F-35000, Rennes, France; Laboratory of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), Warsaw, Poland
| |
Collapse
|
86
|
Larose H, Shami AN, Abbott H, Manske G, Lei L, Hammoud SS. Gametogenesis: A journey from inception to conception. Curr Top Dev Biol 2019; 132:257-310. [PMID: 30797511 PMCID: PMC7133493 DOI: 10.1016/bs.ctdb.2018.12.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Gametogenesis, the process of forming mature germ cells, is an integral part of both an individual's and a species' health and well-being. This chapter focuses on critical male and female genetic and epigenetic processes underlying normal gamete formation through their differentiation to fertilization. Finally, we explore how knowledge gained from this field has contributed to progress in areas with great clinical promise, such as in vitro gametogenesis.
Collapse
Affiliation(s)
- Hailey Larose
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Haley Abbott
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Gabriel Manske
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Lei Lei
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI, United States.
| | - Saher Sue Hammoud
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, United States; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
87
|
Jamieson-Lucy A, Mullins MC. The vertebrate Balbiani body, germ plasm, and oocyte polarity. Curr Top Dev Biol 2019; 135:1-34. [DOI: 10.1016/bs.ctdb.2019.04.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
88
|
Yoshida S. Heterogeneous, dynamic, and stochastic nature of mammalian spermatogenic stem cells. Curr Top Dev Biol 2019; 135:245-285. [DOI: 10.1016/bs.ctdb.2019.04.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
89
|
Świątek P, Urbisz AZ. Architecture and Life History of Female Germ-Line Cysts in Clitellate Annelids. Results Probl Cell Differ 2019; 68:515-551. [PMID: 31598870 DOI: 10.1007/978-3-030-23459-1_21] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Animal female and male germ-line cells often form syncytial units termed cysts, clusters, or clones. Within these cysts, the cells remain interconnected by specific cell junctions known as intercellular bridges or ring canals, which enable cytoplasm to be shared and macromolecules and organelles to be exchanged between cells. Numerous analyses have shown that the spatial organization of cysts and their functioning may differ between the sexes and taxa. The vast majority of our knowledge about the formation and functioning of germ-line cysts comes from studies of model species (mainly Drosophila melanogaster); the other systems of the cyst organization and functioning are much less known and are sometimes overlooked. Here, we present the current state of the knowledge of female germ-line cysts in clitellate annelids (Clitellata), which is a monophyletic taxon of segmented worms (Annelida). The organization of germ-line cysts in clitellates differs markedly from that of the fruit fly and vertebrates. In Clitellata, germ cells are not directly connected one to another, but, as a rule, each cell has one ring canal that connects it to an anuclear central cytoplasmic core, a cytophore. Thus, this pattern of cell distribution is similar to the germ-line cysts of Caenorhabditis elegans. The last decade of studies has revealed that although clitellate female germ-line cysts have a strong morphological plasticity, e.g., cysts may contain from 16 to as many as 2500 cells, the oogenesis always shows a meroistic mode, i.e., the interconnected cells take on different fates; a few (sometimes only one) become oocytes, whereas the rest play the role of supporting (nurse) cells and do not continue oogenesis.This is the first comprehensive summary of the current knowledge on the organization and functioning of female germ-line cysts in clitellate annelids.
Collapse
Affiliation(s)
- Piotr Świątek
- Faculty of Biology and Environmental Protection, Department of Animal Histology and Embryology, University of Silesia in Katowice, Katowice, Poland.
| | - Anna Z Urbisz
- Faculty of Biology and Environmental Protection, Department of Animal Histology and Embryology, University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
90
|
The extraordinary biology and development of marsupial frogs (Hemiphractidae) in comparison with fish, mammals, birds, amphibians and other animals. Mech Dev 2018; 154:2-11. [DOI: 10.1016/j.mod.2017.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/23/2017] [Accepted: 12/28/2017] [Indexed: 11/20/2022]
|
91
|
Palozzi JM, Jeedigunta SP, Hurd TR. Mitochondrial DNA Purifying Selection in Mammals and Invertebrates. J Mol Biol 2018; 430:4834-4848. [DOI: 10.1016/j.jmb.2018.10.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/20/2018] [Accepted: 10/25/2018] [Indexed: 01/19/2023]
|
92
|
Rotgers E, Jørgensen A, Yao HHC. At the Crossroads of Fate-Somatic Cell Lineage Specification in the Fetal Gonad. Endocr Rev 2018; 39:739-759. [PMID: 29771299 PMCID: PMC6173476 DOI: 10.1210/er.2018-00010] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/09/2018] [Indexed: 01/07/2023]
Abstract
The reproductive endocrine systems are vastly different between males and females. This sexual dimorphism of the endocrine milieu originates from sex-specific differentiation of the somatic cells in the gonads during fetal life. Most gonadal somatic cells arise from the adrenogonadal primordium. After separation of the adrenal and gonadal primordia, the gonadal somatic cells initiate sex-specific differentiation during gonadal sex determination with the specification of the supporting cell lineages: Sertoli cells in the testis vs granulosa cells in the ovary. The supporting cell lineages then facilitate the differentiation of the steroidogenic cell lineages, Leydig cells in the testis and theca cells in the ovary. Proper differentiation of these cell types defines the somatic cell environment that is essential for germ cell development, hormone production, and establishment of the reproductive tracts. Impairment of lineage specification and function of gonadal somatic cells can lead to disorders of sexual development (DSDs) in humans. Human DSDs and processes for gonadal development have been successfully modeled using genetically modified mouse models. In this review, we focus on the fate decision processes from the initial stage of formation of the adrenogonadal primordium in the embryo to the maintenance of the somatic cell identities in the gonads when they become fully differentiated in adulthood.
Collapse
Affiliation(s)
- Emmi Rotgers
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, North Carolina
| | - Anne Jørgensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,International Research and Research Training Center in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen, Denmark
| | - Humphrey Hung-Chang Yao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Durham, North Carolina
| |
Collapse
|
93
|
Zhihan T, Xinyi M, Qingying L, Rufei G, Yan Z, Xuemei C, Yanqing G, Yingxiong W, Junlin H. Autophagy participates in cyst breakdown and primordial folliculogenesis by reducing reactive oxygen species levels in perinatal mouse ovaries. J Cell Physiol 2018; 234:6125-6135. [DOI: 10.1002/jcp.27367] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 08/16/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Tu Zhihan
- Laboratory of Reproductive Biology, College of Public Health and Administration, Chongqing Medical University Chongqing China
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University Chongqing China
| | - Mu Xinyi
- Laboratory of Reproductive Biology, College of Public Health and Administration, Chongqing Medical University Chongqing China
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University Chongqing China
- Department of Histology and Embryology College of Basic Medicine, Chongqing Medical University Chongqing China
| | - Li Qingying
- Laboratory of Reproductive Biology, College of Public Health and Administration, Chongqing Medical University Chongqing China
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University Chongqing China
| | - Gao Rufei
- Laboratory of Reproductive Biology, College of Public Health and Administration, Chongqing Medical University Chongqing China
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University Chongqing China
| | - Zhang Yan
- Laboratory of Reproductive Biology, College of Public Health and Administration, Chongqing Medical University Chongqing China
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University Chongqing China
| | - Chen Xuemei
- Laboratory of Reproductive Biology, College of Public Health and Administration, Chongqing Medical University Chongqing China
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University Chongqing China
| | - Geng Yanqing
- Laboratory of Reproductive Biology, College of Public Health and Administration, Chongqing Medical University Chongqing China
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University Chongqing China
| | - Wang Yingxiong
- Laboratory of Reproductive Biology, College of Public Health and Administration, Chongqing Medical University Chongqing China
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University Chongqing China
| | - He Junlin
- Laboratory of Reproductive Biology, College of Public Health and Administration, Chongqing Medical University Chongqing China
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University Chongqing China
| |
Collapse
|
94
|
Wear HM, Eriksson A, Yao HHC, Watanabe KH. Cell-based computational model of early ovarian development in mice. Biol Reprod 2018; 97:365-377. [PMID: 29088396 DOI: 10.1093/biolre/iox089] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/09/2017] [Indexed: 11/13/2022] Open
Abstract
Despite its importance to reproduction, certain mechanisms of early ovarian development remain a mystery. To improve our understanding, we constructed the first cell-based computational model of ovarian development in mice that is divided into two phases: Phase I spans embryonic day 5.5 (E5.5) to E12.5; and Phase II spans E12.5 to postnatal day 2. We used the model to investigate four mechanisms: in Phase I, (i) whether primordial germ cells (PGCs) undergo mitosis during migration; and (ii) if the mechanism for secretion of KIT ligand from the hindgut resembles inductive cell-cell signaling or is secreted in a static manner; and in Phase II, (iii) that changes in cellular adhesion produce germ cell nest breakdown; and (iv) whether localization of primordial follicles in the cortex of the ovary is due to proliferation of granulosa cells. We found that the combination of the first three hypotheses produced results that aligned with experimental images and PGC abundance data. Results from the fourth hypothesis did not match experimental images, which suggests that more detailed processes are involved in follicle localization. Phase I and Phase II of the model reproduce experimentally observed cell counts and morphology well. A sensitivity analysis identified contact energies, mitotic rates, KIT chemotaxis strength, and diffusion rate in Phase I and oocyte death rate in Phase II as parameters with the greatest impact on model predictions. The results demonstrate that the computational model can be used to understand unknown mechanisms, generate new hypotheses, and serve as an educational tool.
Collapse
Affiliation(s)
- Hannah M Wear
- Institute of Environmental Health, Oregon Health & Science University, Portland, OR, USA
| | - Annika Eriksson
- Division of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University Portland, OR, USA
| | - Humphrey Hung-Chang Yao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Karen H Watanabe
- Institute of Environmental Health, Oregon Health & Science University, Portland, OR, USA.,School of Public Health, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
95
|
Hunter N. Oocyte Quality Control: Causes, Mechanisms, and Consequences. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2018; 82:235-247. [PMID: 29743337 DOI: 10.1101/sqb.2017.82.035394] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Oocyte quality and number are key determinants of reproductive life span and success. These variables are shaped in part by the elimination of oocytes that experience problems during the early stages of meiosis. Meiotic prophase-I marks an extended period of genome vulnerability in which epigenetic reprogramming unleashes retroelements and hundreds of DNA double-strand breaks (DSBs) are inflicted to initiate the programmed recombination required for accurate chromosome segregation at the first meiotic division. Expression of LINE-1 retroelements perturbs several aspects of meiotic prophase and is associated with oocyte death during the early stages of meiotic prophase I. Defects in chromosome synapsis and recombination also trigger oocyte loss, but typically at a later stage, as cells transition into quiescence and form primordial follicles. Interrelated pathways that signal defects in DSB repair and chromosome synapsis mediate this late oocyte attrition. Here, I review our current understanding of early and late oocyte attrition based on studies in mouse and describe how these processes appear to be both distinct and overlapping and how they help balance the quality and size of oocyte reserves to maximize fecundity.
Collapse
Affiliation(s)
- Neil Hunter
- Howard Hughes Medical Institute, University of California, Davis, Davis, California 95616.,Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, California 95616.,Department of Molecular and Cellular Biology, University of California, Davis, Davis, California 95616.,Department of Cell Biology and Human Anatomy, University of California, Davis, Davis, California 95616
| |
Collapse
|
96
|
Arendrup FS, Mazaud-Guittot S, Jégou B, Kristensen DM. EDC IMPACT: Is exposure during pregnancy to acetaminophen/paracetamol disrupting female reproductive development? Endocr Connect 2018; 7:149-158. [PMID: 29305399 PMCID: PMC5776669 DOI: 10.1530/ec-17-0298] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 11/20/2017] [Indexed: 12/19/2022]
Abstract
Concern has been raised over chemical-induced disruption of ovary development during fetal life resulting in long-lasting consequences only manifesting themselves much later during adulthood. A growing body of evidence suggests that prenatal exposure to the mild analgesic acetaminophen/paracetamol can cause such a scenario. Therefore, in this review, we discuss three recent reports that collectively indicate that prenatal exposure in a period of 13.5 days post coitum in both rats and mouse can result in reduced female reproductive health. The combined data show that the exposure results in the reduction of primordial follicles, irregular menstrual cycle, premature absence of corpus luteum, as well as reduced fertility, resembling premature ovarian insufficiency syndrome in humans that is linked to premature menopause. This could especially affect the Western parts of the world, where the age for childbirth is continuously being increased and acetaminophen is recommended during pregnancy for pain and fever. We therefore highlight an urgent need for more studies to verify these data including both experimental and epidemiological approaches.
Collapse
Affiliation(s)
| | - Severine Mazaud-Guittot
- Inserm (Institut National de la Santé et de la Recherche Médicale)Irset - Inserm, UMR 1085, Rennes, France
| | - Bernard Jégou
- Inserm (Institut National de la Santé et de la Recherche Médicale)Irset - Inserm, UMR 1085, Rennes, France
- EHESP-School of Public HealthRennes, France
| | - David Møbjerg Kristensen
- Department of NeurologyDanish Headache Center, Rigshospitalet, University of Copenhagen, Denmark
- Inserm (Institut National de la Santé et de la Recherche Médicale)Irset - Inserm, UMR 1085, Rennes, France
| |
Collapse
|
97
|
Lu K, Jensen L, Lei L, Yamashita YM. Stay Connected: A Germ Cell Strategy. Trends Genet 2017; 33:971-978. [PMID: 28947158 DOI: 10.1016/j.tig.2017.09.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/24/2017] [Accepted: 09/05/2017] [Indexed: 01/20/2023]
Abstract
Germ cells develop as a cyst of interconnected sibling cells in a broad range of organisms in both sexes. A well-established function of intercellular connectivity is to transport cytoplasmic materials from 'nurse' cells to oocytes, a critical process for developing functional oocytes in ovaries of many species. However, there are situations where connectivity exists without a nursing mechanism, and the biological meaning of such connectivity remains obscure. In this review, we summarize current knowledge on the formation of intercellular connectivity, and discuss its meaning by visiting multiple examples of germ cell connectivity observed in evolutionarily distant species.
Collapse
Affiliation(s)
- Kevin Lu
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA; Medical Scientist Training Program, University of Michigan, Ann Arbor, MI 48109, USA; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lindy Jensen
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lei Lei
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yukiko M Yamashita
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI 48109, USA; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA; Howard Hughes Medical Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
98
|
McCaig CM, Lin X, Farrell M, Rehain-Bell K, Shakes DC. Germ cell cysts and simultaneous sperm and oocyte production in a hermaphroditic nematode. Dev Biol 2017; 430:362-373. [PMID: 28844904 DOI: 10.1016/j.ydbio.2017.08.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 07/05/2017] [Accepted: 08/08/2017] [Indexed: 11/29/2022]
Abstract
Studies of gamete development in the self-fertile hermaphrodites of Caenorhabditis elegans have significantly contributed to our understanding of fundamental developmental mechanisms. However, evolutionary transitions from outcrossing males and females to self-fertile hermaphrodites have convergently evolved within multiple nematode sub-lineages, and whether the C. elegans pattern of self-fertile hermaphroditism and gamete development is representative remains largely unexplored. Here we describe a pattern of sperm production in the trioecious (male/female/hermaphrodite) nematode Rhabditis sp. SB347 (recently named Auanema rhodensis) that differs from C. elegans in two striking ways. First, while C. elegans hermaphrodites make a one-time switch from sperm to oocyte production, R. sp. SB347 hermaphrodites continuously produce both sperm and oocytes. Secondly, while C. elegans germ cell proliferation is limited to germline stem cells (GSCs), sperm production in R. sp. SB347 includes an additional population of mitotically dividing cells that are a developmental intermediate between GSCs and fully differentiated spermatocytes. These cells are present in males and hermaphrodites but not females, and exhibit key characteristics of spermatogonia - the mitotic progenitors of spermatocytes in flies and vertebrates. Specifically, they exist outside the stem cell niche, increase germ cell numbers by transit-amplifying divisions, and synchronously proliferate within germ cell cysts. We also discovered spermatogonia in other trioecious Rhabditis species, but not in the male/female species Rhabditis axei or the more distant hermaphroditic Oscheius tipulae. The discovery of simultaneous hermaphroditism and spermatogonia in a lab-cultivatable nematode suggests R. sp. SB347 as a richly informative species for comparative studies of gametogenesis.
Collapse
Affiliation(s)
- Caitlin M McCaig
- Department of Biology, College of William and Mary, Williamsburg, VA 23187, USA
| | - Xiaoxue Lin
- Department of Biology, College of William and Mary, Williamsburg, VA 23187, USA
| | - Maureen Farrell
- Department of Biology, College of William and Mary, Williamsburg, VA 23187, USA
| | - Kathryn Rehain-Bell
- Department of Biology, College of William and Mary, Williamsburg, VA 23187, USA
| | - Diane C Shakes
- Department of Biology, College of William and Mary, Williamsburg, VA 23187, USA.
| |
Collapse
|
99
|
A pilgrim's progress: Seeking meaning in primordial germ cell migration. Stem Cell Res 2017; 24:181-187. [PMID: 28754603 DOI: 10.1016/j.scr.2017.07.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 06/08/2017] [Accepted: 07/15/2017] [Indexed: 01/08/2023] Open
Abstract
Comparative studies of primordial germ cell (PGC) development across organisms in many phyla reveal surprising diversity in the route of migration, timing and underlying molecular mechanisms, suggesting that the process of migration itself is conserved. However, beyond the perfunctory transport of cellular precursors to their later arising home of the gonads, does PGC migration serve a function? Here we propose that the process of migration plays an additional role in quality control, by eliminating PGCs incapable of completing migration as well as through mechanisms that favor PGCs capable of responding appropriately to migration cues. Focusing on PGCs in mice, we explore evidence for a selective capacity of migration, considering the tandem regulation of proliferation and migration, cell-intrinsic and extrinsic control, the potential for tumors derived from failed PGC migrants, the potential mechanisms by which migratory PGCs vary in their cellular behaviors, and corresponding effects on development. We discuss the implications of a selective role of PGC migration for in vitro gametogenesis.
Collapse
|
100
|
Wang C, Zhou B, Xia G. Mechanisms controlling germline cyst breakdown and primordial follicle formation. Cell Mol Life Sci 2017; 74:2547-2566. [PMID: 28197668 PMCID: PMC11107689 DOI: 10.1007/s00018-017-2480-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 12/11/2022]
Abstract
In fetal females, oogonia proliferate immediately after sex determination. The progress of mitosis in oogonia proceeds so rapidly that the incompletely divided cytoplasm of the sister cells forms cysts. The oogonia will then initiate meiosis and arrest at the diplotene stage of meiosis I, becoming oocytes. Within each germline cyst, oocytes with Balbiani bodies will survive after cyst breakdown (CBD). After CBD, each oocyte is enclosed by pre-granulosa cells to form a primordial follicle (PF). Notably, the PF pool formed perinatally will be the sole lifelong oocyte source of a female. Thus, elucidating the mechanisms of CBD and PF formation is not only meaningful for solving mysteries related to ovarian development but also contributes to the preservation of reproduction. However, the mechanisms that regulate these phenomena are largely unknown. This review summarizes the progress of cellular and molecular research on these processes in mice and humans.
Collapse
Affiliation(s)
- Chao Wang
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Bo Zhou
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Guoliang Xia
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|