201
|
Sun YC, Sun XF, Dyce PW, Shen W, Chen H. The role of germ cell loss during primordial follicle assembly: a review of current advances. Int J Biol Sci 2017; 13:449-457. [PMID: 28529453 PMCID: PMC5436565 DOI: 10.7150/ijbs.18836] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/25/2017] [Indexed: 12/27/2022] Open
Abstract
In most female mammals, early germline development begins with the appearance of primordial germ cells (PGCs), and develops to form mature oocytes following several vital processes. It remains well accepted that significant germ cell apoptosis and oocyte loss takes place around the time of birth. The transition of the ovarian environment from fetal to neonatal, coincides with the loss of germ cells and the timing of follicle formation. All told it is common to lose approximately two thirds of germ cells during this transition period. The current consensus is that germ cell loss can be attributed, at least in part, to programmed cell death (PCD). Recently, autophagy has been implicated as playing a part in germ cell loss during the time of parturition. In this review, we discuss the major opinions and mechanisms of mammalian ovarian PCD during the process of germ cell loss. We also pay close attention to the function of autophagy in germ cell loss, and speculate that autophagy may also serve as a critical and necessary process during the establishment of primordial follicle pool.
Collapse
Affiliation(s)
- Yuan-Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling Shaanxi 712100, China
| | - Xiao-Feng Sun
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Paul W Dyce
- Department of Animal Sciences, Auburn University, Auburn, AL 36849, USA
| | - Wei Shen
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China
| | - Hong Chen
- College of Animal Science and Technology, Northwest A&F University, Shaanxi Key Laboratory of Molecular Biology for Agriculture, Yangling Shaanxi 712100, China
| |
Collapse
|
202
|
Vanorny DA, Mayo KE. The role of Notch signaling in the mammalian ovary. Reproduction 2017; 153:R187-R204. [PMID: 28283672 DOI: 10.1530/rep-16-0689] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/03/2017] [Accepted: 03/09/2017] [Indexed: 12/21/2022]
Abstract
The Notch pathway is a contact-dependent, or juxtacrine, signaling system that is conserved in metazoan organisms and is important in many developmental processes. Recent investigations have demonstrated that the Notch pathway is active in both the embryonic and postnatal ovary and plays important roles in events including follicle assembly and growth, meiotic maturation, ovarian vasculogenesis and steroid hormone production. In mice, disruption of the Notch pathway results in ovarian pathologies affecting meiotic spindle assembly, follicle histogenesis, granulosa cell proliferation and survival, corpora luteal function and ovarian neovascularization. These aberrations result in abnormal folliculogenesis and reduced fertility. The knowledge of the cellular interactions facilitated by the Notch pathway is an important area for continuing research, and future studies are expected to enhance our understanding of ovarian function and provide critical insights for improving reproductive health. This review focuses on the expression of Notch pathway components in the ovary, and on the multiple functions of Notch signaling in follicle assembly, maturation and development. We focus on the mouse, where genetic investigations are possible, and relate this information to the human ovary.
Collapse
Affiliation(s)
- Dallas A Vanorny
- Department of Molecular Biosciences and Center for Reproductive ScienceNorthwestern University, Evanston, Illinois, USA
| | - Kelly E Mayo
- Department of Molecular Biosciences and Center for Reproductive ScienceNorthwestern University, Evanston, Illinois, USA
| |
Collapse
|
203
|
Developmental effects of imatinib mesylate on follicle assembly and early activation of primordial follicle pool in postnatal rat ovary. Reprod Biol 2017; 17:25-33. [DOI: 10.1016/j.repbio.2016.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 12/26/2022]
|
204
|
Moriwaki M, Moore B, Mosbruger T, Neklason DW, Yandell M, Jorde LB, Welt CK. POLR2C Mutations Are Associated With Primary Ovarian Insufficiency in Women. J Endocr Soc 2017; 1:162-173. [PMID: 29367954 PMCID: PMC5650244 DOI: 10.1210/js.2016-1014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/01/2017] [Indexed: 01/11/2023] Open
Abstract
CONTEXT Primary ovarian insufficiency (POI) results from a premature loss of oocytes, causing infertility and early menopause. The etiology of POI remains unknown in a majority of cases. OBJECTIVE To identify candidate genes in families affected by POI. DESIGN This was a family-based genetic study. SETTING The study was performed at two academic institutions. PATIENTS AND OTHER PARTICIPANTS A family with four generations of women affected by POI (n = 5). Four of these women, three with an associated autoimmune diagnosis, were studied. The controls (n = 387) were recruited for health in old age. INTERVENTION Whole-genome sequencing was performed. MAIN OUTCOME MEASURE Candidate genes were identified by comparing gene mutations in three family members and 387 control subjects analyzed simultaneously using the pedigree Variant Annotation, Analysis and Search Tool. Data were also compared with that in publicly available databases. RESULTS We identified a heterozygous nonsense mutation in a subunit of RNA polymerase II (POLR2C) that synthesizes messenger RNA. A rare sequence variant in POLR2C was also identified in one of 96 women with sporadic POI. POLR2C expression was decreased in the proband compared with women with POI from another cause. Knockdown in an embryonic carcinoma cell line resulted in decreased protein production and impaired cell proliferation. CONCLUSIONS These data support a role for RNA polymerase II mutations as candidates in the etiology of POI. The current data also support results from genome-wide association studies that hypothesize a role for RNA polymerase II subunits in age at menopause in the population.
Collapse
Affiliation(s)
| | - Barry Moore
- UStar Center for Genetic Discovery, Department of Human Genetics
| | | | - Deborah W. Neklason
- Division of Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah 84112
| | - Mark Yandell
- UStar Center for Genetic Discovery, Department of Human Genetics
| | - Lynn B. Jorde
- UStar Center for Genetic Discovery, Department of Human Genetics
| | | |
Collapse
|
205
|
Differentiation of Mouse Primordial Germ Cells into Functional Oocytes In Vitro. Ann Biomed Eng 2017; 45:1608-1619. [PMID: 28243826 PMCID: PMC5489615 DOI: 10.1007/s10439-017-1815-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/15/2017] [Indexed: 01/01/2023]
Abstract
Various complex molecular events in oogenesis cannot be observed in vivo. As a bioengineering technique for female reproductive tissues, in vitro culture systems for female germ cells have been used to analyze oogenesis and preserve germ cells for over 20 years. Recently, we have established a new methodological approach for the culture of primordial germ cells (PGCs) and successfully obtained offspring. Our PGC culture system will be useful to clarify unresolved mechanisms of fertility and sterility from the beginning of mammalian oogenesis, before meiosis. This review summarizes the history of culture methods for mammalian germ cells, our current in vitro system, and future prospects for the culture of germ cells.
Collapse
|
206
|
Wang YY, Sun YC, Sun XF, Cheng SF, Li B, Zhang XF, De Felici M, Shen W. Starvation at birth impairs germ cell cyst breakdown and increases autophagy and apoptosis in mouse oocytes. Cell Death Dis 2017; 8:e2613. [PMID: 28182014 PMCID: PMC5386484 DOI: 10.1038/cddis.2017.3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 01/02/2017] [Accepted: 01/03/2017] [Indexed: 01/12/2023]
Abstract
The female reproductive lifespan is largely determined by the size of primordial follicle pool, which is established following germ cell cyst breakdown around birth. Almost two-third of oocytes are lost during germ cell cysts breakdown, following autophagic and apoptosis mechanisms. To investigate a possible relationship between germ cell cyst breakdown and nutrition supply, we established a starvation model in mouse pups at birth and evaluated the dynamics of cyst breakdown during nutrient deprivation. Our results showed that after 36 h of starvation between 1.5 and 3 d.p.p., indicators of metabolism both at systemic and ovarian level were significantly altered and the germ cell cyst breakdown markedly decreased. We also found that markers of oxidative stress, autophagy and apoptosis were increased and higher number of oocytes in cyst showing autophagic markers and of TUNEL-positive oocytes and somatic cells were present in the ovaries of starved pups. Moreover, the proliferation of pre-granulosa cells and the expression of the oocyte-specific transcription factor Nobox were decreased in such ovaries. Finally, we observed that the ovaries of the starved pups could recover a normal number of follicles after about 3 weeks from re-feeding. In conclusion, these data indicate that nutrient deficiency at birth can generate a number of adaptive metabolic and oxidative responses in the ovaries causing increased apoptosis both in the somatic cells and oocyte and autophagy mainly in these latter and leading to a delay of germ cell cyst breakdown and follicle assembly.
Collapse
Affiliation(s)
- Yong-Yong Wang
- College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China.,College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Yuan-Chao Sun
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Xiao-Feng Sun
- College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China.,College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Shun-Feng Cheng
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Bo Li
- Chengguo Station of Animal Husbandry and Veterinary, Laizhou 261437, China
| | - Xi-Feng Zhang
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome 00133, Italy
| | - Wei Shen
- College of Animal Science and Technology, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao 266109, China
| |
Collapse
|
207
|
Sharma RP, Schuhmacher M, Kumar V. Review on crosstalk and common mechanisms of endocrine disruptors: Scaffolding to improve PBPK/PD model of EDC mixture. ENVIRONMENT INTERNATIONAL 2017; 99:1-14. [PMID: 27697394 DOI: 10.1016/j.envint.2016.09.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/18/2016] [Accepted: 09/19/2016] [Indexed: 06/06/2023]
Abstract
Endocrine disruptor compounds (EDCs) are environment chemicals that cause harmful effects through multiple mechanisms, interfering with hormone system resulting in alteration of homeostasis, reproduction and developmental effect. Many of these EDCs have concurrent exposure with crosstalk and common mechanisms which may lead to dynamic interactions. To carry out risk assessment of EDCs' mixture, it is important to know the detailed toxic pathway, crosstalk of receptor and other factors like critical window of exposure. In this review, we summarize the major mechanism of actions of EDCs with the different/same target organs interfering with the same/different class of hormone by altering their synthesis, metabolism, binding and cellular action. To show the impact of EDCs on life stage development, a case study on female fertility affecting germ cell is illustrated. Based on this summarized discussion, major groups of EDCs are classified based on their target organ, mode of action and potential risk. Finally, a conceptual model of pharmacodynamic interaction is proposed to integrate the crosstalk and common mechanisms that modulate estrogen into the predictive mixture dosimetry model with dynamic interaction of mixture. This review will provide new insight for EDCs' risk assessment and can be used to develop next generation PBPK/PD models for EDCs' mixture analysis.
Collapse
Affiliation(s)
- Raju Prasad Sharma
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
| | - Marta Schuhmacher
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
| | - Vikas Kumar
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain.
| |
Collapse
|
208
|
Ikami K, Nuzhat N, Lei L. Organelle transport during mouse oocyte differentiation in germline cysts. Curr Opin Cell Biol 2017; 44:14-19. [PMID: 28038435 DOI: 10.1016/j.ceb.2016.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 11/25/2016] [Accepted: 12/05/2016] [Indexed: 10/20/2022]
Abstract
During mammalian oogenesis, germ cells undergo oocyte differentiation and oocyte development to form mature oocytes that contain essential components for supporting early embryogenesis. However, only a small fraction of germ cells become mature oocytes and the mechanism of this massive germ cell loss has been unclear. Our recent studies suggested that the formation of functional oocytes and germ cell loss are interlinked by a 'nursing' process in germline cysts during oocyte differentiation in mouse fetal ovaries. 80% of the fetal germ cells sacrifice themselves by donating their cytoplasmic contents to the remaining sister germ cells that differentiate into primary oocytes with augmented developmental potential. In this review, we will summarize the process of mouse oocyte differentiation with a particular focus on organelle transport in germline cysts.
Collapse
Affiliation(s)
- Kanako Ikami
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3045, Ann Arbor, MI 48109, United States
| | - Nafisa Nuzhat
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3045, Ann Arbor, MI 48109, United States
| | - Lei Lei
- Department of Cell and Developmental Biology, University of Michigan Medical School, 109 Zina Pitcher Place, BSRB 3045, Ann Arbor, MI 48109, United States.
| |
Collapse
|
209
|
Abstract
The ovary, the female gonad, serves as the source for the germ cells as well as the major supplier of steroid sex hormones. During embryonic development, the primordial germ cells (PGCs) are specified, migrate to the site of the future gonad, and proliferate, forming structures of germ cells nests, which will eventually break down to generate the primordial follicles (PMFs). Each PMF contains an oocyte arrested at the first prophase of meiosis, surrounded by a flattened layer of somatic pre-granulosa cells. Most of the PMFs are kept dormant and only a selected population is activated to join the growing pool of follicles in a process regulated by both intra- and extra-oocyte factors. The PMFs will further develop into secondary pre-antral follicles, a stage which depends on bidirectional communication between the oocyte and the surrounding somatic cells. Many of the signaling molecules involved in this dialog belong to the transforming growth factor β (TGF-β) superfamily. As the follicle continues to develop, a cavity called antrum is formed. The resulting antral follicles relay on the pituitary gonadotropins, follicle-stimulating hormone (FSH), and luteinizing hormone (LH) for their development. Most of the follicles undergo atretic degeneration and only a subset of the antral follicles, known as the dominant follicles, will reach the preovulatory stage at each reproductive cycle, respond to LH, and subsequently ovulate, releasing a fertilizable oocyte. The remaining somatic cells in the raptured follicle will undergo terminal differentiation and form the corpus luteum, which secretes progesterone necessary to maintain pregnancy.
Collapse
|
210
|
Zhang GL, Sun XF, Feng YZ, Li B, Li YP, Yang F, Nyachoti CM, Shen W, Sun SD, Li L. Zearalenone exposure impairs ovarian primordial follicle formation via down-regulation of Lhx8 expression in vitro. Toxicol Appl Pharmacol 2017; 317:33-40. [PMID: 28089945 DOI: 10.1016/j.taap.2017.01.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/14/2016] [Accepted: 01/08/2017] [Indexed: 11/24/2022]
Abstract
Zearalenone (ZEA) is an estrogenic mycotoxin mainly produced as a secondary metabolite by numerous species of Fusarium. Previous work showed that ZEA had a negative impact on domestic animals with regard to reproduction. The adverse effects and the mechanisms of ZEA on mammalian ovarian folliculogenesis remain largely unknown, particularly its effect on primordial follicle formation. Thus, we investigated the biological effects of ZEA exposure on murine ovarian germ cell cyst breakdown and primordial follicle assembly. Our results demonstrated that newborn mouse ovaries exposed to 10 or 30μM ZEA in vitro had significantly less germ cell numbers compared to the control group. Moreover, the presence of ZEA in vitro increased the numbers of TUNEL and γH2AX positive cells within mouse ovaries and the ratio of mRNA levels of the apoptotic genes Bax/Bcl-2. Furthermore, ZEA exposure reduced the mRNA of oocyte specific genes such as LIM homeobox 8 (Lhx8), newborn ovary homeobox (Nobox), spermatogenesis and oogenesis helix-loop-helix (Sohlh2), and factor in the germline alpha (Figlα) in a dose dependent manner. Exposure to ZEA led to remarkable changes in the Lhx8 3'-UTR DNA methylation dynamics in oocytes and severely impaired folliculogenesis in ovaries after transplantation under the kidney capsules of immunodeficient mice. In conclusion, ZEA exposure impairs mouse primordial follicle formation in vitro.
Collapse
Affiliation(s)
- Guo-Liang Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiao-Feng Sun
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, Shandong 266109, China
| | - Yan-Zhong Feng
- Institute of Animal Sciences, Heilongjiang Academy of Agricultural Sciences, Harbin, Heilongjiang 150086, China
| | - Bo Li
- Chengguo Station of Animal Husbandry and Veterinary, Laizhou 261437, China
| | - Ya-Peng Li
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, Shandong 266109, China
| | - Fan Yang
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, Shandong 266109, China
| | | | - Wei Shen
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, Shandong 266109, China
| | - Shi-Duo Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Lan Li
- Institute of Reproductive Sciences, College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, Shandong 266109, China.
| |
Collapse
|
211
|
Abstract
Fully grown oocytes arrest meiosis at prophase I and deposit maternal RNAs. A subset of maternal transcripts is stored in a dormant state in the oocyte, and the timely driven translation of specific mRNAs guides meiotic progression, the oocyte-embryo transition, and early embryo development. In the absence of transcription, the regulation of gene expression in oocytes is controlled almost exclusively at the level of transcriptome and proteome stabilization and at the level of protein synthesis.This chapter focuses on the recent findings on RNA distribution related to the temporal and spatial translational control of the meiotic cycle progression in mammalian oocytes. We discuss the most relevant mechanisms involved in the organization of the oocyte's maternal transcriptome storage and localization, and the regulation of translation, in correlation with the regulation of oocyte meiotic progression.
Collapse
|
212
|
Chang HM, Qiao J, Leung PCK. Oocyte-somatic cell interactions in the human ovary-novel role of bone morphogenetic proteins and growth differentiation factors. Hum Reprod Update 2016; 23:1-18. [PMID: 27797914 PMCID: PMC5155571 DOI: 10.1093/humupd/dmw039] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/29/2016] [Accepted: 10/11/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Initially identified for their capability to induce heterotopic bone formation,
bone morphogenetic proteins (BMPs) are multifunctional growth factors that belong
to the transforming growth factor β superfamily. Using cellular and
molecular genetic approaches, recent studies have implicated intra-ovarian BMPs as
potent regulators of ovarian follicular function. The bi-directional communication
of oocytes and the surrounding somatic cells is mandatory for normal follicle
development and oocyte maturation. This review summarizes the current knowledge on
the physiological role and molecular determinants of these ovarian regulatory
factors within the human germline-somatic regulatory loop. OBJECTIVE AND RATIONALE The regulation of ovarian function remains poorly characterized in humans because,
while the fundamental process of follicular development and oocyte maturation is
highly similar across species, most information on the regulation of ovarian
function is obtained from studies using rodent models. Thus, this review focuses
on the studies that used human biological materials to gain knowledge about human
ovarian biology and disorders and to develop strategies for preventing, diagnosing
and treating these abnormalities. SEARCH METHODS Relevant English-language publications describing the roles of BMPs or growth
differentiation factors (GDFs) in human ovarian biology and phenotypes were
comprehensively searched using PubMed and the Google Scholar database. The
publications included those published since the initial identification of BMPs in
the mammalian ovary in 1999 through July 2016. OUTCOMES Studies using human biological materials have revealed the expression of BMPs,
GDFs and their putative receptors as well as their molecular signaling in the
fundamental cells (oocyte, cumulus/granulosa cells (GCs) and theca/stroma cells)
of the ovarian follicles throughout follicle development. With the availability of
recombinant human BMPs/GDFs and the development of immortalized human cell lines,
functional studies have demonstrated the physiological role of intra-ovarian
BMPs/GDFs in all aspects of ovarian functions, from follicle development to
steroidogenesis, cell–cell communication, oocyte maturation, ovulation and
luteal function. Furthermore, there is crosstalk between these potent ovarian
regulators and the endocrine signaling system. Dysregulation or naturally
occurring mutations within the BMP system may lead to several female reproductive
diseases. The latest development of recombinant BMPs, synthetic BMP inhibitors,
gene therapy and tools for BMP-ligand sequestration has made the BMP pathway a
potential therapeutic target in certain human fertility disorders; however,
further clinical trials are needed. Recent studies have indicated that GDF8 is an
intra-ovarian factor that may play a novel role in regulating ovarian functions in
the human ovary. WIDER IMPLICATIONS Intra-ovarian BMPs/GDFs are critical regulators of folliculogenesis and human
ovarian functions. Any dysregulation or variations in these ligands or their
receptors may affect the related intracellular signaling and influence ovarian
functions, which accounts for several reproductive pathologies and infertility.
Understanding the normal and pathological roles of intra-ovarian BMPs/GDFs,
especially as related to GC functions and follicular fluid levels, will inform
innovative approaches to fertility regulation and improve the diagnosis and
treatment of ovarian disorders.
Collapse
Affiliation(s)
- Hsun-Ming Chang
- Department of Obstetrics and Gynaecology, Center for Reproductive Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing 100191, P.R. China.,Department of Obstetrics and Gynaecology, Child and Family Research Institute, University of British Columbia, Room 317, 950 West 28 Avenue, Vancouver, British Columbia, Canada V5Z 4H4
| | - Jie Qiao
- Department of Obstetrics and Gynaecology, Center for Reproductive Medicine, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing 100191, P.R. China
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, Child and Family Research Institute, University of British Columbia, Room 317, 950 West 28 Avenue, Vancouver, British Columbia, Canada V5Z 4H4
| |
Collapse
|
213
|
York JP, Ren YA, Zeng J, Bin Zhang, Wang F, Chen R, Liu J, Xia X, Zhang P. Growth Arrest Specific 2 (GAS2) is a Critical Mediator of Germ Cell Cyst Breakdown and Folliculogenesis in Mice. Sci Rep 2016; 6:34956. [PMID: 27734842 PMCID: PMC5062118 DOI: 10.1038/srep34956] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/21/2016] [Indexed: 01/25/2023] Open
Abstract
In the mouse ovary, the primordial follicle pool is established through a diverse array of signaling pathways and tissue remodeling events. Growth arrest specific gene two (GAS2) is a highly conserved cytoskeleton-associated protein whose in vivo function remains unclear. In Drosophila, loss of the GAS2 homolog, Pigs, results in infertility. We demonstrate herein that, in the mouse ovary, GAS2 is expressed in the stromal cells surrounding the oocyte cysts on 16.5 dpc, and in stromal cells surrounding growing follicles during juvenile and adult life. We have generated genetically engineered mice with inactivated Gas2. Gas2 homozygous mutant mice are viable but have severely impaired fertility in females, in which oocyte cyst breakdown is disrupted and follicle growth is impaired, with significantly reduced numbers of large antral follicles and corpora lutea. In these mutant mice, the organization of the basal lamina surrounding developing follicles is severely defective at multiple stages of folliculogenesis. We also found that Notch signaling activity was altered in ovaries from Gas2 null mice around the time of birth and during follicular development later in life. These results indicate that GAS2 is a critical and novel regulator of tissue remodeling in the ovary during oocyte cyst breakdown and folliculogenesis.
Collapse
Affiliation(s)
- J Philippe York
- Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yi Athena Ren
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jie Zeng
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Bin Zhang
- Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA
| | - Fang Wang
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Rui Chen
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Jianqiao Liu
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China
| | - Xuefeng Xia
- The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510150, China.,Houston Methodist Research Institute, 6670 Bertner Ave, Houston, TX 77030, USA.,Biochemistry and Molecular Biology, Baylor College of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Pumin Zhang
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Biochemistry and Molecular Biology, Baylor College of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
214
|
Zhang T, Shen W, De Felici M, Zhang XF. Di(2-ethylhexyl)phthalate: Adverse effects on folliculogenesis that cannot be neglected. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:579-588. [PMID: 27530864 DOI: 10.1002/em.22037] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/30/2016] [Accepted: 07/05/2016] [Indexed: 06/06/2023]
Abstract
Primordial follicle formation and the subsequent transition of follicles through primary and secondary stages constitute crucial events of oogenesis. In particular, in mammals, defects in the processes that precede and accompany the formation of the primordial follicle pool can affect the size of this population significantly, while alterations in follicle activation, growth and maturation can result in premature depletion of the follicle reserve or cause follicle arrest at immature stages. Over the last decade, in vitro and in vivo approaches have been used to provide evidence that exposure to di(2-ethylhexyl)phthalate(DEHP), the most widely used plasticizer, has a deleterious effect on various stages of folliculogenesis in rodents. There is growing concern, supported by epidemiological and experimental data, that DEHP may have similar effects in women. This article reviews the evidence, with particular reference to our own findings, that DEHP may actually exert a variety of adverse effects on mammalian folliculogenesis from early to final stages of oogenesis, including altered development of the primordial germ cells, impaired fetal oocyte survival and meiotic progression, reduced oocyte nest breakdown, acceleration of primordial follicle activation, altered follicle steroidogenesis and increased follicle atresia. These effects can cause serious complications for reproductive and nonreproductive women's health. In addition, emerging data indicate that phthalates, including DEHP, may cause subtle epigenetic changes in germ cells that can be transmitted to subsequent generations, with potential negative effects on human health. Environ. Mol. Mutagen. 57:589-604, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Teng Zhang
- College of Biological and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, 430023, China
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Wei Shen
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata,", Rome, 00133, Italy.
| | - Xi-Feng Zhang
- College of Biological and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, 430023, China.
| |
Collapse
|
215
|
Dranow DB, Hu K, Bird AM, Lawry ST, Adams MT, Sanchez A, Amatruda JF, Draper BW. Bmp15 Is an Oocyte-Produced Signal Required for Maintenance of the Adult Female Sexual Phenotype in Zebrafish. PLoS Genet 2016; 12:e1006323. [PMID: 27642754 PMCID: PMC5028036 DOI: 10.1371/journal.pgen.1006323] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 08/25/2016] [Indexed: 11/30/2022] Open
Abstract
Although the zebrafish is a major model organism, how they determine sex is not well understood. In domesticated zebrafish, sex determination appears to be polygenic, being influenced by multiple genetic factors that may vary from strain to strain, and additionally can be influenced by environmental factors. However, the requirement of germ cells for female sex determination is well documented: animals that lack germ cells, or oocytes in particular, develop exclusively as males. Recently, it has been determined that oocytes are also required throughout the adult life of the animal to maintain the differentiated female state. How oocytes control sex differentiation and maintenance of the sexual phenotype is unknown. We therefore generated targeted mutations in genes for two oocyte produced signaling molecules, Bmp15 and Gdf9 and here report a novel role for Bmp15 in maintaining adult female sex differentiation in zebrafish. Females deficient in Bmp15 begin development normally but switch sex during the mid- to late- juvenile stage, and become fertile males. Additionally, by generating mutations in the aromatase cyp19a1a, we show that estrogen production is necessary for female development and that the function of Bmp15 in female sex maintenance is likely linked to the regulation of estrogen biosynthesis via promoting the development of estrogen-producing granulosa cells in the oocyte follicle.
Collapse
Affiliation(s)
- Daniel B. Dranow
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
| | - Kevin Hu
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
| | - April M. Bird
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
| | - S. Terese Lawry
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
| | - Melissa T. Adams
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
| | - Angelica Sanchez
- Departments of Pediatrics and Molecular Biology, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - James F. Amatruda
- Departments of Pediatrics and Molecular Biology, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Bruce W. Draper
- Department of Molecular and Cellular Biology, University of California Davis, Davis, California, United States of America
| |
Collapse
|
216
|
Complete in vitro generation of fertile oocytes from mouse primordial germ cells. Proc Natl Acad Sci U S A 2016; 113:9021-6. [PMID: 27457928 DOI: 10.1073/pnas.1603817113] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Reconstituting gametogenesis in vitro is a key goal for reproductive biology and regenerative medicine. Successful in vitro reconstitution of primordial germ cells and spermatogenesis has recently had a significant effect in the field. However, recapitulation of oogenesis in vitro remains unachieved. Here we demonstrate the first reconstitution, to our knowledge, of the entire process of mammalian oogenesis in vitro from primordial germ cells, using an estrogen-receptor antagonist that promotes normal follicle formation, which in turn is crucial for supporting oocyte growth. The fundamental events in oogenesis (i.e., meiosis, oocyte growth, and genomic imprinting) were reproduced in the culture system. The most rigorous evidence of the recapitulation of oogenesis was the birth of fertile offspring, with a maximum of seven pups obtained from a cultured gonad. Moreover, cryopreserved gonads yielded functional oocytes and offspring in this culture system. Thus, our in vitro system will enable both innovative approaches for a deeper understanding of oogenesis and a new avenue to create and preserve female germ cells.
Collapse
|
217
|
Zhang J, Liu W, Sun X, Kong F, Zhu Y, Lei Y, Su Y, Su Y, Li J. Inhibition of mTOR Signaling Pathway Delays Follicle Formation in Mice. J Cell Physiol 2016; 232:585-595. [PMID: 27301841 DOI: 10.1002/jcp.25456] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/13/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Jing Zhang
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| | - Wenwen Liu
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| | - Xinhui Sun
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| | - Feifei Kong
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| | - Ye Zhu
- Department of Gynecology; Nanjing Maternal and Child Health Hospital; Nanjing Medical University; Nanjing China
| | - Yue Lei
- Department of Gynecology; Nanjing Maternal and Child Health Hospital; Nanjing Medical University; Nanjing China
| | - Youqiang Su
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| | - Yiping Su
- Department of Gynecology; Nanjing Maternal and Child Health Hospital; Nanjing Medical University; Nanjing China
| | - Jing Li
- State Key Laboratory of Reproductive Medicine; Nanjing Medical University; Nanjing China
| |
Collapse
|
218
|
Koshel E, Galkina S, Saifitdinova A, Dyomin A, Deryusheva S, Gaginskaya E. Ribosomal RNA gene functioning in avian oogenesis. Cell Tissue Res 2016; 366:533-542. [PMID: 27339801 DOI: 10.1007/s00441-016-2444-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/18/2016] [Indexed: 12/18/2022]
Abstract
Despite long-term exploration into ribosomal RNA gene functioning during the oogenesis of various organisms, many intriguing problems remain unsolved. In this review, we describe nucleolus organizer region (NOR) activity in avian oocytes. Whereas oocytes from an adult avian ovary never reveal the formation of the nucleolus in the germinal vesicle (GV), an ovary from juvenile birds possesses both nucleolus-containing and non-nucleolus-containing oocytes. The evolutionary diversity of oocyte NOR functioning and the potential non-rRNA-related functions of the nucleolus in oocytes are also discussed.
Collapse
Affiliation(s)
- Elena Koshel
- Department of Cytology and Histology, Saint-Petersburg State University, Saint-Petersburg, 199034, Russia
| | - Svetlana Galkina
- Department of Cytology and Histology, Saint-Petersburg State University, Saint-Petersburg, 199034, Russia
| | - Alsu Saifitdinova
- Department of Cytology and Histology, Saint-Petersburg State University, Saint-Petersburg, 199034, Russia
| | - Alexandr Dyomin
- Department of Cytology and Histology, Saint-Petersburg State University, Saint-Petersburg, 199034, Russia
| | - Svetlana Deryusheva
- Department of Cytology and Histology, Saint-Petersburg State University, Saint-Petersburg, 199034, Russia.,Department of Embryology, Carnegie Institution for Science, Baltimore, MD 21218, USA
| | - Elena Gaginskaya
- Department of Cytology and Histology, Saint-Petersburg State University, Saint-Petersburg, 199034, Russia.
| |
Collapse
|
219
|
Vaithiyanathan K, Liew SH, Zerafa N, Gamage T, Cook M, O’Reilly LA, Bouillet P, Scott CL, Strasser A, Findlay JK, Hutt KJ. BCL2-modifying factor promotes germ cell loss during murine oogenesis. Reproduction 2016; 151:553-62. [DOI: 10.1530/rep-15-0561] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/25/2016] [Indexed: 11/08/2022]
Abstract
Abstract
Apoptosis plays a prominent role during ovarian development by eliminating large numbers of germ cells from the female germ line. However, the precise mechanisms and regulatory proteins involved in germ cell death are yet to be determined. In this study, we characterised the role of the pro-apoptotic BH3-only protein, BCL2-modifying factor (BMF), in germ cell apoptosis in embryonic and neonatal mouse ovaries. BMF protein was immunohistochemically localised to germ cells at embryonic days 15.5 (E15.5) and E17.5 and postnatal day 1 (PN1), coincident with entry into the meiotic prophase, but was undetectable at E13.5, and only present at low levels at PN3 and PN5. Consistent with this expression pattern, loss of BMF in female mice was associated with a decrease in apoptosis at E15.5 and E17.5. Furthermore, increased numbers of germ cells were found in ovaries from Bmf−/− mice compared with WT animals at E15.5 and PN1. However, germ cell numbers were comparable between Bmf−/− and WT ovaries at PN3, PN5 and PN10. Collectively, these data indicate that BMF mediates foetal oocyte loss and its action limits the maximal number of germ cells attained in the developing ovary, but does not influence the number of primordial follicles initially established in ovarian reserve.
Collapse
|
220
|
Collins JK, Jones KT. DNA damage responses in mammalian oocytes. Reproduction 2016; 152:R15-22. [PMID: 27069010 DOI: 10.1530/rep-16-0069] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/07/2016] [Indexed: 01/31/2023]
Abstract
DNA damage acquired during meiosis can lead to infertility and miscarriage. Hence, it should be important for an oocyte to be able to detect and respond to such events in order to make a healthy egg. Here, the strategies taken by oocytes during their stages of growth to respond to DNA damaging events are reviewed. In particular, recent evidence of a novel pathway in fully grown oocytes helps prevent the formation of mature eggs with DNA damage. It has been found that fully grown germinal vesicle stage oocytes that have been DNA damaged do not arrest at this point in meiosis, but instead undergo meiotic resumption and stall during the first meiotic division. The Spindle Assembly Checkpoint, which is a well-known mitotic pathway employed by somatic cells to monitor chromosome attachment to spindle microtubules, appears to be utilised by oocytes also to respond to DNA damage. As such maturing oocytes are arrested at metaphase I due to an active Spindle Assembly Checkpoint. This is surprising given this checkpoint has been previously studied in oocytes and considered to be weak and ineffectual because of its poor ability to be activated in response to microtubule attachment errors. Therefore, the involvement of the Spindle Assembly Checkpoint in DNA damage responses of mature oocytes during meiosis I uncovers a novel second function for this ubiquitous cellular checkpoint.
Collapse
Affiliation(s)
- Josie K Collins
- Centre for Biological SciencesFaculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK ;
| | - Keith T Jones
- Centre for Biological SciencesFaculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK ;
| |
Collapse
|
221
|
Zhao L, Du X, Huang K, Zhang T, Teng Z, Niu W, Wang C, Xia G. Rac1 modulates the formation of primordial follicles by facilitating STAT3-directed Jagged1, GDF9 and BMP15 transcription in mice. Sci Rep 2016; 6:23972. [PMID: 27050391 PMCID: PMC4822123 DOI: 10.1038/srep23972] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/01/2016] [Indexed: 11/17/2022] Open
Abstract
The size of the primordial follicle pool determines the reproductive potential of mammalian females, and establishment of the pool is highly dependent on specific genes expression. However, the molecular mechanisms by which the essential genes are regulated coordinately to ensure primordial follicle assembly remain a mystery. Here, we show that the small GTPase Rac1 plays an indispensable role in controlling the formation of primordial follicles in mouse ovary. Employing fetal mouse ovary organ culture system, we demonstrate that disruption of Rac1 retarded the breakdown of germline cell cysts while Rac1 overexpression accelerated the formation of primordial follicles. In addition, in vivo inhibitor injection resulted in the formation of multi-oocyte follicles. Subsequent investigation showed that Rac1 induced nuclear import of STAT3 by physical binding. In turn, nuclear STAT3 directly activated the transcription of essential oocyte-specific genes, including Jagged1, GDF9, BMP15 and Nobox. Further, GDF9 and BMP15 regulated the translation of Notch2 via mTORC1 activation in pregranulosa cells. Overexression or addition of Jagged1, GDF9 and BMP15 not only reversed the effect of Rac1 disruption, but also accelerated primordial follicle formation via Notch2 signaling activation. Collectively, these results indicate that Rac1 plays important roles as a key regulator in follicular assembly.
Collapse
Affiliation(s)
- Lihua Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Xinhua Du
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Kun Huang
- 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
| | - Zhen Teng
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Wanbao Niu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Chao Wang
- 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
| |
Collapse
|
222
|
Wang S, Liu J, Li X, Ji X, Zhang J, Wang Y, Cui S. MiR-125b Regulates Primordial Follicle Assembly by Targeting Activin Receptor Type 2a in Neonatal Mouse Ovary. Biol Reprod 2016; 94:83. [PMID: 26962113 DOI: 10.1095/biolreprod.115.131128] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Accepted: 01/26/2016] [Indexed: 01/20/2023] Open
Abstract
The establishment of the primordial follicle pool is crucial for fertility in mammalian females, and the interruption of overall micro-RNA production byDicer1conditional knockout in the female reproductive system results in infertility. However, there are few reports about the functions of individual micro-RNA in regulating primordial follicle assembly. The present study aimed to investigate the function of miR-125b, which is conserved and preferentially expressed in mammalian ovary during primordial follicle assembly. Detection of miR-125b in the developing mouse ovaries by real-time PCR and in situ hybridization showed that it was highly expressed perinatally and specifically located in the ovarian somatic cells. MiR-125b overexpression blocked the process of primordial follicle assembly in cultured newborn mouse ovaries, while its knockdown promoted this process. Further studies showed that miR-125b regulated the activin/Smad2 signaling in neonatal mouse ovary by directly targeting the 3'-untranslated region of activin receptor type 2a (Acvr2a). Overexpression of miR-125b in neonatal mouse ovary suppressed theAcvr2aprotein level, attenuating activin/Smad2 signaling, while knockdown of miR-125b showed the opposite effects. In addition, recombinant human activin A (rh-ActA) down-regulated miR-125b in the neonatal mouse ovary. Overexpression of miR-125b attenuated the promoting effects of rh-ActA on primordial follicle assembly. Taken together, these data suggest that miR-125b blocks the process of primordial follicle assembly, and miR-125b may play this role by regulating the expression ofAcvr2ain the activin/Smad2 signaling pathway.
Collapse
Affiliation(s)
- Shufen Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Jiali Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Xinqiang Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Xiaowen Ji
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Jianfang Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Yue Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Sheng Cui
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| |
Collapse
|
223
|
|
224
|
Zama AM, Bhurke A, Uzumcu M. Effects of Endocrine-disrupting Chemicals on Female Reproductive Health. ACTA ACUST UNITED AC 2016. [DOI: 10.2174/1874070701610010054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endocrine-disrupting chemicals (EDCs) are increasingly prevalent in the environment and the evidence demonstrates that they affect reproductive health, has been accumulating for the last few decades. In this review of recent literature, we present evidence of the effects of estrogen-mimicking EDCs on female reproductive health especially the ovaries and uteri. As representative EDCs, data from studies with a pharmaceutical estrogen, diethylstilbestrol (DES), an organochlorine pesticide methoxychlor (MXC), a phytoestrogen (genistein), and a chemical used in plastics, bisphenol a (BPA) have been presented. We also discuss the effects of a commonly found plasticizer in the environment, a phthalate (DEHP), even though it is not a typical estrogenic EDC. Collectively, these studies show that exposures during fetal and neonatal periods cause developmental reprogramming leading to adult reproductive disease. Puberty, estrous cyclicity, ovarian follicular development, and uterine functions are all affected by exposure to these EDCs. Evidence that epigenetic modifications are involved in the progression to adult disease is also presented.
Collapse
|
225
|
Niu W, Wang Y, Wang Z, Xin Q, Wang Y, Feng L, Zhao L, Wen J, Zhang H, Wang C, Xia G. JNK signaling regulates E-cadherin junctions in germline cysts and determines primordial follicle formation in mice. Development 2016; 143:1778-87. [PMID: 27013242 DOI: 10.1242/dev.132175] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 03/11/2016] [Indexed: 12/23/2022]
Abstract
Physiologically, the size of the primordial follicle pool determines the reproductive lifespan of female mammals, while its establishment largely depends on a process of germline cyst breakdown during the perinatal period. The mechanisms regulating this process are poorly understood. Here we demonstrate that c-Jun amino-terminal kinase (JNK) signaling is crucial for germline cyst breakdown and primordial follicle formation. JNK was specifically localized in oocytes and its activity increased as germline cyst breakdown progressed. Importantly, disruption of JNK signaling with a specific inhibitor (SP600125) or knockdown technology (Lenti-JNK-shRNAs) resulted in significantly suppressed cyst breakdown and primordial follicle formation in cultured mouse ovaries. Our results show that E-cadherin is intensely expressed in germline cysts, and that its decline is necessary for oocyte release from the cyst. However, inhibition of JNK signaling leads to aberrantly enhanced localization of E-cadherin at oocyte-oocyte contact sites. WNT4 expression is upregulated after SP600125 treatment. Additionally, similar to the effect of SP600125 treatment, WNT4 overexpression delays cyst breakdown and is accompanied by abnormal E-cadherin expression patterns. In conclusion, our results suggest that JNK signaling, which is inversely correlated with WNT4, plays an important role in perinatal germline cyst breakdown and primordial follicle formation by regulating E-cadherin junctions between oocytes in mouse ovaries.
Collapse
Affiliation(s)
- Wanbao Niu
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Ye Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Zhengpin Wang
- Laboratory of Cellular and Developmental Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qiliang Xin
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Yijing Wang
- National Institute of Biological Sciences, Zhongguancun Life Science Park, Changping, Beijing 102206, China
| | - Lizhao Feng
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Lihua Zhao
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Jia Wen
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Hua Zhang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Chao Wang
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| | - Guoliang Xia
- State Key Laboratory of Agrobiotechnology, College of Biological Science, China Agricultural University, Beijing 100193, China
| |
Collapse
|
226
|
Tsoulis MW, Chang PE, Moore CJ, Chan KA, Gohir W, Petrik JJ, Vickers MH, Connor KL, Sloboda DM. Maternal High-Fat Diet-Induced Loss of Fetal Oocytes Is Associated with Compromised Follicle Growth in Adult Rat Offspring. Biol Reprod 2016; 94:94. [PMID: 26962114 PMCID: PMC4861169 DOI: 10.1095/biolreprod.115.135004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 02/25/2016] [Indexed: 11/12/2022] Open
Abstract
Maternal obesity predisposes offspring to metabolic and reproductive dysfunction. We have shown previously that female rat offspring born to mothers fed a high-fat (HF) diet throughout pregnancy and lactation enter puberty early and display aberrant reproductive cyclicity. The mechanisms driving this reproductive phenotype are currently unknown thus we investigated whether changes in ovarian function were involved. Wistar rats were mated and randomized to: dams fed a control diet (CON) or dams fed a HF diet from conception until the end of lactation (HF). Ovaries were collected from fetuses at Embryonic Day (E) 20, and neonatal ovaries at Day 4 (P4), prepubertal ovaries at P27 and adult ovaries at P120. In a subset of offspring, the effects of a HF diet fed postweaning were evaluated. The present study shows that fetuses of mothers fed a HF diet had significantly fewer oocytes at E20, and in neonates, have reduced AMH signaling that may facilitate an increased number of assembled primordial follicles. Both prepubertally and in adulthood, ovaries show increased follicular atresia. As adults, offspring have reduced FSH responsiveness, low expression levels of estrogen receptor alpha (Eralpha), the oocyte-secreted factor, Gdf9, oocyte-specific RNA binding protein, Dazl, and high expression levels of the granulosa-cell derived factor, AMH, in antral follicles. Together, these data suggest that ovarian compromise in offspring born to HF-fed mothers may arise from changes already observable in the fetus and neonate and in the long term, associated with increased follicular atresia through adulthood.
Collapse
Affiliation(s)
- Michael W Tsoulis
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Pauline E Chang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Caroline J Moore
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Kaitlyn A Chan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Wajiha Gohir
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - James J Petrik
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Mark H Vickers
- Liggins Institute and Gravida, National Centre for Growth and Development, University of Auckland, Aukland, New Zealand
| | - Kristin L Connor
- Department of Health Sciences, Carleton University, Ottawa, Ontario, Canada
| | - Deborah M Sloboda
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada Department of Obstetrics and Gynecology, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
227
|
Marchetti F, Massarotti A, Yauk CL, Pacchierotti F, Russo A. The adverse outcome pathway (AOP) for chemical binding to tubulin in oocytes leading to aneuploid offspring. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2016; 57:87-113. [PMID: 26581746 DOI: 10.1002/em.21986] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/13/2015] [Accepted: 10/13/2015] [Indexed: 06/05/2023]
Abstract
The Organisation for Economic Co-operation and Development (OECD) has launched the Adverse Outcome Pathway (AOP) Programme to advance knowledge of pathways of toxicity and improve the use of mechanistic information in risk assessment. An AOP links a molecular initiating event (MIE) to an adverse outcome (AO) through intermediate key events (KE). Here, we present the scientific evidence in support of an AOP whereby chemicals that bind to tubulin cause microtubule depolymerization resulting in spindle disorganization followed by altered chromosome alignment and segregation and the generation of aneuploidy in female germ cells, ultimately leading to aneuploidy in the offspring. Aneuploidy, an abnormal number of chromosomes that is not an exact multiple of the haploid number, is a well-known cause of human disease and represents a major cause of infertility, pregnancy failure, and serious genetic disorders in the offspring. Among chemicals that induce aneuploidy in female germ cells, a large majority impairs microtubule dynamics and spindle function. Colchicine, a prototypical chemical that binds to tubulin and causes microtubule depolymerization, is used here to illustrate the AOP. This AOP is specific to female germ cells exposed during the periovulation period. Although the majority of the data come from rodent studies, the available evidence suggests that the MIE and KEs are conserved across species and would occur in human oocytes. The development of AOPs related to mutagenicity in germ cells is expected to aid the identification of potential hazards to germ cell genomic integrity and support regulatory efforts to protect population health.
Collapse
Affiliation(s)
- Francesco Marchetti
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - Alberto Massarotti
- Dipartimento Di Scienze Del Farmaco, Università Degli Studi Del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Carole L Yauk
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, Canada
| | - Francesca Pacchierotti
- Division of Health Protection Technologies, Laboratory of Biosafety and Risk Assessment, ENEA CR Casaccia, Rome, Italy
| | | |
Collapse
|
228
|
Role of Notch signaling in granulosa cell proliferation and polyovular follicle induction during folliculogenesis in mouse ovary. Cell Tissue Res 2016; 365:197-208. [PMID: 26899251 DOI: 10.1007/s00441-016-2371-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 01/26/2016] [Indexed: 12/17/2022]
Abstract
In the fetal mouse ovary, oocytes are connected by an intercellular bridge and form germ cell cysts. Folliculogenesis begins after birth. To study the role of Notch signaling in folliculogenesis, double-immunohistochemical localization of laminin and Ki-67 was performed in mouse ovaries from embryonic day 17.5 (E17.5) to postnatal day 4 (P4). Most cysts and follicles contained Ki-67-negative cells; however, a few Ki-67-positive cells were present in cysts from E17.5 through P4, indicating that a small number of pre-granulosa cells continue to proliferate during folliculogenesis. To examine the effects of an inhibitor of Notch signaling (DAPT) and a synthetic estrogen (diethylstilbestrol [DES]) on folliculogenesis, an organ-culture system was established. The numbers of cysts, primordial follicles (PrFs) and primary follicles were unchanged by DES, whereas the total number of PrFs and of PrFs with Ki-67-negative cells was reduced by DAPT. In organ-cultured neonatal ovaries, only DAPT treatment increased degenerating cells defined as oocytes. On the contrary, the number of polyovular follicles (PFs) and the PF incidence were significantly increased in ovaries organ-cultured with DES at day 20 post-grafting. In organ-cultured fetal and neonatal ovaries, DAPT reduced Notch 3 and Hey2 mRNAs, whereas DES increased Hey2 mRNA. These results suggest that Notch signaling in fetal ovaries is involved with PrF assembly by the regulation of oocyte survival rather than by cell proliferation. In PF induction, as a result of the disruption of interactions between oocytes and pre-granulosa cells, DES and Notch signaling act independently.
Collapse
|
229
|
Shimada M, Umehara T, Hoshino Y. Roles of epidermal growth factor (EGF)-like factor in the ovulation process. Reprod Med Biol 2016; 15:201-216. [PMID: 29259438 DOI: 10.1007/s12522-016-0236-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 02/01/2016] [Indexed: 12/22/2022] Open
Abstract
Luteinizing hormone (LH) surge stimulates preovulatory follicles to induce the ovulation process, including oocyte maturation, cumulus expansion, and granulosa cell luteinization. The matured oocytes surrounded by an expanded cumulus cell layer are released from follicles to the oviduct. However, LH receptors are dominantly expressed in granulosa cells, but less in cumulus cells and are not expressed in oocytes, indicating that the secondary factors expressed and secreted from LH-stimulated granulosa cells are required for the induction of the ovulation process. Prostaglandin and progesterone are well-known factors that are produced in granulosa cells and then stimulate in both granulosa and cumulus cells. The mutant mice of prostaglandin synthase (Ptgs2KO mice) or progesterone receptor (PRKO mice) revealed that the functions were essential to accomplish the ovulation process, but not to induce the ovulation process. To identify the factors initiating the transfer of the stimuli of LH surge from granulosa cells to cumulus cells, M. Conti's lab and our group performed microarray analysis of granulosa cells and identified the epidermal growth factor (EGF)-like factor, amphiregulin (AREG), epiregulin (EREG), and β-cellulin (BTC) that act on EGF receptor (EGFR) and then induce the ERK1/2 and Ca2+-PLC pathways in cumulus cells. When each of the pathways was down-regulated using a pharmacological approach or gene targeting study, the induction of cumulus expansion and oocyte maturation were dramatically suppressed, indicating that both pathways are inducers of the ovulation process. However, an in vitro culture study also revealed that the EGFR-induced unphysiological activation of PKC in cumulus cells accelerated oocyte maturation with low cytostatic activity. Thus, the matured oocytes are not arrested at the metaphase II (MII) stage and then spontaneously form pronuclei. The expression of another type of EGF-like factor, neuregulin 1 (NRG1), that does not act on EGFR, but selectively binds to ErbB3 is observed in granulosa cells after the LH surge. NRG1 supports EGFR-induced ERK1/2 phosphorylation, but reduces PKC activity to physiological level in the cumulus cells, which delays the timing of meiotic maturation of oocytes to adjust the timing of ovulation. Thus, both types of EGF-like factor are rapidly induced by LH surge and then stimulate cumulus cells to control ERK1/2 and PKC pathways, which results in the release of matured oocytes with a fertilization competence.
Collapse
Affiliation(s)
- Masayuki Shimada
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science Hiroshima University Higashi-Hiroshima 739-8528 Hiroshima Japan
| | - Takashi Umehara
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science Hiroshima University Higashi-Hiroshima 739-8528 Hiroshima Japan
| | - Yumi Hoshino
- Laboratory of Reproductive Endocrinology, Graduate School of Biosphere Science Hiroshima University Higashi-Hiroshima 739-8528 Hiroshima Japan
| |
Collapse
|
230
|
Teng Z, Wang C, Wang Y, Huang K, Xiang X, Niu W, Feng L, Zhao L, Yan H, Zhang H. Gap junctions are essential for murine primordial follicle assembly immediately before birth. Reproduction 2016; 151:105-15. [PMID: 26554027 DOI: 10.1530/rep-15-0282] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2023]
Abstract
The reserve of primordial follicles determines the reproductive ability of the female mammal over its reproductive life. The primordial follicle is composed of two types of cells: oocytes and surrounding pre-granulosa cells. However, the underlying mechanism regulating primordial follicle assembly is largely undefined. In this study, we found that gap junction communication (GJC) established between the ovarian cells in the perinatal mouse ovary may be involved in the process. First, gap junction structures between the oocyte and surrounding pre-granulosa cells appear at about 19.0 dpc (days post coitum). As many as 12 gap junction-related genes are upregulated at birth, implying that a complex communication may exist between ovarian cells, because specifically silencing the genes of individual gap junction proteins, such as Gja1, Gja4 or both, has no influence on primordial follicle assembly. On the other hand, non-specific blockers of GJC, such as carbenoxolone (CBX) and 18α-glycyrrhetinic acid (AGA), significantly inhibit mouse primordial follicle assembly. We proved that the temporal window for establishment of GJC in the fetal ovary is from 19.5 dpc to 1 dpp (days postpartum). In addition, the expression of ovarian somatic cell (OSC)-specific genes, such as Notch2, Foxl2 and Irx3, was negatively affected by GJC blockers, whereas oocyte-related genes, such as Ybx2, Nobox and Sohlh1, were hardly affected, implying that the establishment of GJC during this period may be more important to OSCs than to oocytes. In summary, our results indicated that GJC involves in the mouse primordial follicle assembly process at a specific temporal window that needs Notch signaling cross-talking.
Collapse
Affiliation(s)
- Zhen Teng
- State Key Laboratory of Agro-BiotechnologyCollege of Biological Science, China Agricultural University, Beijing 100193, China
| | - Chao Wang
- State Key Laboratory of Agro-BiotechnologyCollege of Biological Science, China Agricultural University, Beijing 100193, China
| | - Yijing Wang
- State Key Laboratory of Agro-BiotechnologyCollege of Biological Science, China Agricultural University, Beijing 100193, China
| | - Kun Huang
- State Key Laboratory of Agro-BiotechnologyCollege of Biological Science, China Agricultural University, Beijing 100193, China
| | - Xi Xiang
- State Key Laboratory of Agro-BiotechnologyCollege of Biological Science, China Agricultural University, Beijing 100193, China
| | - Wanbao Niu
- State Key Laboratory of Agro-BiotechnologyCollege of Biological Science, China Agricultural University, Beijing 100193, China
| | - Lizhao Feng
- State Key Laboratory of Agro-BiotechnologyCollege of Biological Science, China Agricultural University, Beijing 100193, China
| | - Lihua Zhao
- State Key Laboratory of Agro-BiotechnologyCollege of Biological Science, China Agricultural University, Beijing 100193, China
| | - Hao Yan
- State Key Laboratory of Agro-BiotechnologyCollege of Biological Science, China Agricultural University, Beijing 100193, China
| | - Hua Zhang
- State Key Laboratory of Agro-BiotechnologyCollege of Biological Science, China Agricultural University, Beijing 100193, China
| |
Collapse
|
231
|
Grossman H, Shalgi R. A Role of MicroRNAs in Cell Differentiation During Gonad Development. Results Probl Cell Differ 2016; 58:309-36. [PMID: 27300184 DOI: 10.1007/978-3-319-31973-5_12] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are a group of small noncoding RNA molecules that play a major role in posttranscriptional regulation of gene expression and are expressed in an organ-specific manner. One miRNA can potentially regulate the expression of several genes, depending on cell type and differentiation stage. miRNAs are differentially expressed in the male and female gonads and have an organ-specific reproductive function. Exerting their affect through germ cells and gonadal somatic cells, miRNAs regulate key proteins necessary for gonad development. The role of miRNAs in the testes is only starting to emerge though they have been shown to be required for adequate spermatogenesis. Widely explored in the ovary, miRNAs were suggested to play a fundamental role in follicles' assembly, growth, differentiation, and ovulation. In this chapter, we focus on data obtained from mice in which distinct proteins that participate in the biosynthesis of miRNAs were conditionally knocked out from germ cells (spermatogonial cells or oocytes) or gonadal somatic cells (Sertoli or granulosa cells). We detail recent advances in identification of particular miRNAs and their significance in the development and function of male and female gonads. miRNAs can serve as biomarkers and therapeutic agents of pathological conditions; thus, elucidating the branched and complex network of reproduction-related miRNAs will aid understanding of gonads' physiology and managing reproduction disorders.
Collapse
Affiliation(s)
- Hadas Grossman
- Department of Cell Biology and Development, Tel Aviv University, Ramat Aviv, Israel
| | - Ruth Shalgi
- Department of Cell Biology and Development, Tel Aviv University, Ramat Aviv, Israel.
| |
Collapse
|
232
|
Świątek P, Płachno BJ, Marchant R, Gorgoń S, Krodkiewska M, Małota K, Urbisz AZ. Germ-line cells do not form syncytial cysts in the ovaries of the basal clitellate annelid Capilloventer australis. ZOOL ANZ 2016. [DOI: 10.1016/j.jcz.2015.12.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
233
|
El-Hayek S, Clarke HJ. Control of Oocyte Growth and Development by Intercellular Communication Within the Follicular Niche. Results Probl Cell Differ 2016; 58:191-224. [PMID: 27300180 DOI: 10.1007/978-3-319-31973-5_8] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In the mammalian ovary, each oocyte grows and develops within its own structural and developmental niche-the follicle. Together with the female germ cell in the follicle are somatic granulosa cells, specialized companion cells that surround the oocyte and provide support to it, and an outer layer of thecal cells that serve crucial roles including steroid synthesis. These follicular compartments function as a single physiological unit whose purpose is to produce a healthy egg, which upon ovulation can be fertilized and give rise to a healthy embryo, thus enabling the female germ cell to fulfill its reproductive potential. Beginning from the initial stage of follicle formation and until terminal differentiation at ovulation, oocyte and follicle growth depend absolutely on cooperation between the different cellular compartments. This cooperation synchronizes the initiation of oocyte growth with follicle activation. During growth, it enables metabolic support for the follicle-enclosed oocyte and allows the follicle to fulfill its steroidogenic potential. Near the end of the growth period, intra-follicular interactions prevent the precocious meiotic resumption of the oocyte and ensure its nuclear differentiation. Finally, cooperation enables the events of ovulation, including meiotic maturation of the oocyte and expansion of the cumulus granulosa cells. In this chapter, we discuss the cellular interactions that enable the growing follicle to produce a healthy oocyte, focusing on the communication between the germ cell and the surrounding granulosa cells.
Collapse
Affiliation(s)
- Stephany El-Hayek
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada
- Department of Biology, McGill University, Montreal, QC, Canada
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Block E-M0.2218, Montreal, QC, Canada, H4A 3J1
| | - Hugh J Clarke
- Department of Obstetrics and Gynecology, McGill University, Montreal, QC, Canada.
- Department of Biology, McGill University, Montreal, QC, Canada.
- Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Block E-M0.2218, Montreal, QC, Canada, H4A 3J1.
- Department of Experimental Medicine, McGill University, Montreal, QC, Canada.
| |
Collapse
|
234
|
Sex Specification and Heterogeneity of Primordial Germ Cells in Mice. PLoS One 2015; 10:e0144836. [PMID: 26700643 PMCID: PMC4689518 DOI: 10.1371/journal.pone.0144836] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/23/2015] [Indexed: 11/19/2022] Open
Abstract
In mice, primordial germ cells migrate into the genital ridges by embryonic day 13.5 (E13.5), where they are then subjected to a sex-specific fate with female and male primordial germ cells undergoing mitotic arrest and meiosis, respectively. However, the sex-specific basis of primordial germ cell differentiation is poorly understood. The aim of this study was to investigate the sex-specific features of mouse primordial germ cells. We performed RNA-sequencing (seq) of E13.5 female and male mouse primordial germ cells using next-generation sequencing. We identified 651 and 428 differentially expressed transcripts (>2-fold, P < 0.05) in female and male primordial germ cells, respectively. Of these, many transcription factors were identified. Gene ontology and network analysis revealed differing functions of the identified female- and male-specific genes that were associated with primordial germ cell acquisition of sex-specific properties required for differentiation into germ cells. Furthermore, DNA methylation and ChIP-seq analysis of histone modifications showed that hypomethylated gene promoter regions were bound with H3K4me3 and H3K27me3. Our global transcriptome data showed that in mice, primordial germ cells are decisively assigned to a sex-specific differentiation program by E13.5, which is necessary for the development of vital germ cells.
Collapse
|
235
|
Physiologic Course of Female Reproductive Function: A Molecular Look into the Prologue of Life. J Pregnancy 2015; 2015:715735. [PMID: 26697222 PMCID: PMC4678088 DOI: 10.1155/2015/715735] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 10/29/2015] [Indexed: 12/27/2022] Open
Abstract
The genetic, endocrine, and metabolic mechanisms underlying female reproduction are numerous and sophisticated, displaying complex functional evolution throughout a woman's lifetime. This vital course may be systematized in three subsequent stages: prenatal development of ovaries and germ cells up until in utero arrest of follicular growth and the ensuing interim suspension of gonadal function; onset of reproductive maturity through puberty, with reinitiation of both gonadal and adrenal activity; and adult functionality of the ovarian cycle which permits ovulation, a key event in female fertility, and dictates concurrent modifications in the endometrium and other ovarian hormone-sensitive tissues. Indeed, the ultimate goal of this physiologic progression is to achieve ovulation and offer an adequate environment for the installation of gestation, the consummation of female fertility. Strict regulation of these processes is important, as disruptions at any point in this evolution may equate a myriad of endocrine-metabolic disturbances for women and adverse consequences on offspring both during pregnancy and postpartum. This review offers a summary of pivotal aspects concerning the physiologic course of female reproductive function.
Collapse
|
236
|
Yamashiro C, Hirota T, Kurimoto K, Nakamura T, Yabuta Y, Nagaoka SI, Ohta H, Yamamoto T, Saitou M. Persistent Requirement and Alteration of the Key Targets of PRDM1 During Primordial Germ Cell Development in Mice. Biol Reprod 2015; 94:7. [PMID: 26586842 DOI: 10.1095/biolreprod.115.133256] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/10/2015] [Indexed: 12/24/2022] Open
Abstract
Primordial germ cells (PGCs) are the foundation of totipotency and vital for reproduction and heredity. PGCs in mice arise from the epiblast around Embryonic Day (E) 7.0, migrate through the hindgut endoderm, and colonize and proliferate in the embryonic gonads until around E13.5 prior to their differentiation either into prospermatogonia or oogonia. PRDM1, a transcriptional repressor, plays an essential role in PGC specification that includes robustly repressing a somatic mesodermal program. Using an inducible conditional knockout system, we show here that PRDM1 is critically required throughout PGC development. When Prdm1 was deleted in migrating PGCs at E9.5 or E10.5, or in male gonadal PGCs at E11.5, PGCs were eliminated by apoptosis from around E10.5, E11.5, or E13.5, respectively. When Prdm1 was deleted in female gonadal PGCs at E11.5, PGCs progressed into the first meiotic prophase in an apparently normal fashion, but the oogonia exhibited an aberrant pachytene phenotype, undergoing abrupt apoptosis from around E16.5. The escape of a fraction of PGCs (∼10%) from the Prdm1 deletion was sufficient to recover fairly normal germ cell pools, both in male and female adults. The key targets of PRDM1 in migrating and/or gonadal PGCs, including genes for development, apoptosis, and prospermatogonial differentiation, showed only a modest overlap with those upon PGC specification, and were enriched with histone H3 lysine 27 trimethylation (H3K27me3). Our findings provide critical insight into the mechanism for maintaining the transcriptional integrity of PGCs.
Collapse
Affiliation(s)
- Chika Yamashiro
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO), Kyoto, Japan
| | - Takayuki Hirota
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO), Kyoto, Japan
| | - Kazuki Kurimoto
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO), Kyoto, Japan
| | - Tomonori Nakamura
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO), Kyoto, Japan
| | - Yukihiro Yabuta
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO), Kyoto, Japan
| | - So I Nagaoka
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Ohta
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO), Kyoto, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan JST, Agency for Medical Research and Development (AMED) - Core Research for Evolutional Science and Technology (CREST), Saitama, Japan
| | - Mitinori Saitou
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan Japan Science and Technology Agency (JST), Exploratory Research for Advanced Technology (ERATO), Kyoto, Japan Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
237
|
Teng Z, Wang C, Wang Y, Huang K, Xiang X, Niu W, Feng L, Zhao L, Yan H, Zhang H, Xia G. S100A8, An Oocyte-Specific Chemokine, Directs the Migration of Ovarian Somatic Cells During Mouse Primordial Follicle Assembly. J Cell Physiol 2015; 230:2998-3008. [PMID: 25953201 DOI: 10.1002/jcp.25032] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/04/2015] [Indexed: 01/22/2023]
Abstract
In the mammalian ovaries, the primordial follicle pool determines the reproductive capability over the lifetime of a female. The primordial follicle is composed of two cell members, namely the oocyte and the pre-granulosa cells that encircle the oocyte. However, it is unclear what factors are involved in the reorganization of the two distinct cells into one functional unit. This study was performed to address this issue. Firstly, in an in vitro reconstruction system, dispersed ovarian cells from murine fetal ovaries at 19.0 days post coitum (dpc) reassembled into follicle-like structures, independent of the physical distance between the cells, implying that either oocytes or ovarian somatic cells (OSCs) were motile. We then carried out a series of transwell assay experiments, and determined that it was in fact 19.0 dpc OSCs (as opposed to oocytes), which exhibited a significant chemotactic response to both fetal bovine serum and oocytes themselves. We observed that S100A8, a multi-functional chemokine, may participate in the process as it is mainly expressed in oocytes within the cysts/plasmodia. S100A8 significantly promoted the number of migrating OSCs by 2.5 times in vitro, of which 66.9% were FOXL2 protein-positive cells, implying that the majority of motile OSCs were pre-granulosa cells. In addition, an S100A8-specific antibody inhibited the formation of follicle-like reconstruction cell mass in vitro. And, the primordial follicle formation was reduced when S100a8-specific siRNA was applied onto in vitro cultured 17.5 dpc ovary. Therefore, S100A8 could be a chemokine of oocyte origin, which attracts OSCs to form the primordial follicles.
Collapse
Affiliation(s)
- Zhen Teng
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Chao Wang
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Yijing Wang
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Kun Huang
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Xi Xiang
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Wanbao Niu
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Lizhao Feng
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Lihua Zhao
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Hao Yan
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Hua Zhang
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| | - Guoliang Xia
- State Key Laboratory of Agro-biotechnology, College of Biological Science, China Agricultural University, Beijing,, China
| |
Collapse
|
238
|
Abstract
The adult mammalian ovary is devoid of definitive germline stem cells. As such, female reproductive senescence largely results from the depletion of a finite ovarian follicle pool that is produced during embryonic development. Remarkably, the crucial nature and regulation of follicle assembly and survival during embryogenesis is just coming into focus. This developmental pathway involves the coordination of meiotic progression and the breakdown of germ cell cysts into individual oocytes housed within primordial follicles. Recent evidence also indicates that genetic and environmental factors can specifically perturb primordial follicle assembly. Here, we review the cellular and molecular mechanisms by which the mammalian ovarian reserve is established, highlighting the presence of a crucial checkpoint that allows survival of only the highest-quality oocytes.
Collapse
Affiliation(s)
- Kathryn J Grive
- Brown University, MCB Graduate Program, Providence, RI 02912, USA
| | | |
Collapse
|
239
|
Malki S, Tharp ME, Bortvin A. A Whole-Mount Approach for Accurate Quantitative and Spatial Assessment of Fetal Oocyte Dynamics in Mice. Biol Reprod 2015; 93:113. [PMID: 26423126 DOI: 10.1095/biolreprod.115.132118] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/23/2015] [Indexed: 11/01/2022] Open
Abstract
Depletion of oocytes from the embryonic ovary is a key feature of mammalian oogenesis; however, the rational and molecular bases for this phenomenon remain poorly understood. Presently in the field, the most systematic analysis used to understand the effect of a given molecular pathway on fetal oocyte attrition is to count the number of oocytes in ovaries at different stages of development. This analysis is commonly done using a sampling method based on sectioning of the ovary, a technique that includes many laborious steps culminating in an inaccurate estimate of oocyte number contained within that ovary. This inability to generate data that are directly comparable between labs hinders the field and raises questions about the timing and rate of oocyte depletion. Therefore, we set out to implement a robust method that can be easily used by most research laboratories to study the dynamics of oogenesis during fetal mouse ovary development in both normal and experimental conditions. Here we describe an approach to accurately count the total number of oocytes in embryonic ovaries. This method is based on whole-mount immunofluorescence, tissue clearing with sucrose and ScaleA2 reagent, and automatic detection and counting of germ cells in intact ovaries using confocal microscopy and three-dimensional software analyses. We demonstrate the power of the method by assessing variation of fetal oocyte numbers between left and right ovaries and between litters of mice. Finally, we anticipate that the method could be adopted to the analysis of substages of meiotic prophase I and ovarian somatic cells.
Collapse
Affiliation(s)
- Safia Malki
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland
| | - Marla E Tharp
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | - Alex Bortvin
- Department of Embryology, Carnegie Institution for Science, Baltimore, Maryland
| |
Collapse
|
240
|
Suzuki H, Kanai-Azuma M, Kanai Y. From Sex Determination to Initial Folliculogenesis in Mammalian Ovaries: Morphogenetic Waves along the Anteroposterior and Dorsoventral Axes. Sex Dev 2015; 9:190-204. [DOI: 10.1159/000440689] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2015] [Indexed: 11/19/2022] Open
|
241
|
Yaba A, Ordueri NEG, Tanriover G, Sahin P, Demir N, Celik-Ozenci C. Expression of CCM2 and CCM3 during mouse gonadogenesis. J Assist Reprod Genet 2015; 32:1497-507. [PMID: 26386873 DOI: 10.1007/s10815-015-0559-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/20/2015] [Indexed: 11/25/2022] Open
Abstract
PURPOSE Three cerebral cavernous malformation (CCM) proteins, CCM1, CCM2, and CCM3, regulate cell-cell adhesion, cell shape and polarity, and most likely cell adhesion to extracellular matrix. Recently, CCM2 and CCM3 are known to be expressed in control and varicocele-induced rat testes, but little is known about these proteins during gonadogenesis. This led us to study the CCM proteins during the mouse gonadogenesis. METHODS Neonatal (PND 0), postnatal, and adult mice testes and ovaries were obtained from mice. CCM2 and CCM3 expression were analyzed during mouse testicular and ovarian development by immunohistochemistry and quantitative real-time PCR. RESULTS The results showed that in both sexes, Ccm2 and Ccm3 mRNA and protein were first detectable after gonadogenesis when the gonads were well differentiated and remained present until the adult stage. In the testis, CCM2 and CCM3 expression were restricted to the nuclei of Sertoli cells, suggesting a conserved role in testicular differentiation. In the ovary, the CCM2 and CCM3 proteins were localized in the cytoplasm of oocytes, suggesting an unexpected role during oogenesis. Quantitative real-time PCR (qRT-PCR) results showed that expression of Ccm2 and Ccm3 genes could play a role in the regulation of mouse gonadogenesis translational activation upon testicular and ovarian development. CONCLUSIONS The localization of CCM2 and CCM3 proteins show their different functions for CCM2 and CCM3 which may have important roles in testicular and ovarian differentiation. In conclusion, CCM2 and CCM3 may be involved in establishing the differential expression pattern in developing mouse testis and ovary.
Collapse
Affiliation(s)
- Aylin Yaba
- Department of Histology and Embryology, İstanbul Medipol University School of Medicine, İstanbul, Turkey
| | - N Ece Gungor Ordueri
- Department of Histology and Embryology, Akdeniz University School of Medicine, 07070 Campus, Antalya, Turkey
| | - Gamze Tanriover
- Department of Histology and Embryology, Akdeniz University School of Medicine, 07070 Campus, Antalya, Turkey
| | - Pinar Sahin
- Department of Histology and Embryology, Akdeniz University School of Medicine, 07070 Campus, Antalya, Turkey
| | - Necdet Demir
- Department of Histology and Embryology, Akdeniz University School of Medicine, 07070 Campus, Antalya, Turkey
| | - Ciler Celik-Ozenci
- Department of Histology and Embryology, Akdeniz University School of Medicine, 07070 Campus, Antalya, Turkey.
| |
Collapse
|
242
|
Stanley JA, Arosh JA, Burghardt RC, Banu SK. A fetal whole ovarian culture model for the evaluation of CrVI-induced developmental toxicity during germ cell nest breakdown. Toxicol Appl Pharmacol 2015; 289:58-69. [PMID: 26348139 DOI: 10.1016/j.taap.2015.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 08/18/2015] [Accepted: 09/01/2015] [Indexed: 01/08/2023]
Abstract
Prenatal exposure to endocrine disrupting chemicals (EDCs), including bisphenol A, dioxin, pesticides, and cigarette smoke, has been linked to several ovarian diseases such as premature ovarian failure (POF) and early menopause in women. Hexavalent chromium (CrVI), one of the more toxic heavy metals, is widely used in more than 50 industries. As one of the world's leading producers of Cr compounds, the U.S. is facing growing challenges in protecting human health against adverse effects of CrVI. Our recent findings demonstrated that in vivo CrVI exposure during gestational period caused POF in F1 offspring. Our current research focus is three-fold: (i) to identify the effect of CrVI on critical windows of great vulnerability of fetal ovarian development; (ii) to understand the molecular mechanism of CrVI-induced POF; (iii) to identify potential intervention strategies to mitigate or inhibit CrVI effects. In order to accomplish these goals we used a fetal whole ovarian culture system. Fetuses were removed from the normal pregnant rats on gestational day 13.5. Fetal ovaries were cultured in vitro for 12 days, and treated with or without 0.1 ppm potassium dichromate (CrVI) from culture day 2-8, which recapitulated embryonic day 14.5-20.5, in vivo. Results showed that CrVI increased germ cell/oocyte apoptosis by increasing caspase 3, BAX, p53 and PUMA; decreasing BCL2, BMP15, GDF9 and cKIT; and altering cell cycle regulatory genes and proteins. This model system may serve as a potential tool for high throughput testing of various drugs and/or EDCs in particular to assess developmental toxicity of the ovary.
Collapse
Affiliation(s)
- Jone A Stanley
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Joe A Arosh
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Sakhila K Banu
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
243
|
Cordeiro MH, Kim SY, Ebbert K, Duncan FE, Ramalho-Santos J, Woodruff TK. Geography of follicle formation in the embryonic mouse ovary impacts activation pattern during the first wave of folliculogenesis. Biol Reprod 2015; 93:88. [PMID: 26246221 DOI: 10.1095/biolreprod.115.131227] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 08/04/2015] [Indexed: 11/01/2022] Open
Abstract
During embryonic development, mouse female germ cells enter meiosis in an anterior-to-posterior wave believed to be driven by retinoic acid. It has been proposed that ovarian follicle formation and activation follow the same general wave of meiotic progression; however, the precise anatomic specification of these processes has not been delineated. Here, we created a mouse line using Mvh, Gdf9, and Zp3 promoters to drive distinct temporal expression of three fluorescent proteins in the oocytes and to identify where the first follicle cohort develops. The fluorescent profile revealed that the first growing follicles consistently appeared in a specific region of the ovary, the anterior-dorsal region, which led us to analyze if meiotic onset occurred earlier in the dorsal ovarian region. Surprisingly, in addition to the anterior-to-posterior wave, we observed an early meiotic entry in the ventral region of the ovary. This additional anatomic stratification of meiosis contrasts with the localization of the initial follicle formation and activation in the dorsal region of the ovary. Therefore, our study suggests that the specification of cortical and medullar areas in the ventral and dorsal regions on the ovary, rather than the onset of meiosis, impacts where the first follicle activation event occurs.
Collapse
Affiliation(s)
- Marília H Cordeiro
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal Doctoral Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - So-Youn Kim
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Katherine Ebbert
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Francesca E Duncan
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - João Ramalho-Santos
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Teresa K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
244
|
Abstract
Primordial follicles (PF) are formed when somatic cells differentiate into flattened pregranulosa cells, invaginate into the oocyte nests and encircle individual oocytes. We hypothesize that BMP2 regulates PF formation by promoting the transition of germ cells into oocytes and somatic cells into pregranulosa cells. E15 hamster ovaries were cultured for 8 days corresponding to postnatal day 8 (P8) in vivo, with or without BMP2, and the formation of PF was examined. BMP2 was expressed in the oocytes as well as ovarian somatic cells during development. BMP2 exposure for the first two days or the last two days or the entire 8 days of culture led to increase in PF formation suggesting that BMP2 affected both germ cell transition and somatic cell differentiation. Whereas an ALK2/3 inhibitor completely blocked BMP2-induced PF formation, an ALK2-specific inhibitor was partially effective, suggesting that BMP2 affected PF formation via both ALK2 and ALK3. BMP2 also reduced apoptosis in vitro. Further, more meiotic oocytes were present in BMP2 exposed ovaries. In summary, the results provide the first evidence that BMP2 regulates primordial follicle formation by promoting germ cell to oocyte transition and somatic cell to pre-granulosa cells formation and it acts via both ALK2 and ALK3.
Collapse
Affiliation(s)
| | - Shyamal K Roy
- 1] Department of Cellular and Integrative Physiology, and Obstetrics and Gynecology [2] University of Nebraska Medical Center, Omaha, NE
| |
Collapse
|
245
|
Lim AK, Knowles BB. Controlling Endogenous Retroviruses and Their Chimeric Transcripts During Natural Reprogramming in the Oocyte. J Infect Dis 2015; 212 Suppl 1:S47-51. [DOI: 10.1093/infdis/jiu567] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
246
|
Ren Y, Suzuki H, Jagarlamudi K, Golnoski K, McGuire M, Lopes R, Pachnis V, Rajkovic A. Lhx8 regulates primordial follicle activation and postnatal folliculogenesis. BMC Biol 2015; 13:39. [PMID: 26076587 PMCID: PMC4487509 DOI: 10.1186/s12915-015-0151-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/10/2015] [Indexed: 12/23/2022] Open
Abstract
Background The early stages of ovarian follicle formation—beginning with the breakdown of germ cell cysts and continuing with the formation of primordial follicles and transition to primary and secondary follicles—are critical in determining reproductive life span and fertility. Previously, we discovered that global knockouts of germ cell-specific transcriptional co-regulators Sohlh1, Sohlh2, Lhx8, and Nobox, cause rapid oocyte loss and ovarian failure. Also factors such as Nobox and Sohlh1 are associated with human premature ovarian failure. In this study, we developed a conditional knockout of Lhx8 to study oocyte-specific pathways in postnatal folliculogenesis. Results The conditional deficiency of Lhx8 in the oocytes of primordial follicles leads to massive primordial oocyte activation, in part, by indirectly interacting with the PI3K-AKT pathway, as shown by synergistic effects on FOXO3 nucleocytoplasmic translocation and rpS6 activation. However, LHX8 does not directly regulate members of the PI3K-AKT pathway; instead, we show that LHX8 represses Lin28a expression, a known regulator of mammalian metabolism and of the AKT/mTOR pathway. LHX8 can bind to the Lin28a promoter, and the depletion of Lin28a in Lhx8-deficient oocytes partially suppresses primordial oocyte activation. Moreover, unlike the PI3K-AKT pathway, LHX8 is critical beyond primordial follicle activation, and blocks the primary to secondary follicle transition. Conclusions Our results indicate that the LHX8-LIN28A pathway is essential in the earliest stages of primordial follicle activation, and LHX8 is an important oocyte-specific transcription factor in the ovary for regulating postnatal folliculogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s12915-015-0151-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu Ren
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Hitomi Suzuki
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Experimental Animal Model for Human Disease, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8510, Japan.
| | - Krishna Jagarlamudi
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Kayla Golnoski
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Megan McGuire
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Rita Lopes
- Division of Molecular Neurobiology, MRC National Institute of Medical Research, London, NW7 1AA, UK.
| | - Vassilis Pachnis
- Division of Molecular Neurobiology, MRC National Institute of Medical Research, London, NW7 1AA, UK.
| | - Aleksandar Rajkovic
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
247
|
Wang Z, Niu W, Wang Y, Teng Z, Wen J, Xia G, Wang C. Follistatin288 Regulates Germ Cell Cyst Breakdown and Primordial Follicle Assembly in the Mouse Ovary. PLoS One 2015; 10:e0129643. [PMID: 26076381 PMCID: PMC4468113 DOI: 10.1371/journal.pone.0129643] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 05/11/2015] [Indexed: 11/18/2022] Open
Abstract
In mammals, the primordial follicle pool represents the entire reproductive potential of a female. The transforming growth factor-β (TGF-β) family member activin (ACT) contributes to folliculogenesis, although the exact mechanism is not known. The role of FST288, the strongest ACT-neutralizing isoform of follistatin (FST), during cyst breakdown and primordial follicle formation in the fetal mice ovary was assessed using an in vitro culture system. FST was continuously expressed in the oocytes as well as the cuboidal granulosa cells of growing follicles in perinatal mouse ovaries. Treatment with FST288 delayed germ cell nest breakdown, particularly near the periphery of the ovary, and dramatically decreased the percentage of primordial follicles. In addition, there was a dramatic decrease in proliferation of granulosa cells and somatic cell expression of Notch signaling was impaired. In conclusion, FST288 impacts germ cell nest breakdown and primordial follicle assembly by inhibiting somatic cell proliferation.
Collapse
Affiliation(s)
- Zhengpin Wang
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Wanbao Niu
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Yijing Wang
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Zhen Teng
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Jia Wen
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Guoliang Xia
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Chao Wang
- State Key Laboratory of Agro-Biotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, People’s Republic of China
- * E-mail:
| |
Collapse
|
248
|
Zhou C, Wang W, Peretz J, Flaws JA. Bisphenol A exposure inhibits germ cell nest breakdown by reducing apoptosis in cultured neonatal mouse ovaries. Reprod Toxicol 2015; 57:87-99. [PMID: 26049153 DOI: 10.1016/j.reprotox.2015.05.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/30/2015] [Accepted: 05/26/2015] [Indexed: 11/30/2022]
Abstract
Bisphenol A is a known endocrine disrupting chemical and reproductive toxicant. Previous studies indicate that in utero BPA exposure increases the percentage of germ cells in nests and decreases the percentage of primordial follicles. However, the mechanism by which BPA affects germ cell nest breakdown is unknown. Thus, we hypothesized that BPA inhibits germ cell nest breakdown by interfering with oxidative stress and apoptosis pathways. To test our hypothesis, ovaries from newborn mice were collected and cultured with vehicle (dimethyl sulfoxide, DMSO) or different doses of BPA (0.1, 1, 5, and 10μg/mL). Ovaries then were subjected to histological evaluation of germ cell nests and primordial follicles or to measurements of factors that regulate oxidative stress and apoptosis. Our results indicate that in vitro BPA exposure significantly inhibits germ cell nest breakdown by altering the expression of key ovarian apoptotic genes, but not by interfering with the oxidative stress pathway.
Collapse
Affiliation(s)
- Changqing Zhou
- Department of Comparative Biosciences, University of Illinois, Urbana, IL 61802, USA.
| | - Wei Wang
- Department of Comparative Biosciences, University of Illinois, Urbana, IL 61802, USA.
| | - Jackye Peretz
- Department of Comparative Biosciences, University of Illinois, Urbana, IL 61802, USA.
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois, Urbana, IL 61802, USA.
| |
Collapse
|
249
|
Sun T, Pepling ME, Diaz FJ. Lats1 Deletion Causes Increased Germ Cell Apoptosis and Follicular Cysts in Mouse Ovaries. Biol Reprod 2015; 93:22. [PMID: 26040669 DOI: 10.1095/biolreprod.114.118604] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 05/26/2015] [Indexed: 12/13/2022] Open
Abstract
The Hippo signaling pathway is essential for regulating proliferation and apoptosis in mammalian cells. The LATS1 kinase is a core member of the Hippo signaling pathway that phosphorylates and inactivates the transcriptional co-activators YAP1 and WWTR1. Deletion of Lats1 results in low neonate survival and ovarian stromal tumors in surviving adults, but the effects of Lats1 on early follicular development are not understood. Here, the expression of Hippo pathway components including Wwtr1, Stk4, Stk3, Lats2, and Yap1 transcripts were decreased by 50% in mouse ovaries between 2 and 8 days of age while expression was maintained from 8 days to 21 days and after priming with eCG. LATS1, LATS2, and MOB1B were localized to both germ and somatic cells of primordial to antral follicles. Interestingly, YAP1 was predominantly cytoplasmic, whereas WWTR1 was nuclear in oocytes and somatic cells. Deletion of Lats1 caused an increase in germ cell apoptosis from 1.7% in control ovaries to 3.6% in Lats1 mutant ovaries and a 58% and 32% decrease in primordial and activated follicle numbers in cultured mutant ovaries. Surprisingly, there was an increase in Bmp15 but not Gdf9, Figla, Nobox transcripts or the somatic-specific transcripts Amh and Wnt4 in cultured Lats1 mutant ovaries. Last, Lats1 mutant ovaries developed ovarian cysts at a higher frequency (43%) than heterozygous (24%) and control ovaries (8%). Results showed that the Hippo pathway is active in ovarian follicles and that LATS1 is required to maintain the pool of germ cells and primordial follicles.
Collapse
Affiliation(s)
- Tianyanxin Sun
- Center for Reproductive Biology and Health, Department of Animal Science, The Pennsylvania State University, University Park, Pennsylvania
| | | | - Francisco J Diaz
- Center for Reproductive Biology and Health, Department of Animal Science, The Pennsylvania State University, University Park, Pennsylvania
| |
Collapse
|
250
|
Hua J, Xu B, Yang Y, Ban R, Iqbal F, Cooke HJ, Zhang Y, Shi Q. Follicle Online: an integrated database of follicle assembly, development and ovulation. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2015; 2015:bav036. [PMID: 25931457 PMCID: PMC4414955 DOI: 10.1093/database/bav036] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 03/31/2015] [Indexed: 11/16/2022]
Abstract
Folliculogenesis is an important part of ovarian function as it provides the oocytes for female reproductive life. Characterizing genes/proteins involved in folliculogenesis is fundamental for understanding the mechanisms associated with this biological function and to cure the diseases associated with folliculogenesis. A large number of genes/proteins associated with folliculogenesis have been identified from different species. However, no dedicated public resource is currently available for folliculogenesis-related genes/proteins that are validated by experiments. Here, we are reporting a database ‘Follicle Online’ that provides the experimentally validated gene/protein map of the folliculogenesis in a number of species. Follicle Online is a web-based database system for storing and retrieving folliculogenesis-related experimental data. It provides detailed information for 580 genes/proteins (from 23 model organisms, including Homo sapiens, Mus musculus, Rattus norvegicus, Mesocricetus auratus, Bos Taurus, Drosophila and Xenopus laevis) that have been reported to be involved in folliculogenesis, POF (premature ovarian failure) and PCOS (polycystic ovary syndrome). The literature was manually curated from more than 43 000 published articles (till 1 March 2014). The Follicle Online database is implemented in PHP + MySQL + JavaScript and this user-friendly web application provides access to the stored data. In summary, we have developed a centralized database that provides users with comprehensive information about genes/proteins involved in folliculogenesis. This database can be accessed freely and all the stored data can be viewed without any registration. Database URL:http://mcg.ustc.edu.cn/sdap1/follicle/index.php
Collapse
Affiliation(s)
- Juan Hua
- Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan
| | - Bo Xu
- Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan
| | - Yifan Yang
- Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan
| | - Rongjun Ban
- Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan
| | - Furhan Iqbal
- Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and
| | - Howard J Cooke
- Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan
| | - Yuanwei Zhang
- Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and
| | - Qinghua Shi
- Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China, Center for Reproductive Medicine, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, China, Department of Statistics, University of Kentucky, Lexington, KY 40506, USA, Collaborative Innovation Center of Genetics and Development, Fudan University, 2005 Songhu Road, Shanghai 200438, China, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China and Institute of Pure and Applied Biology, Bahauddin Zakariya University Multan, 60800, Pakistan Molecular and Cell Genetics Laboratory, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, University of Science and
| |
Collapse
|