1
|
Alexander AK, Rodriguez KF, Chen YY, Amato CM, Estermann MA, Nicol B, Xu X, Hung-Chang Yao H. Single-nucleus multiomics reveals the gene-regulatory networks underlying sex determination of murine primordial germ cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.19.581036. [PMID: 39386556 PMCID: PMC11463670 DOI: 10.1101/2024.02.19.581036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Accurate specification of female and male germ cells during embryonic development is critical for sexual reproduction. Primordial germ cells (PGCs) are the bipotential precursors of mature gametes that commit to an oogenic or spermatogenic fate in response to sex-determining cues from the fetal gonad. The critical processes required for PGCs to integrate and respond to signals from the somatic environment in gonads are not understood. In this study, we developed the first single-nucleus multiomics map of chromatin accessibility and gene expression during murine PGC development in both XX and XY embryos. Profiling of cell-type specific transcriptomes and regions of open chromatin from the same cell captured the molecular signatures and gene networks underlying PGC sex determination. Joint RNA and ATAC data for single PGCs resolved previously unreported PGC subpopulations and cataloged a multimodal reference atlas of differentiating PGC clusters. We discovered that regulatory element accessibility precedes gene expression during PGC development, suggesting that changes in chromatin accessibility may prime PGC lineage commitment prior to differentiation. Similarly, we found that sexual dimorphism in chromatin accessibility and gene expression increased temporally in PGCs. Combining single-nucleus sequencing data, we computationally mapped the cohort of transcription factors that regulate the expression of sexually dimorphic genes in PGCs. For example, the gene regulatory networks of XX PGCs are enriched for the transcription factors, TFAP2c, TCFL5, GATA2, MGA, NR6A1, TBX4, and ZFX. Sex-specific enrichment of the forkhead-box and POU6 families of transcription factors was also observed in XY PGCs. Finally, we determined the temporal expression patterns of WNT, BMP, and RA signaling during PGC sex determination, and our discovery analyses identified potentially new cell communication pathways between supporting cells and PGCs. Our results illustrate the diversity of factors involved in programming PGCs towards a sex-specific fate.
Collapse
Affiliation(s)
- Adriana K. Alexander
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Karina F. Rodriguez
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Yu-Ying Chen
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Ciro M. Amato
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Martin A. Estermann
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Barbara Nicol
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Xin Xu
- Epigenetics & Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| | - Humphrey Hung-Chang Yao
- Reproductive Developmental Biology Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA
| |
Collapse
|
2
|
Telfer EE, Grosbois J, Odey YL, Rosario R, Anderson RA. Making a good egg: human oocyte health, aging, and in vitro development. Physiol Rev 2023; 103:2623-2677. [PMID: 37171807 PMCID: PMC10625843 DOI: 10.1152/physrev.00032.2022] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 05/03/2023] [Accepted: 05/06/2023] [Indexed: 05/13/2023] Open
Abstract
Mammalian eggs (oocytes) are formed during fetal life and establish associations with somatic cells to form primordial follicles that create a store of germ cells (the primordial pool). The size of this pool is influenced by key events during the formation of germ cells and by factors that influence the subsequent activation of follicle growth. These regulatory pathways must ensure that the reserve of oocytes within primordial follicles in humans lasts for up to 50 years, yet only approximately 0.1% will ever be ovulated with the rest undergoing degeneration. This review outlines the mechanisms and regulatory pathways that govern the processes of oocyte and follicle formation and later growth, within the ovarian stroma, through to ovulation with particular reference to human oocytes/follicles. In addition, the effects of aging on female reproductive capacity through changes in oocyte number and quality are emphasized, with both the cellular mechanisms and clinical implications discussed. Finally, the details of current developments in culture systems that support all stages of follicle growth to generate mature oocytes in vitro and emerging prospects for making new oocytes from stem cells are outlined.
Collapse
Affiliation(s)
- Evelyn E Telfer
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Johanne Grosbois
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Yvonne L Odey
- Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Roseanne Rosario
- Centre for Discovery Brain Sciences, Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard A Anderson
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
3
|
Review: Role and regulatory mechanism of inhibin in animal reproductive system. Theriogenology 2023; 202:10-20. [PMID: 36878034 DOI: 10.1016/j.theriogenology.2023.02.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 03/03/2023]
Abstract
Inhibin (INH) is a glycoprotein hormone secreted by the gonads that inhibit the synthesis and secretion of follicle-stimulating hormone (FSH). Increasing evidence indicates that INH plays a significant role in the development of the reproductive system including follicle development, ovulation rate, corpus luteum formation and ablation, steroid hormone synthesis and spermatogenesis, subsequently affecting the reproductive capacity of animals such as litter size and egg production. There are currently three main views on how INH inhibits FSH synthesis and secretion: influencing the activity of adenylate cyclase, the expression of follicle-stimulating hormone receptor or gonadotropin-releasing hormone receptor, and the competition system of inhibin-activin. This review discusses the current findings on the structure, function, and mechanism of action of INH in the reproductive system of animals.
Collapse
|
4
|
Evans ET, Horst B, Arend RC, Mythreye K. Evolving roles of activins and inhibins in ovarian cancer pathophysiology. Am J Physiol Cell Physiol 2023; 324:C428-C437. [PMID: 36622068 PMCID: PMC9902228 DOI: 10.1152/ajpcell.00178.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 01/10/2023]
Abstract
Activins and inhibins are unique members of the transforming growth factor-β (TGFβ) family of growth factors, with the ability to exert autocrine, endocrine, and paracrine effects in a wide range of complex physiologic and pathologic processes. Although first isolated within the pituitary, emerging evidence suggests broader influence beyond reproductive development and function. Known roles of activin and inhibin in angiogenesis and immunity along with correlations between gene expression and cancer prognosis suggest potential roles in tumorigenesis. Here, we present a review of the current understanding of the biological role of activins and inhibins as it relates to ovarian cancers, summarizing the underlying signaling mechanisms and physiologic influence, followed by detailing their roles in cancer progression, diagnosis, and treatment.
Collapse
Affiliation(s)
- Elizabeth T Evans
- Department of Gynecologic Oncology, Heersink School of Medicine, University of Alabama School of Medicine, Birmingham, Alabama
| | - Ben Horst
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rebecca C Arend
- Department of Gynecologic Oncology, Heersink School of Medicine, University of Alabama School of Medicine, Birmingham, Alabama
| | - Karthikeyan Mythreye
- Department of Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
5
|
A New Bioreactor to Promote Human Follicular Growth with or without Activin A in Transgender Men. REPRODUCTIVE MEDICINE 2023. [DOI: 10.3390/reprodmed4010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The aim of the present study was to evaluate the effect of activin A on the activation of in vitro folliculogenesis of human ovarian tissues from transgender men with or without our new compartmented chitosan hydrogel microbioreactor (“three-dimensional (3D)-structure”) enabling a three-dimensional tissue culture. Five fresh ovarian human tissues were cultured in vitro for 20 or 22 days in four groups with 100 ng/mL activin A or without activin A during the last six to eight days of culture, and within a 3D-structure or without the 3D-structure in standard conditions. Follicular density and quality were evaluated, and follicular diameters were measured. Estradiol secretion was quantified. Proliferation and apoptosis through immunostaining were also performed. The proportion of primordial follicles was significantly reduced, and the proportion of primary and secondary follicles was significantly increased in all four groups (p < 0.001). Tertiary follicles were observed in the four culture groups. Activin A supplementation did not significantly affect the follicular density, follicular quality, follicular growth, or estradiol secretion (p > 0.05). The 3D-structure increased the density of primary follicles and decreased the estradiol secretion (p < 0.001). Follicular proliferation was significantly lower in the 3D-structure group compared to the non-3D-structure group (p = 0.008). Regarding follicular apoptosis, it was significantly higher in the activin group compared to the non-activin group (p = 0.006). Activin A did not seem to play a key role in the in vitro folliculogenesis activation in our culture conditions. However, the results may indicate that the 3D-structure could be more physiological and could prevent a detrimental in vitro folliculogenesis flare-up.
Collapse
|
6
|
The use of intraovarian injection of autologous platelet rich plasma (PRP) in patients with poor ovarian response and premature ovarian insufficiency. Curr Opin Obstet Gynecol 2022; 34:133-137. [PMID: 35645011 DOI: 10.1097/gco.0000000000000784] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Intraovarian injection of platelet rich plasma (PRP) is a novel treatment for patients with poor ovarian response (POR) and primary ovarian insufficiency (POI). This article reviews the latest literature on the effect of PRP on markers of ovarian reserve, oocyte and embryo yield, and live birth for these poor prognosis patients. RECENT FINDINGS Several case series and one prospective trial have demonstrated improvements in markers of ovarian reserve in patients with POI and POR and improved oocyte and embryo yields in patients with POR. These studies report multiple live births in patients who had previously failed treatment. The positive effects of PRP persist throughout the literature despite the fact that multiple protocols for preparing and injecting PRP exist, with no consensus on the optimal protocol. SUMMARY Intra-ovarian injection of PRP is a promising new technology for poor prognosis patients. Rigorous and appropriately controlled clinical trials are warranted to confirm the utility of this treatment for improving patients' ability to successfully conceive.
Collapse
|
7
|
Liu N, Wang S, Yao Q, Li Y, Hu H, Xiaorong T, Ran H, Price CA, Jiang Z. Activin A attenuates apoptosis of granulosa cells in atretic follicles through ERβ-induced autophagy. Reprod Domest Anim 2022; 57:625-634. [PMID: 35244300 DOI: 10.1111/rda.14103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 02/25/2022] [Indexed: 11/30/2022]
Abstract
It is well known that about 99% of ovarian follicles in mammals suffer from a degenerative process known as atresia, which is a huge waste of genetic resource in female animals. Studies have shown that activin A (ACT-A) is located in ovarian granulosa cells and has different effects in granulosa cell depending on species. Although granulosa cells play a critical role during follicular atresia, the mechanism of action of ACT-A in bovine ovarian granulosa cells (BGC) is poorly understood. In this study, we firstly determined the apoptosis of BGCs isolated from growth follicles and atretic follicles, respectively. Then, BGC isolated from atretic follicles were used as a model to elucidate the role of ACT-A in cattle ovary. The results showed that apoptosis occurred in both growing follicles and atretic follicles, and the percentage of apoptotic cells in atretic follicles was higher than that in growing follicles. The current results indicated that ACT-A can attenuate apoptosis of BGC through maintaining the function of BGC in atretic follicles. Increased ERβ induced by ACT-A promoted BGC autophagy but had no effect on apoptosis. In summary, this study suggests that ACT-A attenuates BGC apoptosis in atretic follicles by ERβ-mediated autophagy signaling.
Collapse
Affiliation(s)
- Ning Liu
- Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, College of Animal Science and Technology, Northwest Agriculture & Forestry University, 712100, Yangling, Shaanxi, China
| | - Shiyou Wang
- Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, College of Animal Science and Technology, Northwest Agriculture & Forestry University, 712100, Yangling, Shaanxi, China
| | - Qichun Yao
- Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, College of Animal Science and Technology, Northwest Agriculture & Forestry University, 712100, Yangling, Shaanxi, China
| | - Yuanyou Li
- Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, College of Animal Science and Technology, Northwest Agriculture & Forestry University, 712100, Yangling, Shaanxi, China
| | - Hai Hu
- Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, College of Animal Science and Technology, Northwest Agriculture & Forestry University, 712100, Yangling, Shaanxi, China
| | - Tang Xiaorong
- Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, College of Animal Science and Technology, Northwest Agriculture & Forestry University, 712100, Yangling, Shaanxi, China
| | - Haohan Ran
- College of Animal Science and Animal Medicine, Tianjin Agricultural University, Jintong Road, Xiqing District, 300380, Tianjin, China
| | - Christopher A Price
- Centre de recherche en reproduction et fertilité, Faculté de médecine vétérinaire, Université de Montréal, St-Hyacinthe, QC, J2S 7C6, Canada
| | - Zhongliang Jiang
- Key Laboratory of Animal Genetic, Breeding and Reproduction in Shaanxi Province, College of Animal Science and Technology, Northwest Agriculture & Forestry University, 712100, Yangling, Shaanxi, China
| |
Collapse
|
8
|
Abstract
In vitro systems capable of reconstituting the process of mouse oogenesis are now being established to help develop further understanding of the mechanisms underlying oocyte/follicle development and differentiation. These systems could also help increase the production of useful livestock or genetically modified animals, and aid in identifying the causes of infertility in humans. Recently, we revealed, using an in vitro system for recapitulating oogenesis, that the activation of the estrogen signaling pathway induces abnormal follicle formation, that blocking estrogen-induced expression of anti-Müllerian hormone is crucial for normal follicle formation, and that the production of α-fetoprotein in fetal liver tissue is involved in normal in vivo follicle formation. In mouse fetuses, follicle formation is not carried out by factors within the ovaries but is instead orchestrated by distal endocrine factors. This review outlines findings from genetics, endocrinology, and in vitro studies regarding the factors that can affect the formation of primordial follicles in mammals.
Collapse
|
9
|
Bao Y, Yao X, Li X, Ei-Samahy MA, Yang H, Liang Y, Liu Z, Wang F. INHBA transfection regulates proliferation, apoptosis and hormone synthesis in sheep granulosa cells. Theriogenology 2021; 175:111-122. [PMID: 34537472 DOI: 10.1016/j.theriogenology.2021.09.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 12/14/2022]
Abstract
Inhibin subunit beta A (INHBA) participates in the synthesis of inhibin A, activin A and activin AB. Here we investigated the effect and molecular mechanism of INHBA on proliferation, apoptosis and hormone synthesis in sheep granulosa cells (GCs) using in vitro transfection. We first noticed that INHBA expression increased with follicle diameter and was widely distributed in ovarian tissue. The proliferation rate of GCs was significantly increased and decreased with overexpression and silence of INHBA, respectively, compared with the negative controls. INHBA transfection affected GC proliferation and apoptosis, regulating the expression of many cell cycle-related and apoptosis-related genes. INHBA overexpression significantly decreased activin and estradiol secretion while increasing inhibin and progesterone secretion. The expression of follicle-stimulating hormone beta subunit was significantly decreased and increased with INHBA overexpression and knockdown, respectively. Notably, silence of INHBA inhibited the expression of many transforming growth factor beta-related genes. Overall, the functional molecule of INHBA gene may be associated with follicular development via regulating proliferation, apoptosis and folliculogenesis-related hormone secretion of sheep GCs. In addition, our findings may contribute to a better understanding of the law of follicular development and thus improve the reproductive performance of female animals.
Collapse
Affiliation(s)
- Yongjin Bao
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaolei Yao
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xiaodan Li
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, 210095, China
| | - M A Ei-Samahy
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, 210095, China
| | - Hua Yang
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yaxu Liang
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zifei Liu
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, 210095, China
| | - Feng Wang
- Hu Sheep Academy, Nanjing Agricultural University, Nanjing, 210095, China; Jiangsu Livestock Embryo Engineering Laboratory, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
10
|
Ma L, Li C, Lian S, Xu B, Yuan J, Lu J, Yang H, Guo J, Ji H. ActivinA activates Notch1-Shh signaling to regulate proliferation in C2C12 skeletal muscle cells. Mol Cell Endocrinol 2021; 519:111055. [PMID: 33045308 DOI: 10.1016/j.mce.2020.111055] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/30/2022]
Abstract
The myoblast is a precursor cell that rebuilds muscle tissue after trauma in human and animal skeletal muscle tissue. Proliferation of myoblasts is important for skeletal muscle damage repair and is controlled by numerous transcription factors and signals. The regulation of these signaling pathways and their complex interactions are not fully understood. This study aims to determine the physiological functions of Activin A, Notch and Sonic Hedgehog (Shh) signaling in the proliferation of mouse C2C12 myoblasts and to explore their interactions. Activin A facilitated proliferation of C2C12 cells and promoted the conversion of G1 into S phase in cell cycle, whereas addition of the receptor inhibitor SB431542 attenuated the proliferation activity of rActA on C2C12 cells. Activin A also activated Notch and Shh signaling, while blockage of these pathways attenuated the function of Activin A in cell cycle. Inhibition of the Notch signaling by Notch response inhibitor DAPT significantly down-regulated the expression of Shh signaling molecules, whereas exogenous rShh reversed the inhibition of C2C12 cells proliferative activity induced by DAPT, indicating Notch signaling act upstream of the Shh pathway. Furthermore, inhibition of Notch signaling weakened the activation of Activin A-mediated Shh signaling. Taken together, our results provide a novel role of Activin A in regulating the proliferation of C2C12 skeletal muscle cells, which impacts ActivinA-Notch1-Shh signaling pathways.
Collapse
Affiliation(s)
- Li Ma
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Chengxu Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shuai Lian
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Bin Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jianbin Yuan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jingjing Lu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Huanmin Yang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jingru Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hong Ji
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China.
| |
Collapse
|
11
|
Wasti S, Sah N, Kuehu DL, Kim YS, Jha R, Mishra B. Expression of follistatin is associated with egg formation in the oviduct of laying hens. Anim Sci J 2020; 91:e13396. [PMID: 32468659 DOI: 10.1111/asj.13396] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 03/20/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022]
Abstract
The objective of this study was to examine the expression profiles of follistatin (FST) and its associated molecules (MSTN, INHA, INHBB, INHBA, ACVR2A, and ACVR2B) in the oviduct of laying hens at 3 hr and 20 hr post-ovulation (p.o., n = 5; 35 weeks old), molting (n = 5; 60 weeks old), and non-laying (n = 4; 35-60 weeks old) hens and also to localize the FST by using immunohistochemistry assay. Expression of FST was significantly higher (p < .05), and MSTN was lower in the uterus of laying hens around 15-20 hr p.o. (during eggshell formation), however, their expressions in the magnum remain unchanged across different physiological stages of hens. FST was mainly expressed in the luminal and glandular epithelium of the uterine tissues, and their expression intensity was highest in laying hens during the eggshell mineralization. There was a relatively increased expression of INHA in the magnum of laying hens around 3 hr p.o. as compared to non-laying and molting hens. At the same time (3 hr p.o.), there was a significant (p < .05) decrease in the expression of the INHBB, ACVR2A, and ACV2B. These results indicate that follistatin may regulate the differentiation of uterine luminal and glandular epithelium during eggshell biomineralization.
Collapse
Affiliation(s)
- Sanjeev Wasti
- Department of Human Nutrition Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Nirvay Sah
- Department of Human Nutrition Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Donna L Kuehu
- Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Yong S Kim
- Department of Human Nutrition Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Rajesh Jha
- Department of Human Nutrition Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Birendra Mishra
- Department of Human Nutrition Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, HI, USA
| |
Collapse
|
12
|
Appiah Adu-Gyamfi E, Tanam Djankpa F, Nelson W, Czika A, Kumar Sah S, Lamptey J, Ding YB, Wang YX. Activin and inhibin signaling: From regulation of physiology to involvement in the pathology of the female reproductive system. Cytokine 2020; 133:155105. [PMID: 32438278 DOI: 10.1016/j.cyto.2020.155105] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/14/2020] [Indexed: 12/17/2022]
Abstract
Activins and inhibins - comprising activin A, B, AB, C and E, and inhibin A and B isoforms - belong to the transforming growth factor beta (TGFβ) superfamily. They regulate several biological processes, including cellular proliferation, differentiation and invasiveness, to enhance the formation and functioning of many human tissues and organs. In this review, we have discussed the role of activin and inhibin signaling in the physiological and female-specific pathological events that occur in the female reproductive system. The up-to-date evidence indicates that these cytokines regulate germ cell development, follicular development, ovulation, uterine receptivity, decidualization and placentation through the activation of several signaling pathways; and that their dysregulated expression is involved in the pathogenesis and pathophysiology of the numerous diseases, including pregnancy complications, that disturb reproduction. Hence, some of the isoforms have been suggested as potential biomarkers and therapeutic targets for the management of some of these diseases. Tackling the research directions highlighted in this review will enhance a detailed comprehension and the clinical utility of these cytokines.
Collapse
Affiliation(s)
- Enoch Appiah Adu-Gyamfi
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Francis Tanam Djankpa
- Department of Physiology, School of Medical Sciences, University of Cape Coast, Cape Coast, Ghana.
| | - William Nelson
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, People's Republic of China; Department of Environmental and Occupational Health, School of Public Health and Social Sciences, Muhimbili University of Health and Allied Sciences, Dar es salaam, Tanzania.
| | - Armin Czika
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Sanjay Kumar Sah
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Jones Lamptey
- Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, People's Republic of China; Kumasi Centre for Collaborative Research in Tropical Medicine, KCCR, Ghana.
| | - Yu-Bin Ding
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Ying-Xiong Wang
- Department of Reproductive Sciences, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China; Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
13
|
Kanduc D, Shoenfeld Y. Human Papillomavirus Epitope Mimicry and Autoimmunity: The Molecular Truth of Peptide Sharing. Pathobiology 2019; 86:285-295. [PMID: 31593963 DOI: 10.1159/000502889] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Accepted: 08/22/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To define the cross-reactivity potential and the consequent autoimmunity intrinsic to viral versus human peptide sharing. METHODS Using human papillomavirus (HPV) infection/active immunization as a research model, the experimentally validated HPV L1 epitopes catalogued at the Immune Epitope DataBase were analyzed for peptide sharing with the human proteome. RESULTS The final data show that the totality of the immunoreactive HPV L1 epi-topes is mostly composed by peptides present in human proteins. CONCLUSIONS Immunologically, the high extent of peptide sharing between the HPV L1 epitopes and human proteins invites to revise the concept of the negative selection of self-reactive lymphocytes. Pathologically, the data highlight a cross-reactive potential for a spectrum of autoimmune diseases that includes ovarian failure, systemic lupus erythematosus (SLE), breast cancer and sudden death, among others. Therapeutically, analyzing already validated immunoreactive epitopes filters out the peptide sharing possibly exempt of self-reactivity, defines the effective potential for pathologic autoimmunity, and allows singling out peptide epitopes for safe immunotherapeutic protocols.
Collapse
Affiliation(s)
- Darja Kanduc
- Department of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Bari, Italy,
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Affiliated to Tel-Aviv, University School of Medicine, Ramat Gan, Israel.,I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian, Federation, Sechenov University, Moscow, Russian Federation
| |
Collapse
|
14
|
Reader KL, Marino FE, Nicholson HD, Risbridger GP, Gold EJ. Role of activin C in normal ovaries and granulosa cell tumours of mice and humans. Reprod Fertil Dev 2019; 30:958-968. [PMID: 29207252 DOI: 10.1071/rd17250] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 11/08/2017] [Indexed: 12/30/2022] Open
Abstract
Activins and inhibins play important roles in the development, growth and function of the ovary. Mice lacking inhibin develop granulosa cell tumours in their ovaries that secrete activin A, and these tumours are modulated by increased activin C expression. The aim of the present study was to identify where activin C is expressed in mouse and human ovaries and whether overexpression of activin C modulates normal follicular development in mice. Immunohistochemical staining for the activin βC subunit was performed on sections from mouse and human ovaries and human adult granulosa cell tumours. Stereology techniques were used to quantify oocyte and follicular diameters, and the percentage of different follicular types in ovaries from wild-type mice and those underexpressing inhibin α and/or overexpressing activin C. Staining for activin βC was observed in the oocytes, granulosa cells, thecal cells and surface epithelium of mouse and human ovaries, and in the granulosa-like cells of adult granulosa cell tumours. Overexpression of activin C in mice did not alter follicular development compared with wild-type mice, but it did modulate the development of abnormal early stage follicles in inhibin α-null mice. These results provide further evidence of a role for activin C in the ovary.
Collapse
Affiliation(s)
- Karen L Reader
- Department of Anatomy, University of Otago, Dunedin 9054, New Zealand
| | | | - Helen D Nicholson
- Department of Anatomy, University of Otago, Dunedin 9054, New Zealand
| | - Gail P Risbridger
- Consortium and Cancer Program Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Vic. 3800, Australia
| | - Elspeth J Gold
- Department of Anatomy, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
15
|
Chu YL, Xu YR, Yang WX, Sun Y. The role of FSH and TGF-β superfamily in follicle atresia. Aging (Albany NY) 2019; 10:305-321. [PMID: 29500332 PMCID: PMC5892684 DOI: 10.18632/aging.101391] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/23/2018] [Indexed: 01/02/2023]
Abstract
Most of the mammalian follicles undergo a degenerative process called “follicle atresia”. Apoptosis of granulosa cells is the main characteristic of follicle atresia. Follicle stimulating hormone (FSH) and the transforming growth factor β (TGF-β) superfamily have important regulatory functions in this process. FSH activates protein kinase A and cooperating with insulin receptor substrates, it promotes the PI3K/Akt pathway which weakens apoptosis. Both Smad or non-Smad signaling of the transforming growth factor β superfamily seem to be related to follicle atresia, and the effect of several important family members on follicle atresia is concluded in this article. FSH and TGF-β are likely to mutually influence each other and what we have already known about the possible underlying molecular mechanism is also discussed below.
Collapse
Affiliation(s)
- Yu-Lan Chu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ya-Ru Xu
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wan-Xi Yang
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yi Sun
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
16
|
Nagashima JB, Wildt DE, Travis AJ, Songsasen N. Activin promotes growth and antral cavity expansion in the dog ovarian follicle. Theriogenology 2019; 129:168-177. [PMID: 30856402 PMCID: PMC6445547 DOI: 10.1016/j.theriogenology.2019.02.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/29/2019] [Accepted: 02/21/2019] [Indexed: 01/16/2023]
Abstract
Understanding regulators of folliculogenesis remains limited in the domestic dog (Canis familiaris), which challenges our ability to develop in vitro follicle culture systems for canid genome rescue efforts. Here, we investigated the influence of activin on dog follicle development and survival, oocyte quality, and FSH receptor expression in culture. Preantral (150 - ≤230 μm diameter), early antral (231 - ≤330 μm), and antral (>330-550 μm) stage follicles were encapsulated in a fibrin-alginate hydrogel with 0, 100, or 200 ng/ml rhActivin plus 0, 0.1, 1, or 10 μg/ml FSH for 12 or 21 d of in vitro culture. All follicle groups increased in diameter (P < 0.05) with activin acting synergistically with FSH to improve (P < 0.05) growth and antral cavity expansion (to >630 μm) in early antral and antral cohorts. This complementary effect was not linked to changes in FSHR mRNA expression (P > 0.05). Although not influencing (P > 0.05) follicle survival or transzonal projection (TZP) density in shorter term 12 d culture, activin in the presence of 1 ng/ml FSH maintained TZP density from the 12-21 d interval. Activin also increased oocyte diameter and improved nuclear integrity compared to un-supplemented controls. These results indicate that activin acts synergistically with FSH to promote growth and antral cavity expansion of the dog follicle in vitro, information useful to formulating an effective culture microenvironment for this species.
Collapse
Affiliation(s)
- Jennifer B Nagashima
- Smithsonian Conservation Biology Institute, National Zoological Park, Front Royal, VA, 22630, USA; Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| | - David E Wildt
- Smithsonian Conservation Biology Institute, National Zoological Park, Front Royal, VA, 22630, USA
| | - Alexander J Travis
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA; Atkinson Center for a Sustainable Future, Cornell University, Ithaca, NY, 14853, USA
| | - Nucharin Songsasen
- Smithsonian Conservation Biology Institute, National Zoological Park, Front Royal, VA, 22630, USA
| |
Collapse
|
17
|
Hummitzsch K, Hatzirodos N, Irving-Rodgers HF, Hartanti MD, Perry VEA, Anderson RA, Rodgers RJ. Morphometric analyses and gene expression related to germ cells, gonadal ridge epithelial-like cells and granulosa cells during development of the bovine fetal ovary. PLoS One 2019; 14:e0214130. [PMID: 30901367 PMCID: PMC6430378 DOI: 10.1371/journal.pone.0214130] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 03/07/2019] [Indexed: 12/24/2022] Open
Abstract
Cells on the surface of the mesonephros give rise to replicating Gonadal Ridge Epithelial-Like (GREL) cells, the first somatic cells of the gonadal ridge. Later germ cells associate with the GREL cells in the ovigerous cords, and the GREL cells subsequently give rise to the granulosa cells in follicles. To examine these events further, 27 bovine fetal ovaries of different gestational ages were collected and prepared for immunohistochemical localisation of collagen type I and Ki67 to identify regions of the ovary and cell proliferation, respectively. The non-stromal cortical areas (collagen-negative) containing GREL cells and germ cells and later in development, the follicles with oocytes and granulosa cells, were analysed morphometrically. Another set of ovaries (n = 17) were collected and the expression of genes associated with germ cell lineages and GREL/granulosa cells were quantitated by RT-PCR. The total volume of non-stromal areas in the cortex increased significantly and progressively with ovarian development, plateauing at the time the surface epithelium developed. However, the proportion of non-stromal areas in the cortex declined significantly and progressively throughout gestation, largely due to a cessation in growth of the non-stroma cells and the continued growth of stroma. The proliferation index in the non-stromal area was very high initially and then declined substantially at the time follicles formed. Thereafter, it remained low. The numerical density of the non-stromal cells was relatively constant throughout ovarian development. The expression levels of a number of genes across gestation either increased (AMH, FSHR, ESR1, INHBA), declined (CYP19A1, ESR2, ALDH1A1, DSG2, OCT4, LGR5) or showed no particular pattern (CCND2, CTNNB1, DAZL, FOXL2, GATA4, IGFBP3, KRT19, NR5A1, RARRES1, VASA, WNT2B). Many of the genes whose expression changed across gestation, were positively or negatively correlated with each other. The relationships between these genes may reflect their roles in the important events such as the transition of ovigerous cords to follicles, oogonia to oocytes or GREL cells to granulosa cells.
Collapse
Affiliation(s)
- Katja Hummitzsch
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Nicholas Hatzirodos
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Helen F. Irving-Rodgers
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- School of Medical Science, Griffith University, Gold Coast Campus, Gold Coast, Queensland, Australia
| | - Monica D. Hartanti
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Viv E. A. Perry
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington, Leicestershire, United Kingdom
| | - Richard A. Anderson
- Medical Research Council Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Raymond J. Rodgers
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
- * E-mail:
| |
Collapse
|
18
|
Wang JJ, Zhai QY, Zhang RQ, Ge W, Liu JC, Li L, Sun ZY, De Felici M, Shen W. Effects of activin A on the transcriptome of mouse oogenesis in vitro. J Cell Physiol 2019; 234:14339-14350. [PMID: 30633354 DOI: 10.1002/jcp.28135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 12/18/2018] [Indexed: 01/01/2023]
Abstract
From the previous research, it has been supported that activin A (ActA) is conducive to ovarian development in vitro. In the present paper, with the aim to identify the molecular pathways through which ActA can influence processes of the fetal and early postnatal oogenesis, we analyzed the transcriptome of embryonic ovaries (12.5 days postcoitum) in vitro cultured with or without ActA for 6 days, as well as the produced oocytes for 28 days, and further compared the gene expression profile with their in vivo counterparts. With the confirmation of designed test, we found that the addition of ActA to the ovary culture tended, generally, to align oocyte gene expression to the in vivo condition, in particular of a number of genes involved in meiosis and epigenetic modifications of histones. In particular, we identified DNA recombination during the oocyte meiotic prophase I and lysine trimethylation of the histone H3K27 during the oocyte growth phase as molecular pathways modulated by ActA.
Collapse
Affiliation(s)
- Jun-Jie Wang
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, China
| | - Qiu-Yue Zhai
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, China
| | - Rui-Qian Zhang
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, China
| | - Wei Ge
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, China
| | - Jing-Cai Liu
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, China
| | - Lan Li
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, China
| | - Zhong-Yi Sun
- Center for Reproductive Medicine, Urology Department, Peking University Shenzhen Hospital, Shenzhen, China
| | - Massimo De Felici
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Wei Shen
- College of Life Sciences, Institute of Reproductive Sciences, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
19
|
Bloise E, Ciarmela P, Dela Cruz C, Luisi S, Petraglia F, Reis FM. Activin A in Mammalian Physiology. Physiol Rev 2019; 99:739-780. [DOI: 10.1152/physrev.00002.2018] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Activins are dimeric glycoproteins belonging to the transforming growth factor beta superfamily and resulting from the assembly of two beta subunits, which may also be combined with alpha subunits to form inhibins. Activins were discovered in 1986 following the isolation of inhibins from porcine follicular fluid, and were characterized as ovarian hormones that stimulate follicle stimulating hormone (FSH) release by the pituitary gland. In particular, activin A was shown to be the isoform of greater physiological importance in humans. The current understanding of activin A surpasses the reproductive system and allows its classification as a hormone, a growth factor, and a cytokine. In more than 30 yr of intense research, activin A was localized in female and male reproductive organs but also in other organs and systems as diverse as the brain, liver, lung, bone, and gut. Moreover, its roles include embryonic differentiation, trophoblast invasion of the uterine wall in early pregnancy, and fetal/neonate brain protection in hypoxic conditions. It is now recognized that activin A overexpression may be either cytostatic or mitogenic, depending on the cell type, with important implications for tumor biology. Activin A also regulates bone formation and regeneration, enhances joint inflammation in rheumatoid arthritis, and triggers pathogenic mechanisms in the respiratory system. In this 30-yr review, we analyze the evidence for physiological roles of activin A and the potential use of activin agonists and antagonists as therapeutic agents.
Collapse
Affiliation(s)
- Enrrico Bloise
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| | - Pasquapina Ciarmela
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| | - Cynthia Dela Cruz
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| | - Stefano Luisi
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| | - Felice Petraglia
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| | - Fernando M. Reis
- Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Obstetrics and Gynecology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Department of Molecular and Developmental Medicine, Obstetrics and Gynecological Clinic, University of Siena, Siena, Italy; and Department of Biomedical, Experimental and Clinical Sciences, Division of Obstetrics and
| |
Collapse
|
20
|
|
21
|
Jørgensen A, Macdonald J, Nielsen JE, Kilcoyne KR, Perlman S, Lundvall L, Langhoff Thuesen L, Juul Hare K, Frederiksen H, Andersson AM, Skakkebæk NE, Juul A, Sharpe RM, Rajpert-De Meyts E, Mitchell RT. Nodal Signaling Regulates Germ Cell Development and Establishment of Seminiferous Cords in the Human Fetal Testis. Cell Rep 2018; 25:1924-1937.e4. [PMID: 30428358 DOI: 10.1016/j.celrep.2018.10.064] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/14/2018] [Accepted: 10/17/2018] [Indexed: 01/26/2023] Open
Abstract
Disruption of human fetal testis development is widely accepted to underlie testicular germ cell cancer (TGCC) origin and additional disorders within testicular dysgenesis syndrome (TDS). However, the mechanisms for the development of testicular dysgenesis in humans are unclear. We used ex vivo culture and xenograft approaches to investigate the importance of Nodal and Activin signaling in human fetal testis development. Inhibition of Nodal, and to some extent Activin, signaling disrupted seminiferous cord formation, abolished AMH expression, reduced androgen secretion, and decreased gonocyte numbers. Subsequent xenografting of testicular tissue rescued the disruptive effects on seminiferous cords and somatic cells but not germ cell effects. Stimulation of Nodal signaling increased the number of germ cells expressing pluripotency factors, and these persisted after xenografting. Our findings suggest a key role for Nodal signaling in the regulation of gonocyte differentiation and early human testis development with implications for the understanding of TGCC and TDS origin.
Collapse
Affiliation(s)
- Anne Jørgensen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark.
| | - Joni Macdonald
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - John E Nielsen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Karen R Kilcoyne
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Signe Perlman
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Lene Lundvall
- Department of Gynaecology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Lea Langhoff Thuesen
- Department of Obstetrics and Gynaecology, Hvidovre University Hospital, Kettegård Alle 30, Hvidovre, Denmark
| | - Kristine Juul Hare
- Department of Obstetrics and Gynaecology, Hvidovre University Hospital, Kettegård Alle 30, Hvidovre, Denmark
| | - Hanne Frederiksen
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Anna-Maria Andersson
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Niels E Skakkebæk
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Anders Juul
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Richard M Sharpe
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Ewa Rajpert-De Meyts
- Department of Growth and Reproduction, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100 Copenhagen, Denmark; International Research and Research Training Centre in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Blegdamsvej 9, 2100 Copenhagen, Denmark
| | - Rod T Mitchell
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK
| |
Collapse
|
22
|
Turathum B, Roytrakul S, Changsangfa C, Sroyraya M, Tanasawet S, Kitiyanant Y, Saikhun K. Missing and overexpressing proteins in domestic cat oocytes following vitrification and in vitro maturation as revealed by proteomic analysis. Biol Res 2018; 51:27. [PMID: 30124164 PMCID: PMC6100730 DOI: 10.1186/s40659-018-0176-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/12/2018] [Indexed: 12/18/2022] Open
Abstract
Background The domestic cat serves as an animal model for assisted reproductive studies of endangered felid species. To date, there are no data on the protein alterations following cryopreservation of oocytes in felid family. Methods Immature (germinal vesicle) domestic cat oocytes were vitrified in the vitrification solution containing 35% ethylene glycol, 20% DMSO and 0.5 mM sucrose. The vitrified-warmed oocytes were matured (metaphase II) in vitro and subjected to proteomic analysis using 1DE SDS-PAGE prefractionation combined with LC–MS/MS. Results A total of 1712 proteins were identified in in vitro matured oocytes. Of the 1712 proteins, 1454 proteins were found in both groups, whereas, 258 proteins were differentially expressed between control and vitrified-warmed groups. In vitrified-warmed oocytes, the missing proteins were membrane and nuclear proteins; whereas, apoptosis and DNA repair proteins were overrepresented. Conclusions The identified missing and overexpressed proteins in vitrified-warmed oocytes represent potential markers of cryoinjuries and the developmental pathways of oocytes. The findings of differential expressed proteins may contribute to effective ways of proteome analysis of oocyte/embryo quality in order to assess safety of cryopreservation in felid species.
Collapse
Affiliation(s)
- Bongkoch Turathum
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, 12121, Thailand
| | - Chinarat Changsangfa
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand
| | - Morakot Sroyraya
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Supita Tanasawet
- Department of Anatomy, Faculty of Science, Prince of Songkla University, Hat Yai, 90110, Thailand
| | - Yindee Kitiyanant
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | - Kulnasan Saikhun
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, 73170, Thailand.
| |
Collapse
|
23
|
Wang C, Zhou B, Xia G. Mechanisms controlling germline cyst breakdown and primordial follicle formation. Cell Mol Life Sci 2017; 74:2547-2566. [PMID: 28197668 PMCID: PMC11107689 DOI: 10.1007/s00018-017-2480-6] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 12/11/2022]
Abstract
In fetal females, oogonia proliferate immediately after sex determination. The progress of mitosis in oogonia proceeds so rapidly that the incompletely divided cytoplasm of the sister cells forms cysts. The oogonia will then initiate meiosis and arrest at the diplotene stage of meiosis I, becoming oocytes. Within each germline cyst, oocytes with Balbiani bodies will survive after cyst breakdown (CBD). After CBD, each oocyte is enclosed by pre-granulosa cells to form a primordial follicle (PF). Notably, the PF pool formed perinatally will be the sole lifelong oocyte source of a female. Thus, elucidating the mechanisms of CBD and PF formation is not only meaningful for solving mysteries related to ovarian development but also contributes to the preservation of reproduction. However, the mechanisms that regulate these phenomena are largely unknown. This review summarizes the progress of cellular and molecular research on these processes in mice and humans.
Collapse
Affiliation(s)
- Chao Wang
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Bo Zhou
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China
| | - Guoliang Xia
- State Key Laboratory for Agro-Biotechnology, College of Biological Science, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
24
|
Kawashima I, Kawamura K. Disorganization of the germ cell pool leads to primary ovarian insufficiency. Reproduction 2017; 153:R205-R213. [PMID: 28289071 DOI: 10.1530/rep-17-0015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 02/10/2017] [Accepted: 03/13/2017] [Indexed: 01/26/2023]
Abstract
The mammalian ovary is an organ that controls female germ cell development, storing them and releasing mature oocytes for transporting to the oviduct. During the fetal stage, female germ cells change from a proliferative state to meiosis before forming follicles with the potential for the growth of surrounding somatic cells. Understanding of molecular and physiological bases of germ cell development in the fetal ovary contributed not only to the elucidation of genetic disorders in primary ovarian insufficiency (POI), but also to the advancement of novel treatments for patients with POI. Accumulating evidence indicates that mutations in NOBOX, DAZL and FIGLAgenes are associated with POI. In addition, cell biology studies revealed the important roles of these genes as essential translational factors for germ cell development. Recent insights into the role of the PI3K (phosphatidylinositol 3-kinase)-Akt signaling pathway in primordial follicle activation allowed the development of a new infertility treatment, IVA (in vitro activation), leading to successful pregnancy/delivery in POI patients. Furthermore, elucidation of genetic dynamics underlying female germ cell development could allow regeneration of oocytes from ES (embryonic stem)/iPS (induced pluripotent stem) cells in mammals. The purpose of this review is to summarize basic findings related to female germ cell development and potential clinical implications, especially focusing on POI etiologies. We also summarize evolving new POI therapies based on IVA as well as oocyte regeneration.
Collapse
Affiliation(s)
- Ikko Kawashima
- Department of Advanced Reproductive MedicineSt. Marianna University School of Medicine, Kawasaki City, Kanagawa, Japan
| | - Kazuhiro Kawamura
- Department of Advanced Reproductive MedicineSt. Marianna University School of Medicine, Kawasaki City, Kanagawa, Japan
| |
Collapse
|
25
|
Shi L, Resaul J, Owen S, Ye L, Jiang WG. Clinical and Therapeutic Implications of Follistatin in Solid Tumours. Cancer Genomics Proteomics 2017; 13:425-435. [PMID: 27807065 DOI: 10.21873/cgp.20005] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 10/05/2016] [Indexed: 12/20/2022] Open
Abstract
Follistatin (FST), as a single-chain glycosylated protein, has two major isoforms, FST288 and FST315. The FST315 isoform is the predominant form whilst the FST288 variant accounts for less than 5% of the encoded mRNA. FST is differentially expressed in human tissues and aberrant expression has been observed in a variety of solid tumours, including gonadal, gastric and lung cancer, hepatocellular carcinoma, basal cell carcinoma and melanoma. Based on the current evidence, FST is an antagonist of transforming growth factor beta family members, such as activin and bone morphogenetic proteins (BMPs). FST plays a role in tumourigenesis, metastasis and angiogenesis of solid tumours through its interaction with activin and BMPs, thus resulting in pathophysiological function. In terms of diagnosis, prognosis and therapy, FST has shown strong promise. Through a better understanding of its biological functions, potential clinical applications may yet emerge.
Collapse
Affiliation(s)
- Lei Shi
- Urology Department, Yantai Yu Huang Ding Hospital, Yantai, Shandong Province, P.R. China.,Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Jeyna Resaul
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Sioned Owen
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, U.K.
| |
Collapse
|
26
|
Sun R, Sun YC, Ge W, Tan H, Cheng SF, Yin S, Sun XF, Li L, Dyce P, Li J, Yang X, Shi QH, Shen W. The crucial role of Activin A on the formation of primordial germ cell-like cells from skin-derived stem cells in vitro. Cell Cycle 2016; 14:3016-29. [PMID: 26406115 DOI: 10.1080/15384101.2015.1078031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Primordial germ cells (PGCs) are founder cells of the germ cell lineage, and can be differentiated from stem cells in an induced system in vitro. However, the induction conditions need to be optimized in order to improve the differentiation efficiency. Activin A (ActA) is a member of the TGF-β super family and plays an important role in oogenesis and folliculogenesis. In the present study, we found that ActA promoted PGC-like cells (PGCLCs) formation from mouse skin-derived stem cells (SDSCs) in both embryoid body-like structure (EBLS) differentiation and the co-culture stage in a dose dependent manner. ActA treatment (100 ng/ml) during EBLS differentiation stage and further co-cultured for 6 days without ActA significantly increased PGCLCs from 53.2% to 82.8%, and as well as EBLS differentiation without ActA followed by co-cultured with 100 ng/ml ActA for 4 to 12 days with the percentage of PGCLCs increasing markedly in vitro. Moreover, mice treated with ActA at 100 ng/kg body weight from embryonic day (E) 5.5-12.5 led to more PGCs formation. However, the stimulating effects of ActA were interrupted by Smad3 RNAi, and in an in vitro cultured Smad3(-/-) mouse skin cells scenario. SMAD3 is thus likely a key effecter molecule in the ActA signaling pathway. In addition, we found that the expression of some epiblast cell markers, Fgf5, Dnmt3a, Dnmt3b and Wnt3, was increased in EBLSs cultured for 4 days or PGCLCs co-cultured for 12 days with ActA treatment. Interestingly, at 16 days of differentiation, the percentage of PGCLCs was decreased in the presence of ActA, but the expression of meiosis-relative genes, such as Stra8, Dmc1, Sycp3 and Sycp1, was increased. In conclusion, our data here demonstrated that ActA can promote PGCLC formation from SDSCs in vitro, at early stages of differentiation, and affect meiotic initiation of PGCLCs in later stages.
Collapse
Affiliation(s)
- Rui Sun
- a 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 , Anhui , China
| | - Yuan-Chao Sun
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Wei Ge
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Hui Tan
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Shun-Feng Cheng
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Shen Yin
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Xiao-Feng Sun
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Lan Li
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| | - Paul Dyce
- d Department of Animal and Poultry Science ; University of Guelph ; Guelph ; Ontario , Canada
| | - Julang Li
- d Department of Animal and Poultry Science ; University of Guelph ; Guelph ; Ontario , Canada
| | - Xiao Yang
- e Genetic Laboratory of Development and Diseases; Beijing Institute of Biotechnology ; Beijing , China
| | - Qing-Hua Shi
- a 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 , Anhui , China.,f Collaborative Innovation Center of Genetics and Development; Fudan University ; Shanghai , China
| | - Wei Shen
- b Institute of Reproductive Sciences; Qingdao Agricultural University , Qingdao ; Shandong , China.,c Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong; College of Animal Science and Technology; Qingdao Agricultural University , Qingdao ; Shandong , China
| |
Collapse
|
27
|
Wijayarathna R, de Kretser DM. Activins in reproductive biology and beyond. Hum Reprod Update 2016; 22:342-57. [PMID: 26884470 DOI: 10.1093/humupd/dmv058] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/20/2015] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Activins are members of the pleiotrophic family of the transforming growth factor-beta (TGF-β) superfamily of cytokines, initially isolated for their capacity to induce the release of FSH from pituitary extracts. Subsequent research has demonstrated that activins are involved in multiple biological functions including the control of inflammation, fibrosis, developmental biology and tumourigenesis. This review summarizes the current knowledge on the roles of activin in reproductive and developmental biology. It also discusses interesting advances in the field of modulating the bioactivity of activins as a therapeutic target, which would undoubtedly be beneficial for patients with reproductive pathology. METHODS A comprehensive literature search was carried out using PUBMED and Google Scholar databases to identify studies in the English language which have contributed to the advancement of the field of activin biology, since its initial isolation in 1987 until July 2015. 'Activin', 'testis', 'ovary', 'embryonic development' and 'therapeutic targets' were used as the keywords in combination with other search phrases relevant to the topic of activin biology. RESULTS Activins, which are dimers of inhibin β subunits, act via a classical TGF-β signalling pathway. The bioactivity of activin is regulated by two endogenous inhibitors, inhibin and follistatin. Activin is a major regulator of testicular and ovarian development. In the ovary, activin A promotes oocyte maturation and regulates granulosa cell steroidogenesis. It is also essential in endometrial repair following menstruation, decidualization and maintaining pregnancy. Dysregulation of the activin-follistatin-inhibin system leads to disorders of female reproduction and pregnancy, including polycystic ovary syndrome, ectopic pregnancy, miscarriage, fetal growth restriction, gestational diabetes, pre-eclampsia and pre-term birth. Moreover, a rise in serum activin A, accompanied by elevated FSH, is characteristic of female reproductive aging. In the male, activin A is an autocrine and paracrine modulator of germ cell development and Sertoli cell proliferation. Disruption of normal activin signalling is characteristic of many tumours affecting reproductive organs, including endometrial carcinoma, cervical cancer, testicular and ovarian cancer as well as prostate cancer. While activin A and B aid the progression of many tumours of the reproductive organs, activin C acts as a tumour suppressor. Activins are important in embryonic induction, morphogenesis of branched glandular organs, development of limbs and nervous system, craniofacial and dental development and morphogenesis of the Wolffian duct. CONCLUSIONS The field of activin biology has advanced considerably since its initial discovery as an FSH stimulating agent. Now, activin is well known as a growth factor and cytokine that regulates many aspects of reproductive biology, developmental biology and also inflammation and immunological mechanisms. Current research provides evidence for novel roles of activins in maintaining the structure and function of reproductive and other organ systems. The fact that activin A is elevated both locally as well as systemically in major disorders of the reproductive system makes it an important biomarker. Given the established role of activin A as a pro-inflammatory and pro-fibrotic agent, studies of its involvement in disorders of reproduction resulting from these processes should be examined. Follistatin, as a key regulator of the biological actions of activin, should be evaluated as a therapeutic agent in conditions where activin A overexpression is established as a contributing factor.
Collapse
Affiliation(s)
- R Wijayarathna
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia Centre for Reproductive Health, Hudson Institute of Medical Research, 27-31, Wright Street, Clayton, VIC 3168, Australia
| | - D M de Kretser
- Department of Anatomy and Developmental Biology, Monash University, Wellington Road, Clayton, VIC 3800, Australia Centre for Reproductive Health, Hudson Institute of Medical Research, 27-31, Wright Street, Clayton, VIC 3168, Australia
| |
Collapse
|
28
|
Jung D, Kee K. Insights into female germ cell biology: from in vivo development to in vitro derivations. Asian J Androl 2016; 17:415-20. [PMID: 25652637 PMCID: PMC4430939 DOI: 10.4103/1008-682x.148077] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Understanding the mechanisms of human germ cell biology is important for developing infertility treatments. However, little is known about the mechanisms that regulate human gametogenesis due to the difficulties in collecting samples, especially germ cells during fetal development. In contrast to the mitotic arrest of spermatogonia stem cells in the fetal testis, female germ cells proceed into meiosis and began folliculogenesis in fetal ovaries. Regulations of these developmental events, including the initiation of meiosis and the endowment of primordial follicles, remain an enigma. Studying the molecular mechanisms of female germ cell biology in the human ovary has been mostly limited to spatiotemporal characterizations of genes or proteins. Recent efforts in utilizing in vitro differentiation system of stem cells to derive germ cells have allowed researchers to begin studying molecular mechanisms during human germ cell development. Meanwhile, the possibility of isolating female germline stem cells in adult ovaries also excites researchers and generates many debates. This review will mainly focus on presenting and discussing recent in vivo and in vitro studies on female germ cell biology in human. The topics will highlight the progress made in understanding the three main stages of germ cell developments: namely, primordial germ cell formation, meiotic initiation, and folliculogenesis.
Collapse
Affiliation(s)
| | - Kehkooi Kee
- Department of Basic Medical Sciences, Center for Stem Cell Biology and Regenerative Medicine, School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
29
|
Reader KL, Gold E. Activins and activin antagonists in the human ovary and ovarian cancer. Mol Cell Endocrinol 2015; 415:126-32. [PMID: 26277402 DOI: 10.1016/j.mce.2015.08.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/06/2015] [Accepted: 08/09/2015] [Indexed: 12/22/2022]
Abstract
Activins are members of the transforming growth factor β superfamily that play an important role in controlling cell proliferation and differentiation in many organs including the ovary. It is essential that activin signalling be tightly regulated as imbalances can lead to uncontrolled cell proliferation and cancer. This review describes the expression and function of the activins and their known antagonists in both normal and cancerous human ovaries.
Collapse
Affiliation(s)
- Karen L Reader
- Department of Anatomy, University of Otago, PO Box 913, Dunedin 9054, New Zealand.
| | - Elspeth Gold
- Department of Anatomy, University of Otago, PO Box 913, Dunedin 9054, New Zealand
| |
Collapse
|
30
|
Jørgensen A, Lindhardt Johansen M, Juul A, Skakkebaek NE, Main KM, Rajpert-De Meyts E. Pathogenesis of germ cell neoplasia in testicular dysgenesis and disorders of sex development. Semin Cell Dev Biol 2015; 45:124-37. [PMID: 26410164 DOI: 10.1016/j.semcdb.2015.09.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 09/21/2015] [Indexed: 12/29/2022]
Abstract
Development of human gonads is a sex-dimorphic process which evolved to produce sex-specific types of germ cells. The process of gonadal sex differentiation is directed by the action of the somatic cells and ultimately results in germ cells differentiating to become functional gametes through spermatogenesis or oogenesis. This tightly controlled process depends on the proper sequential expression of many genes and signalling pathways. Disturbances of this process can be manifested as a large spectrum of disorders, ranging from severe disorders of sex development (DSD) to - in the genetic male - mild reproductive problems within the testicular dysgenesis syndrome (TDS), with large overlap between the syndromes. These disorders carry an increased but variable risk of germ cell neoplasia. In this review, we discuss the pathogenesis of germ cell neoplasia associated with gonadal dysgenesis, especially in individuals with 46,XY DSD. We summarise knowledge concerning development and sex differentiation of human gonads, with focus on sex-dimorphic steps of germ cell maturation, including meiosis. We also briefly outline the histopathology of germ cell neoplasia in situ (GCNIS) and gonadoblastoma (GDB), which are essentially the same precursor lesion but with different morphological structure dependent upon the masculinisation of the somatic niche. To assess the risk of germ cell neoplasia in different types of DSD, we have performed a PubMed search and provide here a synthesis of the evidence from studies published since 2006. We present a model for pathogenesis of GCNIS/GDB in TDS/DSD, with the risk of malignancy determined by the presence of the testis-inducing Y chromosome and the degree of masculinisation. The associations between phenotype and the risk of neoplasia are likely further modulated in each individual by the constellation of the gene polymorphisms and environmental factors.
Collapse
Affiliation(s)
- Anne Jørgensen
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| | - Marie Lindhardt Johansen
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| | - Anders Juul
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| | - Niels E Skakkebaek
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| | - Katharina M Main
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| | - Ewa Rajpert-De Meyts
- Department of Growth & Reproduction and International Center for Research and Training in Endocrine Disruption of Male Reproduction and Child Health (EDMaRC), Rigshospitalet, University of Copenhagen, Denmark.
| |
Collapse
|
31
|
Liang GJ, Zhang XF, Wang JJ, Sun YC, Sun XF, Cheng SF, Li L, De Felici M, Shen W. Activin A accelerates the progression of fetal oocytes throughout meiosis and early oogenesis in the mouse. Stem Cells Dev 2015; 24:2455-65. [PMID: 26083127 DOI: 10.1089/scd.2015.0068] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Activins can exert several roles in ovary development. However, little is known about their involvement in early mammalian oogenesis. In this study, we reported that activin receptors (including ActRIA, ActRIB, ActRIIA, and ActRIIB) are expressed throughout the development of the mouse ovaries from 12.5 days postcoitum (dpc) to 21 days postparturition (dpp). Moreover, we found that in vitro, the addition of activin A (ActA) to the culture medium of 12.5 dpc ovarian tissues accelerated the progression of oocytes throughout meiotic prophase I stages. This result was reproduced in vivo following administration of ActA to pregnant mice. The in vitro effect of ActA was associated with increased expression of premeiotic and meiotic genes (including Dazl, Spo11, Stra8, Scp3, and Rec8) in the ovarian tissues. Mechanistically, ActA-dependent SMAD3 signaling modulated the expression of members of the retinoic acid (RA) system, including the RA degradation CYP26B1 enzyme and the RA receptors. Finally, ActA promoted the survival and growth of fetal and early postnatal oocytes and primordial follicle assembly both in vitro and in vivo. In conclusion, the present study identifies new roles of ActA in early oogenesis and suggested that ActA and RA might cooperate in promoting meiosis in female germ cells.
Collapse
Affiliation(s)
- Gui-Jin Liang
- 1 Institute of Reproductive Sciences, Qingdao Agricultural University , Qingdao, China .,2 Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Animal Science and Technology, Qingdao Agricultural University , Qingdao, China
| | - Xi-Feng Zhang
- 1 Institute of Reproductive Sciences, Qingdao Agricultural University , Qingdao, China .,2 Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Animal Science and Technology, Qingdao Agricultural University , Qingdao, China
| | - Jun-Jie Wang
- 1 Institute of Reproductive Sciences, Qingdao Agricultural University , Qingdao, China .,2 Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Animal Science and Technology, Qingdao Agricultural University , Qingdao, China
| | - Yuan-Chao Sun
- 1 Institute of Reproductive Sciences, Qingdao Agricultural University , Qingdao, China .,2 Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Animal Science and Technology, Qingdao Agricultural University , Qingdao, China
| | - Xiao-Feng Sun
- 1 Institute of Reproductive Sciences, Qingdao Agricultural University , Qingdao, China .,3 College of Life Science, Qingdao Agricultural University , Qingdao, China
| | - Shun-Feng Cheng
- 1 Institute of Reproductive Sciences, Qingdao Agricultural University , Qingdao, China .,2 Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Animal Science and Technology, Qingdao Agricultural University , Qingdao, China
| | - Lan Li
- 1 Institute of Reproductive Sciences, Qingdao Agricultural University , Qingdao, China .,2 Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Animal Science and Technology, Qingdao Agricultural University , Qingdao, China
| | - Massimo De Felici
- 4 Department of Biomedicine and Prevention, University of Rome Tor Vergata , Rome, Italy
| | - Wei Shen
- 1 Institute of Reproductive Sciences, Qingdao Agricultural University , Qingdao, China .,2 Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, College of Animal Science and Technology, Qingdao Agricultural University , Qingdao, China
| |
Collapse
|
32
|
Bayne RAL, Kinnell HL, Coutts SM, He J, Childs AJ, Anderson RA. GDF9 is transiently expressed in oocytes before follicle formation in the human fetal ovary and is regulated by a novel NOBOX transcript. PLoS One 2015; 10:e0119819. [PMID: 25790371 PMCID: PMC4366263 DOI: 10.1371/journal.pone.0119819] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/16/2015] [Indexed: 12/05/2022] Open
Abstract
During human fetal ovary development, the process of primordial follicle formation is immediately preceded by a highly dynamic period of germ cell and somatic cell reorganisation. This is regulated by germ-cell specific transcription regulators, by the conserved RNA binding proteins DAZL and BOLL and by secreted growth factors of the TGFβ family, including activin βA: these all show changing patterns of expression preceding follicle formation. In mice, the transcription factor Nobox is essential for follicle formation and oocyte survival, and NOBOX regulates the expression of GDF9 in humans. We have therefore characterised the expression of GDF9 in relation to these known key factors during follicle formation in the human fetal ovary. mRNA levels of GDF9, BMP15 and NOBOX were quantified by qRT-PCR and showed dramatic increases across gestation. GDF9 protein expression was localised by immunohistochemistry to the same population of germ cells as those expressing activin βA prior to follicle formation but did not co-localise with either BOLL or DAZL. A novel NOBOX isoform was identified in fetal ovary that was shown to be capable of up-regulating the GDF9 promoter in reporter assays. Thus, during oogenesis in humans, oocytes go through a dynamic and very sharply demarcated sequence of changes in expression of these various proteins, even within individual germ cell nests, likely to be of major functional significance in determining selective germ cell survival at this key stage in ovarian development. Transcriptional variation may contribute to the range of age of onset of POI in women with NOBOX mutations.
Collapse
Affiliation(s)
- Rosemary A. L. Bayne
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
- * E-mail:
| | - Hazel L. Kinnell
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Shiona M. Coutts
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Jing He
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| | - Andrew J. Childs
- Department of Comparative Biomedical Sciences, The Royal Veterinary College, London, United Kingdom
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Queen’s Medical Research Institute, Edinburgh, United Kingdom
| |
Collapse
|
33
|
Epistasis between polymorphisms in ACVR2B and ADAMTS19 is associated with premature ovarian failure. Menopause 2015; 22:212-6. [DOI: 10.1097/gme.0000000000000285] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
34
|
Patel AK, Shah RK, Patel UA, Tripathi AK, Joshi CG. Goat activin receptor type IIB knockdown by muscle specific promoter driven artificial microRNAs. J Biotechnol 2014; 187:87-97. [PMID: 25107506 DOI: 10.1016/j.jbiotec.2014.07.450] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 07/20/2014] [Accepted: 07/25/2014] [Indexed: 01/30/2023]
Abstract
Activin receptor type IIB (ACVR2B) is a transmembrane receptor which mediates signaling of TGF beta superfamily ligands known to function in regulation of muscle mass, embryonic development and reproduction. ACVR2B antagonism has shown to enhance the muscle growth in several disease and transgenic models. Here, we show ACVR2B knockdown by RNA interference using muscle creatine kinase (MCK) promoter driven artificial microRNAs (amiRNAs). Among the various promoter elements tested, the ∼1.26 kb MCK promoter region showed maximum transcriptional activity in goat myoblasts cells. We observed up to 20% silencing in non-myogenic 293T cells and up to 32% silencing in myogenic goat myoblasts by MCK directed amiRNAs by transient transfection. Goat myoblasts stably integrated with MCK directed amiRNAs showed merely 8% silencing in proliferating myoblasts which was increased to 34% upon induction of differentiation at transcript level whereas up to 57% silencing at protein level. Knockdown of ACVR2B by 5'-UTR derived amiRNAs resulted in decreased SMAD2/3 signaling, increased expression of myogenic regulatory factors (MRFs) and enhanced proliferation and differentiation of myoblasts. Unexpectedly, knockdown of ACVR2B by 3'-UTR derived amiRNAs resulted in increased SMAD2/3 signaling, reduced expression of MRFs and suppression of myogenesis. Our study offers muscle specific knockdown of ACVR2B as a potential strategy to enhance muscle mass in the farm animal species.
Collapse
Affiliation(s)
- Amrutlal K Patel
- Department of Animal Biotechnology, College of Veterinary Science and Animal Husbandry, Anand Agricultural University, Anand 388001, India
| | - Ravi K Shah
- Department of Animal Biotechnology, College of Veterinary Science and Animal Husbandry, Anand Agricultural University, Anand 388001, India
| | - Utsav A Patel
- Department of Animal Biotechnology, College of Veterinary Science and Animal Husbandry, Anand Agricultural University, Anand 388001, India
| | - Ajai K Tripathi
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Chaitanya G Joshi
- Department of Animal Biotechnology, College of Veterinary Science and Animal Husbandry, Anand Agricultural University, Anand 388001, India.
| |
Collapse
|
35
|
Makanji Y, Zhu J, Mishra R, Holmquist C, Wong WPS, Schwartz NB, Mayo KE, Woodruff TK. Inhibin at 90: from discovery to clinical application, a historical review. Endocr Rev 2014; 35:747-94. [PMID: 25051334 PMCID: PMC4167436 DOI: 10.1210/er.2014-1003] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
When it was initially discovered in 1923, inhibin was characterized as a hypophysiotropic hormone that acts on pituitary cells to regulate pituitary hormone secretion. Ninety years later, what we know about inhibin stretches far beyond its well-established capacity to inhibit activin signaling and suppress pituitary FSH production. Inhibin is one of the major reproductive hormones involved in the regulation of folliculogenesis and steroidogenesis. Although the physiological role of inhibin as an activin antagonist in other organ systems is not as well defined as it is in the pituitary-gonadal axis, inhibin also modulates biological processes in other organs through paracrine, autocrine, and/or endocrine mechanisms. Inhibin and components of its signaling pathway are expressed in many organs. Diagnostically, inhibin is used for prenatal screening of Down syndrome as part of the quadruple test and as a biochemical marker in the assessment of ovarian reserve. In this review, we provide a comprehensive summary of our current understanding of the biological role of inhibin, its relationship with activin, its signaling mechanisms, and its potential value as a diagnostic marker for reproductive function and pregnancy-associated conditions.
Collapse
Affiliation(s)
- Yogeshwar Makanji
- Department of Obstetrics and Gynecology (Y.M., J.Z., C.H., W.P.S.W., T.K.W.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60610; Center for Molecular Innovation and Drug Discovery (R.M., C.H.), Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208; and Department of Molecular Biosciences (N.B.S., K.E.M., T.K.W.), Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Fowler PA, Childs AJ, Courant F, MacKenzie A, Rhind SM, Antignac JP, Le Bizec B, Filis P, Evans F, Flannigan S, Maheshwari A, Bhattacharya S, Monteiro A, Anderson RA, O'Shaughnessy PJ. In utero exposure to cigarette smoke dysregulates human fetal ovarian developmental signalling. Hum Reprod 2014; 29:1471-89. [PMID: 24847019 DOI: 10.1093/humrep/deu117] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
STUDY QUESTION How does maternal cigarette smoking disturb development of the human fetal ovary? SUMMARY ANSWER Maternal smoking increases fetal estrogen titres and dysregulates several developmental processes in the fetal ovary. WHAT IS KNOWN ALREADY Exposure to maternal cigarette smoking during gestation reduces human fetal ovarian cell numbers, germ cell proliferation and subsequent adult fecundity. STUDY DESIGN, SIZE, DURATION The effects of maternal cigarette smoking on the second trimester human fetal ovary, fetal endocrine signalling and fetal chemical burden were studied. A total of 105 fetuses were studied, 56 from mothers who smoked during pregnancy and 49 from those who did not. PARTICIPANTS/MATERIALS, SETTING METHODS Ovary, liver and plasma samples were collected from electively terminated, normally progressing, second trimester human fetuses. Circulating fetal hormones, levels of 73 fetal ovarian transcripts, protein localization, density of oocytes/primordial follicles and levels of 16 polycyclic aromatic hydrocarbons (PAHs) in the fetal liver were determined. MAIN RESULTS AND THE ROLE OF CHANCE Circulating fetal estrogen levels were very high and were increased by maternal smoking (ANOVA, P = 0.055-0.004 versus control). Smoke exposure also dysregulated (two-way ANOVA, smoking versus gestation weeks interaction, P = 0.046-0.023) four fetal ovarian genes (cytochrome P450 scc [CYP11A1], NOBOX oogenesis homeobox [NOBOX], activator of apoptosis harakiri [HRK], nuclear receptor subfamily 2, group E, member 1 [NR2E1]), shifted the ovarian Inhibin βA/inhibin α ratio (NHBA/INHA) transcript ratio in favour of activin (ANOVA, P = 0.049 versus control) and reduced the proportion of dominant-negative estrogen receptor 2 (ERβ: ESR2) isoforms in half the exposed fetuses. PAHs, ligands for the aryl hydrocarbon receptor (AHR), were increased nearly 6-fold by maternal smoking (ANOVA, P = 0.011 versus control). A fifth transcript, COUP transcription factor 1 (nuclear receptor subfamily 2, group F, member 1: NR2F1, which contains multiple AHR-binding sites), was both significantly increased (ANOVA, P = 0.026 versus control) and dysregulated by (two-way ANOVA, smoking versus gestation weeks interaction, P = 0.021) maternal smoking. NR2F1 is associated with repression of FSHR expression and smoke-exposed ovaries failed to show the normal increase in FSHR expression during the second trimester. There was a significantly higher number of DEAD (Asp-Glu-Ala-Asp) box polypeptide 4 (DDX4) VASA-positive (ANOVA, P = 0.016 versus control), but not POU domain, class 1, transcription factor 1 (POU5F1) OCT3/4-positive, oocytes in smoke-exposed fetuses and this matched with a significantly higher number of primordial follicles (ANOVA, P = 0.024 versus control). LIMITATIONS, REASONS FOR CAUTION The effects of maternal smoking on establishment of the maximum fetal primordial follicle pool cannot be reliably studied in our population since the process is not completed until 28 weeks of gestation and normal fetuses older than 21 weeks of gestation are not available for study. Our data suggest that some fetal ovaries are affected by smoke exposure while others are not, indicating that additional studies, with larger numbers, may show more significant effects. WIDER IMPLICATIONS OF THE FINDINGS Fetal exposure to chemicals in cigarette smoke is known to lead to reduced fecundity in women. Our study suggests, for the first time, that this occurs via mechanisms involving activation of AHR, disruption of inhibin/activin and estrogen signalling, increased exposure to estrogen and dysregulation of multiple molecular pathways in the exposed human fetal ovary. Our data also suggest that alterations in the ESR2 positive and dominant negative isoforms may be associated with reduced sensitivity of some fetuses to increased estrogens and maternal smoking. STUDY FUNDING/COMPETING INTEREST(S) The study was supported by grants from the Chief Scientist Office (Scottish Executive, CZG/1/109, and CZG/4/742), NHS Grampian Endowments (08/02), the European Community's Seventh Framework Programme (FP7/2007-2013) under grant agreement no. 212885, a Society for Reproduction & Fertility summer studentship, Medical Research Scotland (research grant 354 FRG) and the Medical Research Council (WBS: U.1276.00.002.00001 and G1100357). The authors declare they have no competing interests, be it financial, personal or professional.
Collapse
|
37
|
Abstract
The female germline comprises a reserve population of primordial (non-growing) follicles containing diplotene oocytes arrested in the first meiotic prophase. By convention, the reserve is established when all individual oocytes are enclosed by granulosa cells. This commonly occurs prior to or around birth, according to species. Histologically, the ‘reserve’ is the number of primordial follicles in the ovary at any given age and is ultimately depleted by degeneration and progression through folliculogenesis until exhausted. How and when the reserve reaches its peak number of follicles is determined by ovarian morphogenesis and germ cell dynamics involving i) oogonial proliferation and entry into meiosis producing an oversupply of oocytes and ii) large-scale germ cell death resulting in markedly reduced numbers surviving as the primordial follicle reserve. Our understanding of the processes maintaining the reserve comes primarily from genetically engineered mouse models, experimental activation or destruction of oocytes, and quantitative histological analysis. As the source of ovulated oocytes in postnatal life, the primordial follicle reserve requires regulation of i) its survival or maintenance, ii) suppression of development (dormancy), and iii) activation for growth and entry into folliculogenesis. The mechanisms influencing these alternate and complex inter-related phenomena remain to be fully elucidated. Drawing upon direct and indirect evidence, we discuss the controversial concept of postnatal oogenesis. This posits a rare population of oogonial stem cells that contribute new oocytes to partially compensate for the age-related decline in the primordial follicle reserve.
Collapse
|
38
|
Ge ZH, Wang ZX, Yu TL, Yang N, Sun Y, Hao CL, Sun LX. Morphine improved the antitumor effects on MCF-7 cells in combination with 5-Fluorouracil. Biomed Pharmacother 2013; 68:299-305. [PMID: 24210071 DOI: 10.1016/j.biopha.2013.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 09/24/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The most frequently used opioid in cancer pain management is morphine which remains a cornerstone for the management of cancer pain, due to the largest experience existing among physicians and widely availability in a variety of formulation. Considering that analgesics on cancer pain is often under the condition of chemotherapy and 5-Fluorouracil (5-FU) is widely used today as a potent drug for the treatment of advanced cancers, whether analgesics such as morphine, interferes the chemotherapy such as 5-FU, arose as a considerable problem. METHODS In this study, the MCF-7 breast cancer cells were used to determine the antitumor effects of the 5-FU in combination with morphine. The cell proliferation was determined by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay and the apoptosis was determined by the Annexin V/PI staining and flow cytometry. The immunocytochemistry and western blot was used to determine the Bcl-2 and Bax expression. RESULTS It was shown that in MCF-7 cells, the proliferation was inhibited, the apoptosis was promoted, the Bcl-2 expression was suppressed and the Bax expression was promoted by both 5-FU alone and morphine alone, while the superior effects were achieved in combination with the two drugs. CONCLUSION These results suggest that the morphine may have the beneficial effects on the antitumor chemotherapy with 5-FU, in stead of interferential effects.
Collapse
Affiliation(s)
- Zhi-Hua Ge
- Affiliated Hospital of Chengde Medical College, Hebei province, 067000 Chengde, PR China
| | - Zhi-Xue Wang
- Affiliated Hospital of Chengde Medical College, Hebei province, 067000 Chengde, PR China
| | - Tie-Li Yu
- Affiliated Hospital of Chengde Medical College, Hebei province, 067000 Chengde, PR China
| | - Ning Yang
- Affiliated Hospital of Chengde Medical College, Hebei province, 067000 Chengde, PR China
| | - Yu Sun
- Affiliated Hospital of Chengde Medical College, Hebei province, 067000 Chengde, PR China
| | - Chang-Lai Hao
- Affiliated Hospital of Chengde Medical College, Hebei province, 067000 Chengde, PR China.
| | - Li-Xin Sun
- Affiliated Hospital of Chengde Medical College, Hebei province, 067000 Chengde, PR China.
| |
Collapse
|
39
|
Anderson RA, McIlwain L, Coutts S, Kinnell HL, Fowler PA, Childs AJ. Activation of the aryl hydrocarbon receptor by a component of cigarette smoke reduces germ cell proliferation in the human fetal ovary. Mol Hum Reprod 2013; 20:42-8. [PMID: 23979962 PMCID: PMC3867980 DOI: 10.1093/molehr/gat059] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Fetal life is a critical time for female fertility, when germ cells complete proliferation, initiate meiosis and ultimately form the lifetime stock of primordial follicles. Female fertility may be reduced by in utero exposure to cigarette smoke, which contains ligands for the aryl hydrocarbon receptor (AhR). The AhR is a critical regulator of ovarian germ cell survival in mice; thus activation of this receptor in the ovaries of fetuses exposed to maternal cigarette smoke in utero may provide a mechanism by which female fertility is reduced in later life. We have therefore investigated AhR expression in the human fetal ovary, and examined the effects of an AhR ligand present in cigarette smoke, on germ cells in human fetal ovaries cultured in vitro. The results showed that AHR mRNA expression increased 2-fold between first and late second trimester (P = 0.008). AhR protein was confined to germ cells at all gestations, but varied from expression in most germ cells during the first trimester, to only patchy expression by clusters of germ cells at later gestations. Culture of human fetal ovaries with the AhR ligand 9,10-dimethyl-1,2-benzanthracene-3,4-dihydrodiol (DMBA-DHD; a component of cigarette smoke) did not affect germ cell number in vitro, but significantly reduced the proportion of proliferating germ cells by 29% (as assessed by phospho-histone H3 staining (P = 0.04)). Germ cell apoptosis was not significantly affected. These results reveal that germ cells in the human fetal ovary express AhR from the proliferative stage of development through entry into meiosis and beyond, and demonstrate that AhR ligands found in cigarette smoke have the capacity to impair human fetal ovarian germ cell proliferation.
Collapse
Affiliation(s)
- Richard A Anderson
- MRC Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK and
| | | | | | | | | | | |
Collapse
|
40
|
Dumesic DA, Richards JS. Ontogeny of the ovary in polycystic ovary syndrome. Fertil Steril 2013; 100:23-38. [PMID: 23472949 DOI: 10.1016/j.fertnstert.2013.02.011] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 02/05/2013] [Accepted: 02/06/2013] [Indexed: 12/11/2022]
Abstract
Activation of primordial follicles into the growing pool, selection of the dominant follicle, and its eventual ovulation require complex endocrine and metabolic interactions as well as intraovarian paracrine signals to coordinate granulosa cell proliferation, theca cell differentiation, and oocyte maturation. Early preantral follicle development relies mostly upon mesenchymal-epithelial cell interactions, intraovarian paracrine signals, and oocyte-secreted factors, whereas development of the antral follicle depends on circulating gonadotropins as well as locally derived regulators. In women with polycystic ovary syndrome (PCOS), ovarian hyperandrogenism, hyperinsulinemia from insulin resistance, and altered intrafollicular paracrine signaling perturb the activation, survival, growth, and selection of follicles, causing accumulation of small antral follicles within the periphery of the ovary, giving it a polycystic morphology. Altered adipocyte-ovarian interactions further compound these adverse events on follicle development and also can harm the oocyte, particularly in the presence of increased adiposity. Finally, endocrine antecedents of PCOS occur in female infants born to mothers with PCOS, which suggests that interactions between genes and the maternal-fetal hormonal environment may program ovarian function after birth.
Collapse
Affiliation(s)
- Daniel A Dumesic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA.
| | | |
Collapse
|
41
|
Tran S, Zhou X, Lafleur C, Calderon MJ, Ellsworth BS, Kimmins S, Boehm U, Treier M, Boerboom D, Bernard DJ. Impaired fertility and FSH synthesis in gonadotrope-specific Foxl2 knockout mice. Mol Endocrinol 2013; 27:407-21. [PMID: 23340250 DOI: 10.1210/me.2012-1286] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Impairments in pituitary FSH synthesis or action cause infertility. However, causes of FSH dysregulation are poorly described, in part because of our incomplete understanding of mechanisms controlling FSH synthesis. Previously, we discovered a critical role for forkhead protein L2 (FOXL2) in activin-stimulated FSH β-subunit (Fshb) transcription in immortalized cells in vitro. Here, we tested the hypothesis that FOXL2 is required for FSH synthesis in vivo. Using a Cre/lox approach, we selectively ablated Foxl2 in murine anterior pituitary gonadotrope cells. Conditional knockout (cKO) mice developed overtly normally but were subfertile in adulthood. Testis size and spermatogenesis were significantly impaired in cKO males. cKO females exhibited reduced ovarian weight and ovulated fewer oocytes in natural estrous cycles compared with controls. In contrast, ovaries of juvenile cKO females showed normal responses to exogenous gonadotropin stimulation. Both male and female cKO mice were FSH deficient, secondary to diminished pituitary Fshb mRNA production. Basal and activin-stimulated Fshb expression was similarly impaired in Foxl2 depleted primary pituitary cultures. Collectively, these data definitively establish FOXL2 as the first identified gonadotrope-restricted transcription factor required for selective FSH synthesis in vivo.
Collapse
Affiliation(s)
- Stella Tran
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Pangas SA. Regulation of the ovarian reserve by members of the transforming growth factor beta family. Mol Reprod Dev 2012; 79:666-79. [PMID: 22847922 DOI: 10.1002/mrd.22076] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 07/13/2012] [Indexed: 11/08/2022]
Abstract
Genetic or environmental factors that affect the endowment of oocytes, their assembly into primordial follicles, or their subsequent entry into the growing follicle pool can disrupt reproductive function and may underlie disorders such as primary ovarian insufficiency. Mouse models have been instrumental in identifying genes important in ovarian development, and a number of genes now associated with ovarian dysfunction in women were first identified as causing reproductive defects in knockout mice. The transforming growth factor beta (TGFB) family consists of developmentally important growth factors that include the TGFBs, anti-Müllerian hormone (AMH), activins, bone morphogenetic proteins (BMPs), and growth and differentiation factor 9 (GDF9). The ovarian primordial follicle pool is the source of oocytes in adults. Development of this pool can be grossly divided into three key processes: (1) establishment of oocytes during embryogenesis followed by (2) assembly and (3) activation of the primordial follicle. Disruptions in any of these processes may cause reproductive dysfunction. Most members of the TGFB family show pivotal roles in each of these areas. Understanding the phenotypes of various mouse models for this protein family will be directly relevant to understanding how disruptions in TGFB family signaling result in reproductive diseases in women and will present new areas for development of tailored diagnostics and interventions for infertility.
Collapse
Affiliation(s)
- Stephanie A Pangas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA.
| |
Collapse
|
43
|
Knight PG, Satchell L, Glister C. Intra-ovarian roles of activins and inhibins. Mol Cell Endocrinol 2012; 359:53-65. [PMID: 21664422 DOI: 10.1016/j.mce.2011.04.024] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2010] [Revised: 04/26/2011] [Accepted: 04/27/2011] [Indexed: 01/11/2023]
Abstract
Granulosa cells are the main ovarian source of inhibins, activins and activin-binding protein (follistatin) while germ (oogonia, oocytes) and somatic (theca, granulosa, luteal) cells express activin receptors, signaling components and inhibin co-receptor (betaglycan). Activins are implicated in various intra-ovarian roles including germ cell survival and primordial follicle assembly; follicle growth from preantral to mid-antral stages; suppression of thecal androgen production; promotion of granulosa cell proliferation, FSHR and CYP19A1 expression; enhancement of oocyte developmental competence; retardation of follicle luteinization and/or atresia and involvement in luteolysis. Inhibins (primarily inhibin A) are produced in greatest amounts by preovulatory follicles (and corpus luteum in primates) and suppress FSH secretion through endocrine negative feedback. Together with follistatin, inhibins act locally to oppose auto-/paracrine activin (and BMP) signaling thus modulating many of the above processes. The balance between activin-inhibin shifts during follicle development with activin signalling prevailing at earlier stages but declining as inhibin and betaglycan expression rise.
Collapse
Affiliation(s)
- Phil G Knight
- School of Biological Sciences, Hopkins Building, University of Reading, Whiteknights, Reading RG6 6UB, UK.
| | | | | |
Collapse
|
44
|
Zhang ZP, Liang GJ, Zhang XF, Zhang GL, Chao HH, Li L, Sun XF, Min LJ, Pan QJ, Shi QH, Sun QY, De Felici M, Shen W. Growth of mouse oocytes to maturity from premeiotic germ cells in vitro. PLoS One 2012; 7:e41771. [PMID: 22848595 PMCID: PMC3404094 DOI: 10.1371/journal.pone.0041771] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 06/26/2012] [Indexed: 11/23/2022] Open
Abstract
In the present study, we established an in vitro culture system suitable for generating fertilizable oocytes from premeiotic mouse female germ cells. These results were achieved after first establishing an in vitro culture system allowing immature oocytes from 12–14 day- old mice to reach meiotic maturation through culture onto preantral granulosa cell (PAGC) monolayers in the presence of Activin A (ActA). To generate mature oocytes from premeiotic germ cells, pieces of ovaries from 12.5 days post coitum (dpc) embryos were cultured in medium supplemented with ActA for 28 days and the oocytes formed within the explants were isolated and cocultured onto PAGC monolayers in the presence of ActA for 6–7 days. The oocytes were then subjected to a final meiotic maturation assay to evaluate their capability to undergo germinal vesicle break down (GVBD) and reach the metaphase II (MII) stage. We found that during the first 28 days of culture, a significant number of oocytes within the ovarian explants reached nearly full growth and formed preantral follicle-like structures with the surrounding somatic cells. GSH level and Cx37 expression in the oocytes within the explants were indicative of proper developmental conditions. Moreover, the imprinting of Igf2r and Peg3 genes in these oocytes was correctly established. Further culture onto PAGCs in the presence of ActA allowed about 16% of the oocytes to undergo GVBD, among which 17% reached the MII stage during the final 16–18 hr maturation culture. These MII oocytes showed normal spindle and chromosome assembly and a correct ERK1/2 activity. About 35% of the in vitro matured oocytes were fertilized and 53.44% of them were able to reach the 2-cell stage. Finally, around 7% of the 2-cell embryos developed to the morula/blastocyst stage.
Collapse
Affiliation(s)
- Zhi-Peng Zhang
- Key Laboratory of Animal Reproduction and Germplasm Enhancement in Universities of Shandong, Qingdao Agricultural University, Qingdao, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Germline development from human pluripotent stem cells toward disease modeling of infertility. Fertil Steril 2012; 97:1250-9. [DOI: 10.1016/j.fertnstert.2012.04.037] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/23/2012] [Accepted: 04/25/2012] [Indexed: 01/05/2023]
|
46
|
Molecular control of oogenesis. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1896-912. [PMID: 22634430 DOI: 10.1016/j.bbadis.2012.05.013] [Citation(s) in RCA: 259] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2012] [Revised: 05/08/2012] [Accepted: 05/13/2012] [Indexed: 11/20/2022]
Abstract
Oogenesis is a complex process regulated by a vast number of intra- and extra-ovarian factors. Oogonia, which originate from primordial germ cells, proliferate by mitosis and form primary oocytes that arrest at the prophase stage of the first meiotic division until they are fully-grown. Within primary oocytes, synthesis and accumulation of RNAs and proteins throughout oogenesis are essential for oocyte growth and maturation; and moreover, crucial for developing into a viable embryo after fertilization. Oocyte meiotic and developmental competence is gained in a gradual and sequential manner during folliculogenesis and is related to the fact that the oocyte grows in interaction with its companion somatic cells. Communication between oocyte and its surrounding granulosa cells is vital, both for oocyte development and for granulosa cells differentiation. Oocytes depend on differentiated cumulus cells, which provide them with nutrients and regulatory signals needed to promote oocyte nuclear and cytoplasmic maturation and consequently the acquisition of developmental competence.The purpose of this article is to summarize recent knowledge on the molecular aspects of oogenesis and oocyte maturation, and the crucial role of cumulus-cell interactions, highlighting the valuable contribution of experimental evidences obtained in animal models. This article is part of a Special Issue entitled: Molecular Genetics of Human Reproductive Failure.
Collapse
|
47
|
Sarraj MA, Drummond AE. Mammalian foetal ovarian development: consequences for health and disease. Reproduction 2012; 143:151-63. [DOI: 10.1530/rep-11-0247] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The development of a normal ovary during foetal life is essential for the production and ovulation of a high-quality oocyte in adult life. Early in embryogenesis, the primordial germ cells (PGCs) migrate to and colonise the genital ridges. Once the PGCs reach the bipotential gonad, the absence of the sex-determining region on the Y chromosome (SRY) gene and the presence of female-specific genes ensure that the indifferent gonad takes the female pathway and an ovary forms. PGCs enter into meiosis, transform into oogonia and ultimately give rise to oocytes that are later surrounded by granulosa cells to form primordial follicles. Various genes and signals are implicated in germ and somatic cell development, leading to successful follicle formation and normal ovarian development. This review focuses on the differentiation events, cellular processes and molecular mechanisms essential for foetal ovarian development in the mice and humans. A better understanding of these early cellular and morphological events will facilitate further study into the regulation of oocyte development, manifestation of ovarian disease and basis of female infertility.
Collapse
|
48
|
Moore BC, Milnes MR, Kohno S, Katsu Y, Iguchi T, Woodruff TK, Guillette LJ. Altered gonadal expression of TGF-β superfamily signaling factors in environmental contaminant-exposed juvenile alligators. J Steroid Biochem Mol Biol 2011; 127:58-63. [PMID: 21251980 DOI: 10.1016/j.jsbmb.2011.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2010] [Revised: 01/05/2011] [Accepted: 01/10/2011] [Indexed: 11/29/2022]
Abstract
Environmental contaminant exposure can influence gonadal steroid signaling milieus; however, little research has investigated the vulnerability of non-steroidal signaling pathways in the gonads. Here we use American alligators (Alligator mississippiensis) hatched from field-collected eggs to analyze gonadal mRNA transcript levels of the activin-inhibin-follistatin gene expression network and growth differentiation factor 9. The eggs were collected from Lake Woodruff National Wildlife Refuge, a site with minimal anthropogenic influence, and Lake Apopka, a highly contaminated lake adjacent to a former EPA Superfund site. The hatchling alligators were raised for 13 months under controlled conditions, thus limiting differences to embryonic origins. Our data reveal sexually dimorphic mRNA expression in 13-month-old alligator gonads similar to patterns established in vertebrates with genetic sex determination. In addition, we observed a relationship between lake of origin and mRNA expression of activin/inhibin subunits α and βB, follistatin, and growth differentiation factor 9. Our study suggests that embryonic exposure to environmental contaminants can affect future non-steroidal signaling patterns in the gonads of a long-lived species.
Collapse
Affiliation(s)
- Brandon C Moore
- Department of Biology, University of Florida, Gainesville, FL 32611-8525, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Childs AJ, Cowan G, Kinnell HL, Anderson RA, Saunders PTK. Retinoic Acid signalling and the control of meiotic entry in the human fetal gonad. PLoS One 2011; 6:e20249. [PMID: 21674038 PMCID: PMC3108594 DOI: 10.1371/journal.pone.0020249] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Accepted: 04/28/2011] [Indexed: 12/24/2022] Open
Abstract
The development of mammalian fetal germ cells along oogenic or spermatogenic fate trajectories is dictated by signals from the surrounding gonadal environment. Germ cells in the fetal testis enter mitotic arrest, whilst those in the fetal ovary undergo sex-specific entry into meiosis, the initiation of which is thought to be mediated by selective exposure of fetal ovarian germ cells to mesonephros-derived retinoic acid (RA). Aspects of this model are hard to reconcile with the spatiotemporal pattern of germ cell differentiation in the human fetal ovary, however. We have therefore examined the expression of components of the RA synthesis, metabolism and signalling pathways, and their downstream effectors and inhibitors in germ cells around the time of the initiation of meiosis in the human fetal gonad. Expression of the three RA-synthesising enzymes, ALDH1A1, 2 and 3 in the fetal ovary and testis was equal to or greater than that in the mesonephros at 8–9 weeks gestation, indicating an intrinsic capacity within the gonad to synthesise RA. Using immunohistochemistry to detect RA receptors RARα, β and RXRα, we find germ cells to be the predominant target of RA signalling in the fetal human ovary, but also reveal widespread receptor nuclear localization indicative of signalling in the testis, suggesting that human fetal testicular germ cells are not efficiently shielded from RA by the action of the RA-metabolising enzyme CYP26B1. Consistent with this, expression of CYP26B1 was greater in the human fetal ovary than testis, although the sexually-dimorphic expression patterns of the germ cell-intrinsic regulators of meiotic initiation, STRA8 and NANOS2, appear conserved. Finally, we demonstrate that RA induces a two-fold increase in STRA8 expression in cultures of human fetal testis, but is not sufficient to cause widespread meiosis-associated gene expression. Together, these data indicate that while local production of RA within the fetal ovary may be important in regulating the onset of meiosis in the human fetal ovary, mechanisms other than CYP26B1-mediated metabolism of RA may exist to inhibit the entry of germ cells into meiosis in the human fetal testis.
Collapse
Affiliation(s)
- Andrew J Childs
- Medical Research Council Human Reproductive Sciences Unit, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | | | |
Collapse
|
50
|
Maman E, Yung Y, Cohen B, Konopnicki S, Dal Canto M, Fadini R, Kanety H, Kedem A, Dor J, Hourvitz A. Expression and regulation of sFRP family members in human granulosa cells. Mol Hum Reprod 2011; 17:399-404. [DOI: 10.1093/molehr/gar010] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|