1
|
Sage MAG, Duffy DM. Novel Plasma Membrane Androgen Receptor SLC39A9 Mediates Ovulatory Changes in Cells of the Monkey Ovarian Follicle. Endocrinology 2024; 165:bqae071. [PMID: 38889246 PMCID: PMC11212825 DOI: 10.1210/endocr/bqae071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/23/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Follicular androgens are important for successful ovulation and fertilization. The classical nuclear androgen receptor (AR) is a transcription factor expressed in the cells of the ovarian follicle. Androgen actions can also occur via membrane androgen receptor SLC39A9. Studies in fish ovary demonstrated that androgens bind to SLC39A9 and increase intracellular zinc to regulate ovarian cell function. To determine if SLC39A9 is expressed and functional in the key cell types of the mammalian ovulatory follicle, adult female cynomolgus macaques underwent ovarian stimulation. Ovaries or ovarian follicular aspirates were harvested at 0, 12, 24, and 36 hours after human chorionic gonadotropin (hCG). SLC39A9 and AR mRNA and protein were present in granulosa, theca, and vascular endothelial cells across the entire 40-hour ovulatory window. Testosterone, bovine serum albumin-conjugated testosterone (BSA-T), and androstenedione stimulated zinc influx in granulosa, theca, and vascular endothelial cells. The SLC39A9-selective agonist (-)-epicatechin also stimulated zinc influx in vascular endothelial cells. Taken together, these data support the conclusion that SLC39A9 activation via androgen induces zinc influx in key ovarian cells. Testosterone, BSA-T, and androstenedione each increased proliferation in vascular endothelial cells, indicating the potential involvement of SLC39A9 in ovulatory angiogenesis. Vascular endothelial cell migration also increased after treatment with testosterone, but not after treatment with BSA-T or androstenedione, suggesting that androgens stimulate vascular endothelial cell migration through nuclear AR but not SLC39A9. The presence of SLC39A9 receptors and SLC39A9 activation by follicular androstenedione concentrations suggests that androgen activation of ovarian SLC39A9 may regulate ovulatory changes in the mammalian follicle.
Collapse
Affiliation(s)
- Megan A G Sage
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Diane M Duffy
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| |
Collapse
|
2
|
Perry GA, Ketchum JN, Quail LK. Importance of preovulatory estradiol on uterine receptivity and luteal function. Anim Reprod 2023; 20:e20230061. [PMID: 37720725 PMCID: PMC10503890 DOI: 10.1590/1984-3143-ar2023-0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/18/2023] [Indexed: 09/19/2023] Open
Abstract
Animals that exhibited estrus had greater pregnancy success compared to animals that did not exhibit estrus before fixed-time AI (FTAI). Estradiol is synthesized in bovine ovarian follicles under gonadotropin regulation and can directly and indirectly regulate the uterine receptivity and luteal function. Estradiol concentrations at FTAI impacted oviductal gene expression and has been reported to play an important role in establishing the timing of uterine receptivity. These changes have been reported to impact uterine pH and sperm transport to the site of fertilization. After fertilization, preovulatory estradiol has been reported to improve embryo survival likely by mediating changes in uterine blood flow, endometrial thickness and changes in histotroph. Cows with greater estradiol concentrations at the time of GnRH-induced ovulation also had a larger dominant follicle size and greater circulating progesterone concentrations on day 7. Therefore, it is impossible to accurately determine the individual benefit of greater estradiol concentrations prior to ovulation and greater progesterone concentrations following ovulation to pregnancy establishment, as these two measurements are confounded. Research has indicated an importance in the occurrence and timing of increasing preovulatory concentrations of estradiol, but increasing estradiol concentrations by supplementation may not be sufficient to increase fertility. Increased production of estradiol by the preovulatory follicle may be required to enhance fertility through the regulation of sperm transport, fertilization, oviductal secretions, the uterine environment, and embryo survival.
Collapse
Affiliation(s)
| | - Jaclyn Nicole Ketchum
- Texas A&M AgriLife Research, Overton, Texas, United States of America
- Department of Animal Science, Texas A&M University, College Station, Texas, United States of America
| | - Lacey Kay Quail
- Texas A&M AgriLife Research, Overton, Texas, United States of America
- Department of Animal Science, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
3
|
Zheng M, Cadenas J, Pors SE, Esa T, Kristensen SG, Mamsen LS, Adrados CS, Andersen CY. Reducing 3D Hydrogel Stiffness, Addition of Oestradiol in a Physiological Concentration and Increasing FSH Concentration Improve In Vitro Growth of Murine Preantral Follicles. Int J Mol Sci 2023; 24:12499. [PMID: 37569872 PMCID: PMC10419395 DOI: 10.3390/ijms241512499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
This study aimed to optimise culture conditions for murine preantral follicles to improve their growth and survival. Preantral follicles (diameter 100-130 µm) were isolated from prepubertal NMRI mice and individually cultured within alginate beads for 12 days. Three conditions were evaluated: (1) follicle re-encapsulation on day 6 of culture-reducing alginate concentration (0.5% to 0.25% w/v), (2) the presence of oestradiol (E2), and (3) increased follicle-stimulating hormone (FSH) concentration in the culture medium (from 10 to 100 mIU/mL FSH). Follicle morphology and growth, as well as anti-Müllerian hormone (AMH) production, were evaluated. From day 8, re-embedded follicles had a larger average diameter compared to follicles without alginate re-encapsulation (0.5% and 0.25% groups, p < 0.05). Oestradiol (1 µM) had a significantly positive effect on the mean follicular diameter and antrum formation (p < 0.001). Moreover, follicles cultured with 100 mIU/mL FSH showed faster growth (p < 0.05) and significantly higher antrum formation (p < 0.05) compared to the low FSH group. Nevertheless, AMH production was not affected by any of the culture conditions. In conclusion, the growth and survival of mouse preantral follicles during a 12-day period were improved by altering the alginate concentration midways during culture and adding E2 and FSH to the culture medium.
Collapse
Affiliation(s)
- Mengxue Zheng
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
- The Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jesús Cadenas
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
| | - Susanne Elisabeth Pors
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
| | - Tasnim Esa
- The Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800 Kongens Lyngby, Denmark;
| | - Stine Gry Kristensen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
| | - Linn Salto Mamsen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
| | - Cristina Subiran Adrados
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, 2100 Copenhagen, Denmark; (M.Z.); (J.C.); (S.E.P.); (S.G.K.); (L.S.M.); (C.S.A.)
- The Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Claus Yding Andersen
- The Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
4
|
Qin L, Sitticharoon C, Petyim S, Keadkraichaiwat I, Sririwichitchai R, Maikaew P, Churintaraphan M. A Longitudinal Study of the Relationship of Adiponectin with Reproduction in Infertile Women Undergoing IVF/ICSI Treatment, and an Experimental Study in Human Granulosa Cells. Life (Basel) 2023; 13:life13040994. [PMID: 37109523 PMCID: PMC10141627 DOI: 10.3390/life13040994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/17/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
This study investigated the roles of adiponectin in IVF treatment during Phase I (the basal stage before gonadotropin administration), Phase II (approximately 8 days after gonadotropin administration), and Phase III (on the ovum pick-up day), as well as the effects of adiponectin on CYP19A1 and the FSH receptor (FSHR) mRNA expression in a human granulosa-like tumor cell line (KGN). In human subjects (a longitudinal study, n = 30), blood samples were collected in all phases, while follicular fluid (FF) was only collected in Phase III. The participants were classified into successful and unsuccessful groups based on the determination of fetal heartbeats. KGN cells were treated with adiponectin/FSH/IGF-1 (an experimental study, n = 3). There was no difference in the adiponectin levels between successful and unsuccessful pregnancies in the FF (Phase III) and in serum (all phases), as well as among the three phases in both groups. Serum FSH (Phase I) was positively associated with serum adiponectin in the unsuccessful group, but it had a negative association in the successful group (all phases). Serum adiponectin and serum FSH (Phase I) were positively correlated in the unsuccessful group, whereas they were negatively correlated (all phases) in the successful group. The serum adiponectin levels (Phase III) were significantly higher than in the FF in unsuccessful pregnancies, but there was no difference in successful pregnancies. FF adiponectin concentrations were negatively correlated with serum LH in successful subjects. In KGN cells, adiponectin had no influence on CYP19A1 and FSHR mRNA expression. High adiponectin levels in serum compared to FF (Phase III) in unsuccessful subjects might negatively impact IVF treatment.
Collapse
Affiliation(s)
- Lixian Qin
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Chantacha Sitticharoon
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Somsin Petyim
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Issarawan Keadkraichaiwat
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Rungnapa Sririwichitchai
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Pailin Maikaew
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| | - Malika Churintaraphan
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Wanglang Rd., Siriraj, Bangkoknoi, Bangkok 10700, Thailand
| |
Collapse
|
5
|
Lv J, Ge W, Ding Z, Zeng J, Wang W, Duan H, Zhang Y, Zhao X, Hu J. Regulatory role of dihydrotestosterone on BMP-6 receptors in granular cells of sheep antral follicles. Gene 2022; 810:146066. [PMID: 34838638 DOI: 10.1016/j.gene.2021.146066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 10/08/2021] [Accepted: 11/16/2021] [Indexed: 11/19/2022]
Abstract
Bone morphogenetic protein-6 (BMP-6) and dihydrotestosterone (DHT) affect steroid synthesis in follicles and regulate cell proliferation in the ovaries of female animals. However, little is known about granular cells (GCs) in sheep. We identified the key BMP-6 receptors, activin receptor-like kinase(ALK-6), and bone morphogenetic protein receptor type 2 (BMPRII) in sheep follicles using immunohistochemistry (IHC) and immunofluorescence (IF). Both ALK-6 and BMPRII were expressed in the GC layer, GC membranes, and cytoplasm. We evaluated ALK-6 and BMPRII expression at the follicular development stage using quantitative real-time PCR and western blotting to detect sheep GCs from large, medium, and small follicles (diameters of ≥5, 2-5, and ≤2 mm, respectively). The mRNA abundance and protein expression of ALK-6 and BMPRII were significantly higher in GCs from large follicles compared to those in GCs from small follicles (P < 0.05) and were the lowest in GCs from medium follicles. To assess whether DHT affects ALK-6 and BMPRII expression in sheep GCs, we cultured GCs from large follicles in vitro then incubated them with DHT (10-11, 10-9, 10-7 M). We found that 10-7-M DHT significantly inhibited ALK-6 and BMPRII mRNA and protein (P < 0.05). We further explored whether DHT regulates ALK-6 and BMPRII through the nuclear androgen receptor (AR) pathway and found that 10-6-M flutamide, a non-selective androgen inhibitor, partially relieved the inhibitory effect of 10-7-M DHT on ALK-6 and BMPRII expression. Thus, GCs in sheep antral follicles differentially expressed ALK-6 and BMPRII at various stages, indicating that BMP-6 plays different roles to some extent during the development of antral follicles, and that high concentrations of DHT can inhibit the expression of ALK-6 and BMPRII via the androgen receptor pathway in sheep GCs. The present study aimed to determine the expression of the main BMP-6-related main receptors, namely, ALK-6 and BMPRII, during the development of GCs in sheep antral follicles and a potential mechanism of DHT regulation in sheep GCs. Our findings lay a foundation for the further exploration of the effects of ovarian BMP-6 expression on follicular development.
Collapse
Affiliation(s)
- Jianshu Lv
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, China
| | - Wenbo Ge
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, China
| | - Ziqiang Ding
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, China
| | - Jianlin Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, China
| | - Wenjuan Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, China
| | - Hongwei Duan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, China
| | - Yong Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, China
| | - Xingxu Zhao
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, China.
| | - Junjie Hu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China; Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, China.
| |
Collapse
|
6
|
Dai XX, Jiang ZY, Wu YW, Sha QQ, Liu Y, Ding JY, Xi WD, Li J, Fan HY. CNOT6/6L-mediated mRNA degradation in ovarian granulosa cells is a key mechanism of gonadotropin-triggered follicle development. Cell Rep 2021; 37:110007. [PMID: 34788619 DOI: 10.1016/j.celrep.2021.110007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 07/27/2021] [Accepted: 10/26/2021] [Indexed: 12/13/2022] Open
Abstract
CCR4-NOT deadenylase is a major regulator of mRNA turnover. It contains two heterogeneous catalytic subunits CNOT7/8 and CNOT6/6L in vertebrates. The physiological function of each catalytic subunit is unclear due to the gene redundancy. In this study, Cnot6/6l double knockout mice are generated. Cnot6l-/- female mice are infertile, with poor ovarian responses to gonadotropins. Follicle-stimulating hormone (FSH) stimulates the transcription and translation of Cnot6 and Cnot6l in ovarian granulosa cells. CNOT6/6L function as key effectors of FSH in granulosa cells and trigger the clearance of specific transcripts in granulosa cells during preantral to antral follicle transition. These results demonstrate that FSH modulates granulosa cell function by stimulating selective translational activation and degradation of existing mRNAs, in addition to inducing de novo gene transcription. Meanwhile, this study provides in vivo evidence that CNOT6/6L-mediated mRNA deadenylation is dispensable in most somatic cell types, but is essential for female reproductive endocrine regulation.
Collapse
Affiliation(s)
- Xing-Xing Dai
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Zhi-Yan Jiang
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Yun-Wen Wu
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Qian-Qian Sha
- Fertility Preservation Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Yang Liu
- Department of Assisted Reproduction, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong 226000, China
| | - Jia-Yi Ding
- Department of Assisted Reproduction, Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong 226000, China
| | - Wen-Dong Xi
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211100, China
| | - Jing Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 211100, China
| | - Heng-Yu Fan
- Life Sciences Institute, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
7
|
Qin L, Sitticharoon C, Petyim S, Keadkraichaiwat I, Sririwichitchai R, Maikeaw P, Churintaraphan M, Sripong C. Roles of kisspeptin in IVF/ICSI-treated infertile women and in human granulosa cells. Exp Biol Med (Maywood) 2020; 246:996-1010. [PMID: 33327782 DOI: 10.1177/1535370220981006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Kisspeptin, a crucial central regulator of reproduction, has been used as a trigger in in vitro fertilization (IVF) treatment. This study aimed to investigate the roles of kisspeptin in IVF treatment in infertile females (n = 30); and in steroidogenesis in human granulosa-like tumor cell line (KGN). In the human study, blood was collected at three time points including (1) the beginning of gonadotropin stimulation (Phase I), (2) around eight days after gonadotropin stimulation (Phase II), and (3) on the day of ovum pick-up (Phase III). Follicular fluid (FF) was collected at Phase III. Serum human chorionic gonadotropin (hCG) was measured 15 days after embryo transfer and fetal heart beats were determined around 42 days of menstrual cycle to classify the subjects into successful and unsuccessful groups. FF kisspeptin levels were higher in successful compared with unsuccessful subjects (P < 0.01). Kisspeptin levels were significantly higher in FF than in serum in successful subjects (P < 0.05) but were comparable in unsuccessful subjects. Serum kisspeptin was comparable among three phases in the successful group but its levels in Phase III were significantly lower compared with Phase I in the unsuccessful group (P < 0.01). Serum kisspeptin in Phase II/III had positive correlations with serum E2 in Phases II and III and the outcomes of IVF/intracytoplasmic sperm injection (ICSI) treatment including serum hCG levels. For the cell experiment (n = 3), kisspeptin treatment in the presence of FSH together with IGF-1 enhanced CYP19A1 (aromatase) mRNA expression compared with control. FSH alone increased aromatase concentrations in the supernatant compared with control and kisspeptin at the dose of 10-2 mmol/L with FSH enhanced aromatase concentrations in the supernatant compared with FSH alone (P < 0.001 all). In conclusion, kisspeptin enhanced aromatase expression and secretion and was associated with positive outcomes of IVF/ICSI treatment. Further studies regarding supplementation of kisspeptin could reveal its beneficial effects on IVF/ICSI treatment.
Collapse
Affiliation(s)
- Lixian Qin
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chantacha Sitticharoon
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Somsin Petyim
- Department of Obstetrics and Gynecology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Issarawan Keadkraichaiwat
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Rungnapa Sririwichitchai
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Pailin Maikeaw
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Malika Churintaraphan
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chanakarn Sripong
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
8
|
Fàbregues F, Solernou R, Ferreri J, Guimerá M, Peralta S, Casals G, Peñarrubia J, Creus M, Manau D. Comparison of GnRH agonist versus luteal estradiol GnRH antagonist protocol using transdermal testosterone in poor responders. JBRA Assist Reprod 2019; 23:130-136. [PMID: 30614665 PMCID: PMC6501741 DOI: 10.5935/1518-0557.20180090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Objective: Transdermal testosterone has been used in different doses and in different
stimulation protocols in poor responders. The aim of the present study is to
compare the luteal estradiol/GnRH antagonists protocol
versus long GnRH agonists in poor responder patients
according to the Bologna criteria, in which transdermal testosterone has
been used prior to the stimulation with gonadotropins. Methods: In this retrospective analysis, a total of 141 poor responder patients
according to the Bologna criteria were recruited. All patients were treated
with transdermal testosterone preceding ovarian stimulation with
gonadotropins during 5 days. In 53 patients we used the conventional
antagonist protocol (Group 1). In 88 patients (GrH pituitary suppression was
achieved by leuprolide acetate according to the conventional long protocol
(Group 2). We analyzed the ovarian stimulation parameters and IVF
outcomes. Results: Comparing groups 1 and 2, there were no significant differences between
cancellation rates and number of oocytes retrieved. However the total
gonadotropin dose used and the mean length of stimulation were significantly
lower in group 1 when compared to group 2. There were no significant
differences in pregnancy outcomes; however, there was a slight increase in
the implantation rate in group 1 vis-a-vis group 2, although statistical
significance was not achieved. Conclusion: TT in poor responder patients can be effective both with the conventional
agonist's long protocol and with the conventional antagonist's protocol.
However, short regimes with previous estradiol antagonists in the luteal
phase facilitate ovarian stimulation by shortening the days of treatment and
the consumption of gonadotropins
Collapse
Affiliation(s)
- Francesc Fàbregues
- Institut Clinic de Ginecologia, Obstetricia y Neonatología (ICGON). Hospital Clinic de Barcelona. Institut de Investigacions Biomédiques August Pi iSunyer (IDIBAPS)
| | - Roser Solernou
- Institut Clinic de Ginecologia, Obstetricia y Neonatología (ICGON). Hospital Clinic de Barcelona. Institut de Investigacions Biomédiques August Pi iSunyer (IDIBAPS)
| | - Janisse Ferreri
- Institut Clinic de Ginecologia, Obstetricia y Neonatología (ICGON). Hospital Clinic de Barcelona. Institut de Investigacions Biomédiques August Pi iSunyer (IDIBAPS)
| | - Marta Guimerá
- Institut Clinic de Ginecologia, Obstetricia y Neonatología (ICGON). Hospital Clinic de Barcelona. Institut de Investigacions Biomédiques August Pi iSunyer (IDIBAPS)
| | - Sara Peralta
- Institut Clinic de Ginecologia, Obstetricia y Neonatología (ICGON). Hospital Clinic de Barcelona. Institut de Investigacions Biomédiques August Pi iSunyer (IDIBAPS)
| | - Gemma Casals
- Institut Clinic de Ginecologia, Obstetricia y Neonatología (ICGON). Hospital Clinic de Barcelona. Institut de Investigacions Biomédiques August Pi iSunyer (IDIBAPS)
| | - Joana Peñarrubia
- Institut Clinic de Ginecologia, Obstetricia y Neonatología (ICGON). Hospital Clinic de Barcelona. Institut de Investigacions Biomédiques August Pi iSunyer (IDIBAPS)
| | - Montserrat Creus
- Institut Clinic de Ginecologia, Obstetricia y Neonatología (ICGON). Hospital Clinic de Barcelona. Institut de Investigacions Biomédiques August Pi iSunyer (IDIBAPS)
| | - Dolors Manau
- Institut Clinic de Ginecologia, Obstetricia y Neonatología (ICGON). Hospital Clinic de Barcelona. Institut de Investigacions Biomédiques August Pi iSunyer (IDIBAPS)
| |
Collapse
|
9
|
Shen M, Cao Y, Jiang Y, Wei Y, Liu H. Melatonin protects mouse granulosa cells against oxidative damage by inhibiting FOXO1-mediated autophagy: Implication of an antioxidation-independent mechanism. Redox Biol 2018; 18:138-157. [PMID: 30014903 PMCID: PMC6068202 DOI: 10.1016/j.redox.2018.07.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 06/24/2018] [Accepted: 07/06/2018] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress has been described as a prime driver of granulosa cell (GCs) death during follicular atresia. Increasing evidence suggests potential roles of melatonin in protecting GCs from oxidative injury, though the underlying mechanisms remain largely undetermined. Here we first proposed that the inhibition of autophagy through some novel regulators contributes to melatonin-mediated GCs survival under conditions of oxidative stress. Oxidant-induced loss of GCs viability was significantly reduced after melatonin administration, which was correlated with attenuated autophagic signals upon oxidative stimulation both in vivo and in vitro. Compared with melatonin treatment, suppression of autophagy displayed similar preventive effect on GCs death during oxidative stress, but melatonin provided no additional protection in GCs pretreated with autophagy inhibitors. Notably, we found that melatonin-directed regulation of autophagic death was independent of its antioxidation/radical scavenging ability. Further investigations identified FOXO1 as a critical downstream effector of melatonin in promoting GCs survival from oxidative stress-induced autophagy. Specifically, suppression of FOXO1 via the melatonin-phosphatidylinositol 3-kinase (PI3K)-AKT axis not only improved GCs resistance to oxidative stress, but also abolished the autophagic response, from genes expression to the formation of autophagic vacuoles. Moreover, the activation of SIRT1 signaling was required for melatonin-mediated deacetylation of FOXO1 and its interaction with ATG proteins, as well as the inhibition of autophagic death in GCs suffering oxidative stress. These findings reveal a brand new mechanism of melatonin in defense against oxidative damage to GCs by repressing FOXO1, which may be a potential therapeutic target for anovulatory disorders.
Melatonin inhibits oxidative damage in GC without scavenging oxidative stress itself. Melatonin protects GC from oxidative damage via inhibiting autophagic cell death. Inhibition of FOXO1-dependent autophagy by melatonin reduces oxidative damage in GC. Suppression of autophagy through melatonin-PI3K-AKT-FOXO1 axis improves GC survival. Melatonin reduces oxidative injury by inhibiting SIRT1-FOXO1-ATG7-dependent autophagy.
Collapse
Affiliation(s)
- Ming Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yan Cao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yi Jiang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Yinghui Wei
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
10
|
Abstract
Androgen production by the ovary is an essential requirement for normal cyclical secretion of estradiol but its physiological role extends to important actions on both preantral and antral follicle development, including promotion of granulosa cell proliferation. It is likely only in mature antral follicles that androgens encourage apoptosis and consequent follicle atresia, and this may be an important mechanism to ensure mono-follicular ovulation in primates, including humans. Recent studies have provided new insight into the mechanism of androgen signaling in the ovary which involves both genomic and non-genomic effects that are complementary in effecting a cellular response. In polycystic ovary syndrome, a condition characterized by intra-ovarian androgen excess, aberrant development of both preantral and antral follicles is a salient feature. We present evidence that local action of androgens plays a part in such abnormalities. Finally, we review the role of androgens in follicle atresia and conclude that the effects are part of the normal physiology of follicle maturation.
Collapse
|
11
|
Shen M, Jiang Y, Guan Z, Cao Y, Li L, Liu H, Sun SC. Protective mechanism of FSH against oxidative damage in mouse ovarian granulosa cells by repressing autophagy. Autophagy 2017; 13:1364-1385. [PMID: 28598230 DOI: 10.1080/15548627.2017.1327941] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Oxidative stress-induced granulosa cell (GCs) death represents a common reason for follicular atresia. Follicle-stimulating hormone (FSH) has been shown to prevent GCs from oxidative injury, although the underlying mechanism remains to be elucidated. Here we first report that the suppression of autophagic cell death via some novel signaling effectors is engaged in FSH-mediated GCs protection against oxidative damage. The decline in GCs viability caused by oxidant injury was remarkably reduced following FSH treatment, along with impaired macroautophagic/autophagic flux under conditions of oxidative stress both in vivo and in vitro. Blocking of autophagy displayed similar levels of suppression in oxidant-induced cell death compared with FSH treatment, but FSH did not further improve survival of GCs pretreated with autophagy inhibitors. Further investigations revealed that activation of the phosphoinositide 3-kinase (PI3K)-AKT-MTOR (mechanistic target of rapamycin [serine/threonine kinase]) signaling pathway was required for FSH-mediated GCs survival from oxidative stress-induced autophagy. Additionally, the FSH-PI3K-AKT axis also downregulated the autophagic response by targeting FOXO1, whereas constitutive activation of FOXO1 in GCs not only abolished the protection from FSH, but also emancipated the autophagic process, from the protein level of MAP1LC3B-II to autophagic gene expression. Furthermore, FSH inhibited the production of acetylated FOXO1 and its interaction with Atg proteins, followed by a decreased level of autophagic cell death upon oxidative stress. Taken together, our findings suggest a new mechanism involving FSH-FOXO1 signaling in defense against oxidative damage to GCs by restraining autophagy, which may be a potential avenue for the clinical treatment of anovulatory disorders.
Collapse
Affiliation(s)
- Ming Shen
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Yi Jiang
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Zhiqiang Guan
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Yan Cao
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Liechuan Li
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Honglin Liu
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| | - Shao-Chen Sun
- a College of Animal Science and Technology , Nanjing Agricultural University , Nanjing , China
| |
Collapse
|
12
|
Laird M, Thomson K, Fenwick M, Mora J, Franks S, Hardy K. Androgen Stimulates Growth of Mouse Preantral Follicles In Vitro: Interaction With Follicle-Stimulating Hormone and With Growth Factors of the TGFβ Superfamily. Endocrinology 2017; 158:920-935. [PMID: 28324051 PMCID: PMC5460807 DOI: 10.1210/en.2016-1538] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/13/2017] [Indexed: 01/06/2023]
Abstract
Androgens are essential for the normal function of mature antral follicles but also have a role in the early stages of follicle development. Polycystic ovary syndrome (PCOS), the most common cause of anovulatory infertility, is characterized by androgen excess and aberrant follicle development that includes accelerated early follicle growth. We have examined the effects of testosterone and dihydrotestosterone (DHT) on development of isolated mouse preantral follicles in culture with the specific aim of investigating interaction with follicle-stimulating hormone (FSH), the steroidogenic pathway, and growth factors of the TGFβ superfamily that are known to have a role in early follicle development. Both testosterone and DHT stimulated follicle growth and augmented FSH-induced growth and increased the incidence of antrum formation among the granulosa cell layers of these preantral follicles after 72 hours in culture. Effects of both androgens were reversed by the androgen receptor antagonist flutamide. FSH receptor expression was increased in response to both testosterone and DHT, as was that of Star, whereas Cyp11a1 was down-regulated. The key androgen-induced changes in the TGFβ signaling pathway were down-regulation of Amh, Bmp15, and their receptors. Inhibition of Alk6 (Bmpr1b), a putative partner for Amhr2 and Bmpr2, by dorsomorphin resulted in augmentation of androgen-stimulated growth and modification of androgen-induced gene expression. Our findings point to varied effects of androgen on preantral follicle growth and function, including interaction with FSH-activated growth and steroidogenesis, and, importantly, implicate the intrafollicular TGFβ system as a key mediator of androgen action. These findings provide insight into abnormal early follicle development in PCOS.
Collapse
Affiliation(s)
- Mhairi Laird
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Kacie Thomson
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Mark Fenwick
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Jocelyn Mora
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Kate Hardy
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London, United Kingdom
| |
Collapse
|
13
|
Larimore EL, Amundson OL, Bridges GA, McNeel AK, Cushman RA, Perry GA. Changes in ovarian function associated with circulating concentrations of estradiol before a GnRH-induced ovulation in beef cows. Domest Anim Endocrinol 2016; 57:71-9. [PMID: 27565233 DOI: 10.1016/j.domaniend.2016.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/29/2016] [Accepted: 06/01/2016] [Indexed: 11/29/2022]
Abstract
These studies were conducted to evaluate causes for differences in circulating concentrations of estradiol before a GnRH-induced ovulation. Beef cows were synchronized by an injection of GnRH on day -7 and an injection of prostaglandin F2α (PGF2α) on day 0. In experiment 1, blood samples were collected every 3 h from PGF2α on day 0 to hour 33 after PGF2α and at slaughter (hour 36 to 42; n = 10). Cows were assigned to treatment group based on circulating concentrations of estradiol (E2): HighE2 vs LowE2. At slaughter, follicular fluid (FF) and granulosa cells were collected from the dominant follicle. In experiment 2, blood samples (n = 30) were collected every 8 h from PGF2α until the dominant follicle was aspirated via ultrasound-guided follicular aspiration to collect FF and granulosa cells (hour 38 to 46). In experiment 1, HighE2 had increased abundance of 3β-hydroxysteroid dehydrogenase, cytochrome P450 aromatase, and LHR (P ≤ 0.02), and greater concentrations of estradiol and androstenedione (P ≤ 0.02) in the FF. In experiment 2, HighE2 had increased abundance of CYP11A1, 3β-hydroxysteroid dehydrogenase, cytochrome P450 aromatase, and LHR (P ≤ 0.03) vs either LowE2 or GnRHLowE2. There was a tendency (P = 0.07) for LH pulse frequency to be increased in both the GnRHLowE2 and HighE2 compared with LowE2. HighE2 cows experienced increas in circulating concentrations of estradiol compared with LowE2. In conclusion, animals with greater concentrations of circulating estradiol before fixed-time AI experienced an upregulation of the steroidogenic pathway during the preovulatory period.
Collapse
Affiliation(s)
- E L Larimore
- Department of Animal Science, South Dakota State University, Brookings, SD, USA
| | - O L Amundson
- Department of Animal Science, South Dakota State University, Brookings, SD, USA
| | - G A Bridges
- North Central Research and Outreach Center, University of Minnesota, Grand Rapids, MN, USA
| | - A K McNeel
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE, USA
| | - R A Cushman
- USDA, ARS, U.S. Meat Animal Research Center, Clay Center, NE, USA
| | - G A Perry
- Department of Animal Science, South Dakota State University, Brookings, SD, USA.
| |
Collapse
|
14
|
Gal A, Lin PC, Cacioppo JA, Hannon PR, Mahoney MM, Wolfe A, Fernandez-Valdivia R, Lydon JP, Elias CF, Ko C. Loss of Fertility in the Absence of Progesterone Receptor Expression in Kisspeptin Neurons of Female Mice. PLoS One 2016; 11:e0159534. [PMID: 27441639 PMCID: PMC4956300 DOI: 10.1371/journal.pone.0159534] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 07/05/2016] [Indexed: 11/19/2022] Open
Abstract
Ovarian steroids, estradiol and progesterone, play central roles in regulating female reproduction by acting as both positive and negative regulators of gonadotropin-releasing hormone (GnRH) secretion in the hypothalamus. Recent studies have identified kisspeptin neurons of the hypothalamus as the target of estrogenic regulation of GnRH secretion. In this study, we aimed to determine the significance of progesterone receptor (PGR) expression in the kisspeptin neurons. To this end, the Pgr gene was selectively ablated in mouse kisspeptin neurons and the reproductive consequence assessed. The hypothalamus of the Pgr deficient female mouse expressed kisspeptin, the pituitary released LH in response to GnRH stimulation, and the ovary ovulated when stimulated with gonadotropins. However, the mutant mouse gradually lost cyclicity, was unable to generate a LH surge in response to rising estradiol, and eventually became infertile. Taken together, these results indicate that the loss of PGR impairs kisspeptin secretory machinery and therefore that PGR plays a critical role in regulating kisspeptin secretion.
Collapse
Affiliation(s)
- Arnon Gal
- Institute of Veterinary, Animal and Biomedical Sciences, Massey University, Palmerston North, New Zealand
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Po-Ching Lin
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Joseph A. Cacioppo
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Patrick R. Hannon
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Megan M. Mahoney
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| | - Andrew Wolfe
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Rodrigo Fernandez-Valdivia
- Department of Pathology and Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States of America; Tumor Biology Microenvironment Program, Karmanos Cancer Institute, Detroit, MI, United States of America
| | - John P. Lydon
- Center of Reproductive Medicine and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America
| | - Carol F. Elias
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, United States of America
| | - CheMyong Ko
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| |
Collapse
|
15
|
Casarini L, Riccetti L, De Pascali F, Nicoli A, Tagliavini S, Trenti T, La Sala GB, Simoni M. Follicle-stimulating hormone potentiates the steroidogenic activity of chorionic gonadotropin and the anti-apoptotic activity of luteinizing hormone in human granulosa-lutein cells in vitro. Mol Cell Endocrinol 2016; 422:103-114. [PMID: 26690776 DOI: 10.1016/j.mce.2015.12.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 12/07/2015] [Accepted: 12/08/2015] [Indexed: 01/15/2023]
Abstract
Luteinizing hormone (LH) and choriogonadotropin (hCG) are glycoprotein hormones regulating ovarian function and pregnancy, respectively. Since these molecules act on the same receptor (LHCGR), they were traditionally assumed as equivalent in assisted reproduction techniques (ART), although differences between LH and hCG were demonstrated at molecular and physiological level. In this study, we demonstrated for the first time that co-treatment with a follicle-stimulating hormone (FSH) dose in the ART therapeutic range potentiates different LH- and hCG-dependent responses in vitro, measured in terms of cAMP, phospho-CREB, -ERK1/2 and -AKT activation, gene expression, progesterone and estradiol production in human granulosa-lutein cells (hGLC). We show that in the presence of FSH, hCG biopotency is about 5-fold increased, in the presence of FSH, in terms of cAMP activation. Accordingly, CREB phosphorylation and steroid production is increased under hCG and FSH co-treatment. LH effects, evaluated as steroidogenic cAMP/PKA pathway activation, do not change in the presence of FSH, which, however, increases LH-dependent ERK1/2 and AKT, but not CREB phosphorylation, resulting in anti-apoptotic effects. The different modulatory activity of FSH on LH and hCG action in vitro corresponds to their different physiological functions, reflecting proliferative effects exerted by LH during the follicular phase and before trophoblast development, and the high steroidogenic potential of hCG requested to sustain pregnancy from the luteal phase onwards.
Collapse
Affiliation(s)
- Livio Casarini
- Unit of Endocrinology, Dept. Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for the Genomic Research, University of Modena and Reggio Emilia, Modena, Italy.
| | - Laura Riccetti
- Unit of Endocrinology, Dept. Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco De Pascali
- Unit of Endocrinology, Dept. Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessia Nicoli
- Unit of Obstetrics and Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy
| | | | | | - Giovanni Battista La Sala
- Unit of Obstetrics and Gynecology, IRCCS-Arcispedale Santa Maria Nuova, Reggio Emilia, Italy; Dept. of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Reggio Emilia, Italy
| | - Manuela Simoni
- Unit of Endocrinology, Dept. Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for the Genomic Research, University of Modena and Reggio Emilia, Modena, Italy; Dept. of Medicine, Endocrinology, Metabolism and Geriatrics, Azienda USL, Modena, Italy
| |
Collapse
|
16
|
Chakraborty P, Roy SK. Effect of azaline B on follicular development and functions in the hamster. Mol Cell Endocrinol 2015; 400:1-9. [PMID: 25462584 PMCID: PMC4274241 DOI: 10.1016/j.mce.2014.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 11/18/2014] [Accepted: 11/19/2014] [Indexed: 01/08/2023]
Abstract
The usefulness of azaline B, a GnRH antagonist, in suppressing gonadotropin secretion in the golden hamster was examined by examining follicular development, steroidogenesis and expression of steroidogenic enzymes. Serum levels of P and E declined significantly, while FSH or LH was undetectable in azaline B-treated hamsters. FSH significantly increased serum E levels, whereas LH upregulated serum P levels. The formation of antral follicles ceased in azaline-treated hamsters, but was reversed by FSH with or without LH supplement. FSH also activated the primordial follicle pool resulting in increased formation of primary and preantral follicles. Further, an increasing trend in the formation of preantral follicles in response to E or E + P, and the formation of antral follicles in response to E + P treatment was evident. The level of Cyp11a1 mRNA increased markedly in LH- or LH + FSH-treated hamsters, whereas FSH with or without LH upregulated Cyp17a1, Cyp19a1 and Fshr mRNA expression. E without or with P also upregulated ovarian Cyp19a1 mRNA expression. The expression of enzyme protein corroborated the mRNA data. In summary, azaline B is an efficient GnRH antagonist in the hamster, and will be useful in studying the selective effect of gonadotropins on ovarian functions without disrupting the physiological functions of other hormones in ovarian cells.
Collapse
Affiliation(s)
- Prabuddha Chakraborty
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Shyamal K Roy
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Obstetrics and Gynecology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
17
|
Abstract
The current study aims to compare cycle outcomes of two ovarian stimulation protocols in poor responders according to the Bologna criteria; luteal estrogen priming (LE) or letrozole (LZ) co-treatment in antagonist protocol. Following retrospective chart review of a single center, 162 cycles were found eligible for the comparison of two ovarian stimulation protocols. After interpreting data, significantly higher serum estradiol levels, longer duration of cycle, higher number of fertilized oocytes and good quality embryos were detected in patients who received LE. Despite any statistical significance, higher clinical pregnancy rate (CPR) and ongoing pregnancy rate (OPR) per embryo transfer (ET) were detected with LE protocol compared with LZ (12.3% versus 18.2% and 9.6% versus 12.7%, respectively). Younger patients (<40 years) revealed higher CPR and OPR per started cycle compared with older patients (≥40 years) where only OPR was statistically significant. Multivariable analysis demonstrated that basal antral follicle count, peak serum estradiol levels and number of fertilized oocytes were independent variables significantly associated with clinical pregnancies (p < 0.05). In the current analysis, LE or LZ protocols revealed comparable but quite low pregnancy rates in poor responders according to the Bologna criteria. Younger patients were more likely to achieve pregnancy compared to older patients with both protocols.
Collapse
Affiliation(s)
- Emre Göksan Pabuccu
- a Department of Obstetrics and Gynecology, Faculty of Medicine , Ufuk University , Ankara , Turkey and
| | - Gamze Sinem Caglar
- a Department of Obstetrics and Gynecology, Faculty of Medicine , Ufuk University , Ankara , Turkey and
| | - Recai Pabuccu
- a Department of Obstetrics and Gynecology, Faculty of Medicine , Ufuk University , Ankara , Turkey and
- b Centrum Clinic Assisted Reproductive Technology Unit , Ankara , Turkey
| |
Collapse
|
18
|
Lazaros L, Pamporaki C, Vlahos N, Takenaka A, Kitsou C, Kosmas I, Sofikitis N, Stefos T, Zikopoulos K, Hatzi E, Georgiou I. The follicular outcome after standard gonadotropin stimulation is associated with ERα and ERβ genotypes. Endocrine 2014; 47:930-5. [PMID: 24705910 DOI: 10.1007/s12020-014-0249-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 03/21/2014] [Indexed: 10/25/2022]
Abstract
The aim is to study the association of estrogen receptor α (ERα) and estrogen receptor β (ERβ) gene polymorphisms and diplotypes with ovarian response to follicle-stimulating hormone (FSH) stimulation and the hormone levels [FSH, luteinizing hormone (LH), E2] at the third day of the menstrual cycle. Three hundred women undergoing medically assisted reproduction and 300 women with at least one spontaneous pregnancy participated in the study. ERα PvuII and XbaI polymorphisms as well as ERβ AluI polymorphism were genotyped. The FSH, LH, and E2 levels were determined at the third day of the menstrual cycle, while the follicular size, the follicle, and oocyte numbers were recorded during oocyte retrieval. PvuII CC, XbaI GG, and AluI GG women were characterized by increased amounts of large follicles compared to PvuII TT, XbaI AA, and AluI AA women (p = 0.045, 0.01, and 0.033, respectively). The PvuII CC/XbaI GG diplotype was also significantly associated with higher large follicle numbers compared to the PvuII TT/XbaI AA diplotype (p = 0.024). However, no associations were observed between ER gene polymorphisms and the hormonal profile, the follicle/oocyte numbers, and the pregnancy rates. ERα and ERβ genetic variants were associated with ovarian response to standard gonadotropin stimulation of women undergoing in-vitro fertilization affecting mainly the follicular growth.
Collapse
Affiliation(s)
- Leandros Lazaros
- Genetics and IVF Unit, Department of Obstetrics and Gynecology, Medical School, Ioannina University, 45110, Ioannina, Greece
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Involvement of FoxO1 in the effects of follicle-stimulating hormone on inhibition of apoptosis in mouse granulosa cells. Cell Death Dis 2014; 5:e1475. [PMID: 25321482 PMCID: PMC4237239 DOI: 10.1038/cddis.2014.400] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/11/2014] [Accepted: 08/14/2014] [Indexed: 12/23/2022]
Abstract
In mammalian ovaries, follicular atresia occurs periodically and destroys almost all the follicles in the ovary. Follicle-stimulating hormone (FSH) acts as the primary survival factor during follicular atresia by preventing apoptosis in granulosa cells. FoxO1 is a critical factor in promoting follicular atresia and granulosa cell apoptosis. FSH inhibits the induction of FoxO1. In this report, we investigated the role of FSH-FoxO1 pathway in mouse follicular atresia. FSH dampened stress-induced apoptosis and the expression of FoxO1 and pro-apoptosis genes in mouse granulosa cells (MGCs). In contrast, overexpression of FoxO1 inhibited the viability of MGCs and induced the expression of endogenous FoxO1. The signaling cascades involved in regulating FoxO1 activity upon FSH treatment were identified using FSH signaling antagonists. Blocking protein kinase A (PKA), phosphatidylinositol-3 kinase (PI3K) or protein kinase B (AKT) restored the upregulation of FoxO1 and apoptotic signals, which was suppressed by FSH. Moreover, inhibition of PKA or PI3K impaired FSH-induced AKT activity, but inactivation of PI3K or AKT had little effect on PKA activity in the presence of FSH. Correspondingly, constitutive activation of FoxO1 (all three AKT sites were replaced by alanines) also promoted MGC apoptosis despite FSH administration. Furthermore, both luciferase reporter assays and chromatin immunoprecipitation assays showed that FoxO1 directly bound to a FoxO-recognized element site within the FoxO1 promoter and contributed to the regulation of FoxO1 expression in response to FSH. Taken together, we propose a novel model in which FSH downregulates FoxO1-dependent apoptosis in MGCs by coordinating the PKA-PI3K-AKT-FoxO1 axis and FoxO1-FoxO1 positive feedback.
Collapse
|
20
|
Silva M, Ulloa-Leal C, Norambuena C, Fernández A, Adams G, Ratto M. Ovulation-inducing factor (OIF/NGF) from seminal plasma origin enhances Corpus Luteum function in llamas regardless the preovulatory follicle diameter. Anim Reprod Sci 2014; 148:221-7. [DOI: 10.1016/j.anireprosci.2014.05.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 05/21/2014] [Accepted: 05/28/2014] [Indexed: 10/25/2022]
|
21
|
Calder M, Chan YM, Raj R, Pampillo M, Elbert A, Noonan M, Gillio-Meina C, Caligioni C, Bérubé NG, Bhattacharya M, Watson AJ, Seminara SB, Babwah AV. Implantation failure in female Kiss1-/- mice is independent of their hypogonadic state and can be partially rescued by leukemia inhibitory factor. Endocrinology 2014; 155:3065-78. [PMID: 24877624 PMCID: PMC4098000 DOI: 10.1210/en.2013-1916] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The hypothalamic kisspeptin signaling system is a major positive regulator of the reproductive neuroendocrine axis, and loss of Kiss1 in the mouse results in infertility, a condition generally attributed to its hypogonadotropic hypogonadism. We demonstrate that in Kiss1(-/-) female mice, acute replacement of gonadotropins and estradiol restores ovulation, mating, and fertilization; however, these mice are still unable to achieve pregnancy because embryos fail to implant. Progesterone treatment did not overcome this defect. Kiss1(+/-) embryos transferred to a wild-type female mouse can successfully implant, demonstrating the defect is due to maternal factors. Kisspeptin and its receptor are expressed in the mouse uterus, and we suggest that it is the absence of uterine kisspeptin signaling that underlies the implantation failure. This absence, however, does not prevent the closure of the uterine implantation chamber, proper alignment of the embryo, and the ability of the uterus to undergo decidualization. Instead, the loss of Kiss1 expression specifically disrupts embryo attachment to the uterus. We observed that on the day of implantation, leukemia inhibitory factor (Lif), a cytokine that is absolutely required for implantation in mice, is weakly expressed in Kiss1(-/-) uterine glands and that the administration of exogenous Lif to hormone-primed Kiss1(-/-) female mice is sufficient to partially rescue implantation. Taken together, our study reveals that uterine kisspeptin signaling regulates glandular Lif levels, thereby identifying a novel and critical role for kisspeptin in regulating embryo implantation in the mouse. This study provides compelling reasons to explore this role in other species, particularly livestock and humans.
Collapse
|
22
|
Plewka D, Marczyński J, Morek M, Bogunia E, Plewka A. Receptors of hypothalamic-pituitary-ovarian-axis hormone in uterine myomas. BIOMED RESEARCH INTERNATIONAL 2014; 2014:521313. [PMID: 25050358 PMCID: PMC4090522 DOI: 10.1155/2014/521313] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 05/14/2014] [Accepted: 05/26/2014] [Indexed: 11/18/2022]
Abstract
In this study the expression of GnRH, FSH, LH, ER-α, ER-β, and PR receptors was examined in uterine myomas of women in reproductive and perimenopausal age. In cases of GnRH and tropic hormones a membranous and cytoplasmic immunohistochemical reaction was detected, in cases of ER-α and PR the reaction was located in cell nucleus, and in the case of ER-β it manifested also a cytoplasmic location. In some of the examined cases the expression was detected in endometrium, myocytes, and endothelium of blood vessels, in uterine glands and myoma cells. In myometrium the level of GnRH and LH receptors increases with age, whereas the level of progesterone and both estrogen receptors decreases. In myomas of women in reproductive age, independently of their size, expression of GnRH, FSH, and LH receptors was more pronounced than in myometrium. In women of perimenopausal age, independently of myoma size, expression of LH and estrogen α receptors was higher while expression of GnRH receptors was lower than in myometrium. FSH receptor expression was not observed. Expression of estrogen receptor β was not affected by age of the woman or size of myoma. Analysis of obtained results indicates on existing in small myomas local feedback axis between GnRH-LH-progesterone.
Collapse
Affiliation(s)
- Danuta Plewka
- Department of Cytophysiology, Chair of Histology and Embryology, Medical University of Silesia, Medykow 18 Street, 40-752 Katowice, Poland
| | - Jacek Marczyński
- Department of Proteomics, Medical University of Silesia, Jednosci Street 8, 41-200 Sosnowiec, Poland
| | - Michał Morek
- Department of Proteomics, Medical University of Silesia, Jednosci Street 8, 41-200 Sosnowiec, Poland
| | - Edyta Bogunia
- Department of Proteomics, Medical University of Silesia, Jednosci Street 8, 41-200 Sosnowiec, Poland
| | - Andrzej Plewka
- Department of Proteomics, Medical University of Silesia, Jednosci Street 8, 41-200 Sosnowiec, Poland
| |
Collapse
|
23
|
Expression of extracellular matrix components is disrupted in the immature and adult estrogen receptor β-null mouse ovary. PLoS One 2012; 7:e29937. [PMID: 22253831 PMCID: PMC3254630 DOI: 10.1371/journal.pone.0029937] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 12/08/2011] [Indexed: 01/01/2023] Open
Abstract
Within the ovary, Estrogen Receptor β (ERβ) is localized to the granulosa cells of growing follicles. 17β-estradiol (E2) acting via ERβ augments the actions of follicle stimulating hormone in granulosa cells, leading to granulosa cell differentiation and formation of a preovulatory follicle. Adult ERβ-null females are subfertile and possess ovaries with reduced numbers of growing follicles and corpora lutea. Because the majority of E2 production by granulosa cells occurs once puberty is reached, a role for ERβ in the ovary prior to puberty has not been well examined. We now provide evidence that lack of ERβ disrupts gene expression as early as post-natal day (PND) 13, and in particular, we identify a number of genes of the extracellular matrix (ECM) that are significantly higher in ERβ-null follicles than in wildtype (WT) follicles. Considerable changes occur to the ECM occur during normal folliculogenesis to allow for the dramatic growth, cellular differentiation, and reorganization of the follicle from the primary to preovulatory stage. Using quantitative PCR and immunofluorescence, we now show that several ECM genes are aberrantly overexpressed in ERβ-null follicles. We find that Collagen11a1, a protein highly expressed in cartilage, is significantly higher in ERβ-null follicles than WT follicles as early as PND 13, and this heightened expression continues through PND 23–29 into adulthood. Similarly, Nidogen 2, a highly conserved basement membrane glycoprotein, is elevated in ERβ-null follicles at PND 13 into adulthood, and is elevated specifically in the ERβ-null focimatrix, a basal lamina-like matrix located between granulosa cells. Focimatrix laminin and Collagen IV expression were also higher in ERβ-null ovaries than in WT ovaries at various ages. Our findings suggest two novel observations: a) that ERβ regulates granulosa cell gene expression ovary prior to puberty, and b) that ERβ regulates expression of ECM components in the mouse ovary.
Collapse
|
24
|
Chang EM, Han JE, Won HJ, Kim YS, Yoon TK, Lee WS. Effect of estrogen priming through luteal phase and stimulation phase in poor responders in in-vitro fertilization. J Assist Reprod Genet 2011; 29:225-30. [PMID: 22160464 DOI: 10.1007/s10815-011-9685-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 11/20/2011] [Indexed: 10/14/2022] Open
Abstract
PURPOSE To verify whether a novel protocol administering E(2) during the luteal phase of the preceding cycle and during ovarian stimulation in GnRH antagonist cycle could enhance follicular response and hence improve outcomes in poor responders. METHODS In this retrospective analysis, a total of 155 poor responder patients subjected to IVF/ICSI were analyzed. All the patients had history of more than one prior IVF cycle failure with poor response (less than 5 oocytes retrieved and/or maximal E₂ level less than 500 pg/mL) by using conventional long agonist or antagonist protocol. In luteal E2 treatment protocol (n = 86), oral estradiol valerate 4 mg/day was initiated on luteal day 21 and either stopped at menstrual cycle day 3 (Protocol A, n = 28) or continued during the period of ovarian stimulation until the day of hCG injection (Protocol B, n = 58). IVF parameters and pregnancy outcome of luteal E2 treatments group were compared with a standard GnRH antagonist protocol (n = 69) which the patients received no hormonal pretreatment. RESULTS Compared to standard GnRH antagonist protocol, cancellation rate was lower with luteal E2 group (15.1% vs 37.7%, p < 0.01). Moreover, patients treated with luteal estrogen resulted in an increased number of oocytes retrieved (4.5 ± 2.9 vs 3.2 ± 1.9; p < 0.01). A trend toward increase in number of normally fertilized embryos (2.9 ± 2.1vs 2.3 ± 1.9; p = 0.043), and increased prevalence of good quality embryos (51.2% vs 25%; p = 0.047) were noted. Comparing protocol A and B, there were no significant difference between embryologic data, however there were slight increase in ongoing pregnancy rate in protocol B compared to A (27.1% vs 20%, p = 0.357), although statistical significance was not achieved. CONCLUSION Estrogen priming through luteal phase and stimulation phase improved ovarian responsiveness and this may lead to an increase in pregnancy rate in poor responders with failed cycle.
Collapse
Affiliation(s)
- Eun Mi Chang
- Fertility Center of CHA Gangnam Medical Center, Department of Obstetrics and Gynecology, College of Medicine, CHA University, 650-9 Yeoksam, Kangnamgu, Seoul 135-081, Korea
| | | | | | | | | | | |
Collapse
|
25
|
Silvestre F, Boni R, Fissore RA, Tosti E. Ca2+ signaling during maturation of cumulus-oocyte complex in mammals. Mol Reprod Dev 2011; 78:744-56. [PMID: 21656870 DOI: 10.1002/mrd.21332] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 05/11/2011] [Indexed: 11/06/2022]
Abstract
Under the influence of gonadotropins or growth factors, a close cooperation develops between cumulus cells and the oocyte that is implicated in transmitting signals involved in maintaining or releasing the meiotic arrest in the oocyte. While cyclic adenosine 5'-monophosphate (cAMP) is a key molecule in maintaining the meiotic arrest, calcium (Ca(2+)) may play a role in controlling either spontaneous or gonadotropin-induced oocyte maturation, possibly by modulating intracytoplasmic cAMP concentrations via Ca(2+)-sensitive adenylate cyclases. This review focuses on the mechanisms related to the origin of the Ca(2+) wave that travels from the cumulus cells to the oocyte, and discusses the source of variations affecting the dynamics of this wave.
Collapse
Affiliation(s)
- F Silvestre
- Animal Physiology and Evolution Laboratory, Stazione Zoologica Anton Dohrn, Naples, Italy.
| | | | | | | |
Collapse
|
26
|
Development of in vitro culture method for early stage zebrafish (Danio rerio) ovarian follicles for use in cryopreservation studies. Theriogenology 2010; 74:290-303. [DOI: 10.1016/j.theriogenology.2010.02.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Revised: 02/03/2010] [Accepted: 02/05/2010] [Indexed: 11/21/2022]
|
27
|
Rizzo A, Minoia G, Trisolini C, Mutinati M, Spedicato M, Jirillo F, Sciorsci RL. Reactive oxygen species (ROS): involvement in bovine follicular cysts etiopathogenesis. Immunopharmacol Immunotoxicol 2010; 31:631-5. [PMID: 19874233 DOI: 10.3109/08923970902932962] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Ovulation is compared to an acute inflammatory process during which vasoactive agents, prostanoids, leukotrienes and Reactive Oxygen Species (ROS) develop. The aim of this study was to evaluate the levels of ROS in cystic and follicular fluid, in order to establish their involvement in the etiopathogenesis of Cystic Ovarian Follicle (COF) in dairy cows. The study was conducted in 30 healthy cows (group C) and 30 cows affected by COF (group COF). The fluid of follicular cysts and of preovulatory follicles was drawn by means of ultrasound guided aspiration from the cows of both groups. The fluid obtained was analyzed by a photometric analytical system to detect ROS level. ROS concentration was statistically lower in the cystic fluid than in the follicular one (62.4 +/- 13.36 U.Carr vs. 84.89 +/- 26.99 U.Carr) (p<0.05), thus suggesting that an alteration of the cascade responsible for ROS production may be implicated in the complex etipathogenesis of COF.
Collapse
Affiliation(s)
- Annalisa Rizzo
- Department of Animal Production, University of Bari, Bari, Italy
| | | | | | | | | | | | | |
Collapse
|
28
|
Zia-Ur-Rahman, Bukhari SA, Ahmad N, Akhtar N, Ijaz A, Yousaf MS, Haq IU. Dynamics of Follicular Fluid in One-humped Camel (Camelus dromedarius). Reprod Domest Anim 2008; 43:664-71. [DOI: 10.1111/j.1439-0531.2007.00967.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Estrogen receptor subtypes localization shifts in cultured mouse ovarian follicles. Histochem Cell Biol 2008; 129:827-40. [DOI: 10.1007/s00418-008-0408-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2008] [Indexed: 10/22/2022]
|
30
|
Ulloa-Aguirre A, Zariñán T, Pasapera AM, Casas-González P, Dias JA. Multiple facets of follicle-stimulating hormone receptor function. Endocrine 2007; 32:251-63. [PMID: 18246451 DOI: 10.1007/s12020-008-9041-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2007] [Revised: 01/04/2008] [Accepted: 01/14/2008] [Indexed: 10/22/2022]
Abstract
Follicle-stimulating hormone (FSH) is a glycoprotein hormone produced by the anterior pituitary gland. This gonadotropin plays an essential role in reproduction. Its receptor (FSHR) belongs to the superfamily of G protein-coupled receptors (GPCR), specifically the family of rhodopsin-like receptors. Agonist binding to the FSHR triggers the rapid activation of multiple signaling cascades, mainly the cAMP-adenylyl cyclase-protein kinase A cascade, that impact diverse biological effects of FSH in the gonads. As in other G protein-coupled receptors, the several cytoplasmic domains of the FSHR are involved in signal transduction and termination of the FSH signal. Here we summarize some recent information on the signaling cascades activated by FSH as well as on the role of the intracytoplasmic domains of the FSHR in coupling to membrane and cytosolic proteins linked to key biological functions regulated by the FSH-FSHR system.
Collapse
Affiliation(s)
- Alfredo Ulloa-Aguirre
- Research Unit in Reproductive Medicine, Hospital de Ginecobstetricia "Luis Castelazo Ayala", Instituto Mexicano del Seguro Social, Apartado Postal 99-065, Unidad Independencia, C.P. 10101 Mexico, D.F., Mexico.
| | | | | | | | | |
Collapse
|
31
|
Vanholder T, Opsomer G, de Kruif A. Aetiology and pathogenesis of cystic ovarian follicles in dairy cattle: a review. ACTA ACUST UNITED AC 2006; 46:105-19. [PMID: 16597418 DOI: 10.1051/rnd:2006003] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Accepted: 12/05/2005] [Indexed: 11/14/2022]
Abstract
Cystic ovarian follicles (COF) are an important ovarian dysfunction and a major cause of reproductive failure in dairy cattle. Due to the complexity of the disorder and the heterogeneity of the clinical signs, a clear definition is lacking. A follicle becomes cystic when it fails to ovulate and persists on the ovary. Despite an abundance of literature on the subject, the exact pathogenesis of COF is unclear. It is generally accepted that disruption of the hypothalamo-pituitary-gonadal axis, by endogenous and/or exogenous factors, causes cyst formation. Secretion of GnRH/LH from the hypothalamus-pituitary is aberrant, which is attributed to insensitivity of the hypothalamus-pituitary to the positive feedback effect of oestrogens. In addition, several factors can influence GnRH/LH release at the hypothalamo-pituitary level. At the ovarian level, cellular and molecular changes in the growing follicle may contribute to anovulation and cyst formation, but studying follicular changes prior to cyst formation remains extremely difficult. Differences in receptor expression between COF and dominant follicles may be an indication of the pathways involved in cyst formation. The genotypic and phenotypic link of COF with milk yield may be attributed to negative energy balance and the associated metabolic and hormonal adaptations. Altered metabolite and hormone concentrations may influence follicle growth and cyst development, both at the level of the hypothalamus-pituitary and the ovarian level.
Collapse
Affiliation(s)
- Tom Vanholder
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | | | | |
Collapse
|
32
|
Toyama R, Fujimori T, Nabeshima Y, Itoh Y, Tsuji Y, Osamura RY, Nabeshima YI. Impaired regulation of gonadotropins leads to the atrophy of the female reproductive system in klotho-deficient mice. Endocrinology 2006; 147:120-9. [PMID: 16179415 DOI: 10.1210/en.2005-0429] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
klotho-Deficient mice exhibit a syndrome resembling human premature ageing, with multiple pathological phenotypes in tissues including reproductive organs. It was proposed that Klotho might possess the hormonal effects on many organs. In this study, the female reproductive system of klotho mice was examined to reveal the mechanism that brought the female sterility by histological and molecular approaches. We observed cessation of ovarian follicular maturation at the preantral stage and the presence of numerous atretic ovarian follicles and atrophic uteri. In situ hybridization analysis revealed that LH receptor and aromatase P450 were not expressed in the ovaries. These results suggest the impairment of gonadal development during the antral transition process. We next addressed the responsible organs for the failure of antral transition. Transplantation of klotho ovaries to wild-type mice resulted in the ability to bear offspring. Administration of FSH or GnRH induced advanced maturation of ovaries and uteri in klotho mice. These results indicate that the female reproductive organs in klotho mice are potentially functional and that klotho gene deficiency leads to the atrophy of reproductive organs via impairment of the hypothalamic-pituitary axis. Absence of the estrus cycle and constant low trends of both FSH and LH levels were found in female klotho mice. Immunohistochemical analysis revealed that the production of both FSH and LH were decreased in pituitary gland. Taken together, our findings suggest the involvement of klotho in the regulatory control of pituitary hormones.
Collapse
Affiliation(s)
- Ryusuke Toyama
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | | | | | | | | | | | | |
Collapse
|
33
|
Kolibianakis EM, Papanikolaou EG, Fatemi HM, Devroey P. Estrogen and folliculogenesis: is one necessary for the other? Curr Opin Obstet Gynecol 2005; 17:249-53. [PMID: 15870558 DOI: 10.1097/01.gco.0000169101.83342.96] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW During folliculogenesis the primordial follicle undergoes several steps of maturation in order to develop into a preovulatory follicle. The exact role of estrogen during this process has not yet been fully assessed. RECENT FINDINGS Estrogen appears to regulate cyclic gonadotropin release via its action on estrogen receptor alpha in the hypothalamus/hypophysis axis and to enhance folliculogenesis through its actions via estrogen receptor beta in the ovary. In addition, a role of estrogen during the very early stages of folliculogenesis is possible. However, it is likely that oocyte quality and developmental potential are not estrogen dependent. This might explain the lack of association between estrogen and in-vitro fertilization outcome in humans. SUMMARY The advent of knockout mice models has enhanced our understanding of the role of estrogen during folliculogenesis. Existing data suggest that estrogen might be involved in the very early steps of this process, but its role in sustaining ovulation is mainly central.
Collapse
|
34
|
Emmen JMA, Couse JF, Elmore SA, Yates MM, Kissling GE, Korach KS. In vitro growth and ovulation of follicles from ovaries of estrogen receptor (ER){alpha} and ER{beta} null mice indicate a role for ER{beta} in follicular maturation. Endocrinology 2005; 146:2817-26. [PMID: 15731357 DOI: 10.1210/en.2004-1108] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Both estrogen receptor (ER) alpha and beta are expressed within the ovary and lack of either of these receptors affects ovarian function. In this study, the role of ERalpha and ERbeta in folliculogenesis and ovulation was further analyzed. Evaluation of ovarian follicle populations in wild-type and ERbeta knockout (betaERKO) ovaries revealed reduced late antral growth and ovulatory capacity of betaERKO follicles, indicated by reduced numbers of large antral follicles and corpora lutea and increased atresia of large antral follicles. An in vitro culture system was used to study growth, rupture, and luteinization of wild-type, ERalpha knockout (alphaERKO) and betaERKO ovarian follicles. alphaERKO follicles exhibited wild-type-like growth and ovulation rates but an increased capacity to synthesize estradiol. In contrast, betaERKO follicles showed a significant lack of progression from early antral to large antral stage, decreased estradiol production, and reduced ovulation. Expression patterns of several genes involved in follicle maturation and ovulation were analyzed in follicles grown in vitro. Ar, Pgr, and Has2 mRNA expression levels were the same among the three genotypes. However, betaERKO follicles showed reduced expression of Cyp19 mRNA during follicle maturation and reduced Lhcgr and Ptgs2 mRNA expression after human chorionic gonadotropin stimulus. Luteinization occurs normally in alphaERKO and betaERKO follicles, shown by increased progesterone secretion and increased cdkn1b mRNA expression after human chorionic gonadotropin. Collectively, these data indicate that ERbeta, but not ERalpha, plays a direct role in folliculogenesis. ERbeta appears to facilitate follicle maturation from the early antral to the preovulatory stage.
Collapse
Affiliation(s)
- Judith M A Emmen
- Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences/National Institutes of Health, MD B3-02, P.O. Box 12233, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | | | |
Collapse
|
35
|
Perry GA, Smith MF, Lucy MC, Green JA, Parks TE, MacNeil MD, Roberts AJ, Geary TW. Relationship between follicle size at insemination and pregnancy success. Proc Natl Acad Sci U S A 2005; 102:5268-73. [PMID: 15795381 PMCID: PMC556005 DOI: 10.1073/pnas.0501700102] [Citation(s) in RCA: 313] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2004] [Indexed: 11/18/2022] Open
Abstract
Administration of gonadotropin-releasing hormone (GnRH) induces a surge of luteinizing hormone and ovulation in a variety of species, including human beings. Our objectives were to determine the effect of follicle size at the time of ovulation on corpus luteum function and establishment and maintenance of pregnancy in cows in which ovulation was either spontaneous or induced with GnRH. GnRH-induced ovulation of follicles < or approximately = 11 mm in diameter resulted in decreased pregnancy rates and increased late embryonic mortality. This decrease in fertility was associated with lower circulating concentrations of estradiol on the day of insemination, a decreased rate of increase in progesterone after insemination, and, ultimately, decreased circulating concentrations of progesterone. In contrast, ovulatory follicle size had no apparent effect on fertility when ovulation occurred spontaneously. Follicles undergoing spontaneous ovulation do so at a wide range of sizes when they are physiologically mature. Therefore, administration of GnRH to induce ovulation likely initiates a preovulatory gonadotropin surge before some dominant follicles attain physiological maturity. GnRH-induced ovulation of follicles that are physiologically immature has a negative impact on pregnancy rates and late embryonic/fetal survival. These observations in cattle may have implications for assisted reproductive procedures in human beings.
Collapse
Affiliation(s)
- George A Perry
- Department of Animal Sciences, University of Missouri, Columbia, MO 65211, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Pasapera AM, Jiménez-Aguilera MDP, Chauchereau A, Milgrom E, Olivares A, Uribe A, Gutiérrez-Sagal R, Ulloa-Aguirre A. Effects of FSH and 17beta-estradiol on the transactivation of estrogen-regulated promoters and cell proliferation in L cells. J Steroid Biochem Mol Biol 2005; 94:289-302. [PMID: 15857748 DOI: 10.1016/j.jsbmb.2004.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2004] [Accepted: 11/29/2004] [Indexed: 11/26/2022]
Abstract
In the present study, we analyzed human follicle-stimulating hormone (FSH)-induced cell proliferation and transactivation of estrogen-sensitive reporter genes-in L cells stably expressing the human FSH receptor [L-(hFSHR(+)) cells]. In order to dissect the signaling pathways involved in this process, L-(hFSHR(+)) cells were transiently transfected with either the 3X-ERE-TAT-Luc or the ERE-VitA2-TK-CAT reporter genes and treated with FSH or PKA activators (cholera toxin, forskolin and 8-Br-cAMP) in the presence or absence of various kinase inhibitors. We found that FSH and all PKA activators, specifically induced transactivation of both reporter genes. Transactivation of estrogen-sensitive genes by FSH or PKA activators were blocked (approximately 90%) by H89 (PKA inhibitor) and LY294002 but not by Wortmannin (PI3-K inhibitors), 4-OH-tamoxifen, ICI182,780 or SB203580 (p38 MAPK inhibitor); PD98059 (ERK1/2 inhibitor) partially (approximately 30%) blocked the FSH-mediated effect. The combination of FSH and estradiol resulted in a synergistic effect on transactivation as well as on cell proliferation, and this enhancement was attenuated by antiestrogens. We additionally analyzed the participation of the coactivators SRC-1 and cAMP response element binding protein (CREB)-binding protein (CBP) in FSH-evoked estrogen receptor (ER)-dependent transactivation; we found that CBP but not SRC-1 potentiated FSH-induced transcriptional activation of both ER-sensitive reporters, being this effect stronger on the ERE-VitA2-TK-CAT than on the 3X-ERE-TAT-Luc reporter. Thus, in L-(hFSHR(+)) cells FSH induces transcriptional activation of estrogen-sensitive genes through an A-kinase-triggered signaling pathway, using also to a lesser extent the ERK1/2 and p38 pathways. PI3-K is not apparently involved in this FSH-mediated process since LY294002, but not Wortmannin, specifically binds ERs and completely blocks estrogen action. Presumably, CBP cooperates with the ER on genes that contain estrogen responsive elements through mechanisms involving the participation of other proteins and/or basal transcription factors (e.g. CREB), which in turn mediate the transcriptional response of estrogen-sensitive reporter genes to FSH stimulation.
Collapse
Affiliation(s)
- Ana María Pasapera
- Research Unit in Reproductive Medicine, Hospital de Gineco-Obstetricia Luis Castelazo Ayala, Instituto Mexicano del Seguro Social, Apdo. Postal 99-065, Unidad Independencia, México D.F. C.P. 10101, Mexico
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Son DS, Arai KY, Roby KF, Terranova PF. Tumor necrosis factor alpha (TNF) increases granulosa cell proliferation: dependence on c-Jun and TNF receptor type 1. Endocrinology 2004; 145:1218-26. [PMID: 14617571 DOI: 10.1210/en.2003-0860] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TNF alpha has significant in vitro effects on steroidogenesis and folliculogenesis and reproductive alterations occur in TNF receptor type 1 (TNFR1) knockout mice. The present study investigated the effect of in vitro TNF on granulosa cell proliferation from immature mice at 28 d of age, with emphasis on intracellular signaling that regulates granulosa cell proliferation. TNF dose dependently increased granulosa cell proliferation and the proto-oncogene c-Jun protein. However, other Jun family members such as JunD was expressed constitutively and JunB was not expressed. In vitro TNF did not increase c-Jun and proliferation in granulosa cells from TNFR1 knockout mice. The time course of TNF-induced c-Jun revealed biphasic patterns of short-term (3 h) and long-term (24 h) induction. The time courses of Ser63- and Ser73-phospho c-Jun coincided with changes in total c-Jun. Among MAPK cascades, stress-activated protein kinase/c-Jun-NH(2)-teminal kinase signaling was increased transiently in TNF-treated cells, whereas p38MAPK and ERK1 and 2 were not changed. In addition, overexpression of nuclear factor-kappa B and addition of ceramide and 8-bromo-cAMP did not increase c-Jun or proliferation. Antisense oligonucleotides for c-Jun blocked cell proliferation induced by TNF. In conclusion, the above results demonstrate that TNF increased c-Jun by activating stress-activated protein kinase/c-Jun-NH(2)-teminal kinase signaling via TNFR1 in mouse granulosa cells, and the induced c-Jun resulted in increased cell proliferation.
Collapse
Affiliation(s)
- Deok-Soo Son
- Center of Reproductive Sciences, and Department of Molecualr and Integrative Physiology, Unversity of Kansas Medical Center, Kansas City, Kansas 66160, USA
| | | | | | | |
Collapse
|
38
|
Huynh K, Jones G, Thouas G, Britt KL, Simpson ER, Jones MEE. Estrogen is not directly required for oocyte developmental competence. Biol Reprod 2003; 70:1263-9. [PMID: 14695903 DOI: 10.1095/biolreprod.103.022111] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Oocyte maturation and ovulation require a coordinated interaction between gonadotrophs, steroid hormones, and growth factors. The extent to which estrogen is required in this process, however, remains unclear. To better understand the role of estrogen in maintaining developmental competence of mammalian oocytes, we studied the Aromatase knockout (ArKO) mouse, which has been genetically engineered to be incapable of synthesizing endogenous estrogen. Previous studies have established that ArKO female mice are anovulatory with ovaries that progressively degenerate, developing hemorrhagic cystic follicles. In young ArKO females, however, apparently healthy follicles and oocytes have been observed. We investigated if these oocytes could be induced to ovulate, then mature, fertilize, and develop in vitro. Following a standard superovulation protocol, ArKO oocytes did not ovulate. When recovered manually from the ovary, however, ArKO oocytes successfully progressed through in vitro maturation, fertilization, and development to the blastocyst stage at the same rate as wild-type and heterozygote littermates. Therefore, it appears that estrogen is not required for the production and growth of oocytes capable of maturation and complete preimplantation development but is required for continued follicle growth and feedback regulation of ovulation.
Collapse
Affiliation(s)
- Kim Huynh
- Department of Anatomy, Monash University, Clayton, Victoria 3800, Australia
| | | | | | | | | | | |
Collapse
|
39
|
Hu Y, Cortvrindt R, Smitz J. Effects of aromatase inhibition on in vitro follicle and oocyte development analyzed by early preantral mouse follicle culture. Mol Reprod Dev 2002; 61:549-59. [PMID: 11891927 DOI: 10.1002/mrd.10107] [Citation(s) in RCA: 226] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
In vivo studies on folliculogenesis have documented a relation among intrafollicular steroid content, follicle growth, and oocyte development. This study examined how profound changes in androgen/estrogen ratio would affect mouse in vitro follicular development. Arimidex, a potent follicular aromatase inhibitor was used for this purpose. Early preantral follicles were cultured for 12 days up to the preovulatory stage. Oocyte's meiotic maturation, spindle and chromosome configurations, in vitro fertilization and preimplantation embryo development were evaluated. Compared to controls, Arimidex reduced E2 concentration in follicle culture medium by a factor 1000, and an expected simultaneous accumulation of testosterone was measured in the conditioned medium. Arimidex treatment provoked a dose-dependent earlier differentiation of the granulosa cells as judged by an earlier antrallike cavity formation and slightly elevated basal progesterone secretion. Follicle survival exceeded 98% in all groups and all follicles responded normally to HCG/EGF addition on day 12 by cumulus mucification. By the HCG ovulatory challenge, progesterone output was reduced in Arimidex supplemented groups suggesting preovulatory luteinization. These results indicate that in vitro mouse follicles can develop normally under very low levels of estrogens and that a local androgen increase by a factor 3 is not atretogenic. Oocyte growth did not differ among culture conditions. Arimidex treatment induced a dose dependent enhancement of GVBD and polar body formation rate in response to HCG at the end of culture. Spindle and chromosome analyses demonstrated that in all groups, 90% of the oocytes which extruded a polar body had also reached the MII stage. While most of the cultured MII oocytes had a normal spindle and well aligned chromosomes, significantly less oocytes were fertilized in the groups cultured in the presence of Arimidex. Once fertilized, however, there was found to be no difference for preimplantation embryo development between controls and Arimidex treatment. These data suggest that in mice a pronounced estrogenic environment is not essential for in vitro folliculogenesis. Drastic changes in the intrafollicular steroid concentrations do not disrupt meiotic maturation nor compromise early preimplantation development, but adversely affect fertilization of in vitro grown oocytes.
Collapse
Affiliation(s)
- Y Hu
- Follicle Biology Laboratory, Brussels Free University (Dutch Speaking), Laarbeeklaan, Brussels, Belgium
| | | | | |
Collapse
|
40
|
Palter SF, Tavares AB, Hourvitz A, Veldhuis JD, Adashi EY. Are estrogens of import to primate/human ovarian folliculogenesis? Endocr Rev 2001; 22:389-424. [PMID: 11399749 DOI: 10.1210/edrv.22.3.0433] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The notion that estrogens play a meaningful role in ovarian folliculogenesis stems from a large body of in vitro and in vivo experiments carried out in certain rodent models, (e.g., rats) wherein the stimulatory role of estrogen on granulosa cell growth and differentiation is undisputed. However, evidence derived from these polyovulatory species may not be readily generalizable to the monoovulatory subhuman primates, let alone the human. Only recently, significant observations on the ovarian role(s) of estrogen have been reported for the primate/human. It is thus the objective of this communication to review the evidence for and against a role for estrogens in primate/human ovarian follicular development with an emphasis toward the application of the concepts so developed to contemporary reproductive physiology and to the practice of reproductive medicine. The role(s) of estrogens will be examined not only by analyzing the physiological evidence to the effect that these hormones control ovarian function and follicular growth, but also by summarizing the molecular evidence for the existence and distribution of the cognate receptors.
Collapse
Affiliation(s)
- S F Palter
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | |
Collapse
|
41
|
Cushman RA, DeSouza JC, Hedgpeth VS, Britt JH. Effect of long-term treatment with recombinant bovine somatotropin and estradiol on hormone concentrations and ovulatory response of superovulated cattle. Theriogenology 2001; 55:1533-47. [PMID: 11354712 DOI: 10.1016/s0093-691x(01)00500-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The objective was to assess effects of long-term treatment with recombinant bovine somatotropin (bST) and estradiol-17beta (E2) on the number of follicles that ovulated in response to FSH. Non-lactating Holstein and Jersey cows (Trial 1, n=27) and Angus cows and heifers (Trial 2, n=35) received two ear implants of E2 and biweekly injections of bST in a 2 x 2 arrangement of treatments. Estradiol implants were removed 74.6 +/- 1.1 d after insertion and 18.1 +/- 0.9 d after the last biweekly injection of bST. Cows were stimulated with FSH-P beginning 2 d after removal of E2 implants, and PGF2alpha (PGF) was given on the third day of FSH treatment. Ovaries were collected to determine the number of CL at 1 to 2 wk after treatment with PGF. In Trial 2 only, cattle were inseminated at estrus and embryos were collected 6 to 8 d later. Implants of E2 increased (P < 0.01) serum E2 8-fold initially and E2 was still elevated 5-fold at removal of implants. Injections of bST increased (P < 0.01) serum growth hormone (GH) 15-fold and insulin-like growth factor-I (IGF-I) 3-fold. In Trial 1, number of CL was increased by the combination of bST+E2 (P < 0.01). In Trial 2, E2 increased the number of CL (P < 0.05), and bST increased the number of total ova and transferable embryos (P < 0.01). We conclude that long-term treatment with bST and E2 may interact to enhance follicular development and ovulatory response to FSH.
Collapse
Affiliation(s)
- R A Cushman
- Department of Anatomy, Physiological Sciences & Radiology, North Carolina State University Raleigh, USA
| | | | | | | |
Collapse
|
42
|
Couse JF, Curtis Hewitt S, Korach KS. Receptor null mice reveal contrasting roles for estrogen receptor alpha and beta in reproductive tissues. J Steroid Biochem Mol Biol 2000; 74:287-96. [PMID: 11162937 DOI: 10.1016/s0960-0760(00)00105-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- J F Couse
- Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, MD B3-02, PO Box 12233, 27709, Research Triangle Park, NC, USA
| | | | | |
Collapse
|
43
|
Curtis Hewitt S, F Couse J, S Korach K. Estrogen receptor transcription and transactivation: Estrogen receptor knockout mice: what their phenotypes reveal about mechanisms of estrogen action. Breast Cancer Res 2000; 2:345-52. [PMID: 11250727 PMCID: PMC138656 DOI: 10.1186/bcr79] [Citation(s) in RCA: 108] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2000] [Revised: 03/09/2000] [Accepted: 05/26/2000] [Indexed: 11/10/2022] Open
Abstract
Natural, synthetic and environmental estrogens have numerous effects on the development and physiology of mammals. Estrogen is primarily known for its role in the development and functioning of the female reproductive system. However, roles for estrogen in male fertility, bone, the circulatory system and immune system have been established by clinical observations regarding sex differences in pathologies, as well as observations following menopause or castration. The primary mechanism of estrogen action is via binding and modulation of activity of the estrogen receptors (ERs), which are ligand-dependent nuclear transcription factors. ERs are found in highest levels in female tissues critical to reproduction, including the ovaries, uterus, cervix, mammary glands and pituitary gland. Since other affected tissues have extremely low levels of ER, indirect effects of estrogen, for example induction of pituitary hormones that affect the bone, have been proposed. The development of transgenic mouse models that lack either estrogen or ER have proven to be valuable tools in defining the mechanisms by which estrogen exerts its effects in various systems. The aim of this article is to review the mouse models with disrupted estrogen signaling and describe the associated phenotypes.
Collapse
Affiliation(s)
| | - John F Couse
- National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Kenneth S Korach
- National Institutes of Health, Research Triangle Park, North Carolina, USA
| |
Collapse
|
44
|
Couse JF, Bunch DO, Lindzey J, Schomberg DW, Korach KS. Prevention of the polycystic ovarian phenotype and characterization of ovulatory capacity in the estrogen receptor-alpha knockout mouse. Endocrinology 1999; 140:5855-65. [PMID: 10579351 DOI: 10.1210/endo.140.12.7222] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ovarian-derived estradiol plays a critical endocrine role in the regulation of gonadotropin synthesis and secretion from the hypothalamic-pituitary axis. In turn, several para/autocrine effects of estrogen within the ovary are known, including increased ovarian weight, stimulation of granulosa cell growth, augmentation of FSH action, and attenuation of apoptosis. The estrogen receptor-alpha (ERalpha) is present in all three components of the hypothalamic-pituitary-ovarian axis of the mouse. In contrast, estrogen receptor-beta (ERbeta) is easily detectable in ovarian granulosa cells but is low to absent in the pituitary of the adult mouse. This distinct expression pattern for the two ERs suggests the presence of separate roles for each in the regulation of ovarian function. Herein, we definitively show that a lack of ERalpha in the hypothalamic-pituitary axis of the ERalpha-knockout (alphaERKO) mouse results in chronic elevation of serum LH and is the primary cause of the ovarian phenotype of polycystic follicles and anovulation. Prolonged treatment with a GnRH antagonist reduced serum LH levels and prevented the alphaERKO cystic ovarian phenotype. To investigate a direct role for ERalpha within the ovary, immature alphaERKO females were stimulated to ovulate with exogenous gonadotropins. Ovulatory capacity in the immature alphaERKO female was reduced compared with age-matched wild-type (14.5+/-2.9 vs. 40.6+/-2.6 oocytes/animal, respectively); however, oocytes collected from the alphaERKO were able to undergo successful in vitro fertilization. A similar discrepancy in oocyte yield was observed after superovulation of peripubertal (42 days) wild-type and alphaERKO females. In addition, ovaries from immature superovulated alphaERKO females possessed several ovulatory but unruptured follicles. Investigations of the possible reasons for the reduced number of ovulations in the alphaERKO included ribonuclease protection assays to assess the mRNA levels of several markers of follicular maturation and ovulation, including ERbeta, LH-receptor, cyclin-D2, P450-side chain cleavage enzyme, prostaglandin synthase-2, and progesterone receptor. No marked differences in the expression pattern for these mRNAs during the superovulation regimen were observed in the immature alphaERKO ovary compared with that of the wild-type. Serum progesterone levels just before ovulation were slightly lower in the alphaERKO compared with wild-type. These studies indicate that treatment of alphaERKO females with a GnRH antagonist decreased the serum LH levels to within the wild-type range and concurrently prevented development of the characteristic ovarian phenotype of cystic and hemorrhagic follicles. Furthermore, a lack of functional ERalpha within the ovary had no effect on the regulation of several genes required for follicular maturation and ovulation. However, the reduced numbers of ovulations following the administration of exogenous gonadotropins in the alphaERKO suggests an intraovarian role for ERalpha in follicular development and ovulation.
Collapse
Affiliation(s)
- J F Couse
- Receptor Biology Section, Laboratory of Reproductive and Developmental Toxicology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | | | | | | | |
Collapse
|
45
|
Abstract
All scientific investigations begin with distinct objectives: first is the hypothesis upon which studies are undertaken to disprove, and second is the overall aim of obtaining further information, from which future and more precise hypotheses may be drawn. Studies focusing on the generation and use of gene-targeted animal models also apply these goals and may be loosely categorized into sequential phases that become apparent as the use of the model progresses. Initial studies of knockout models often focus on the plausibility of the model based on prior knowledge and whether the generation of an animal lacking the particular gene will prove lethal or not. Upon the successful generation of a knockout, confirmatory studies are undertaken to corroborate previously established hypotheses of the function of the disrupted gene product. As these studies continue, observations of unpredicted phenotypes or, more likely, the lack of a phenotype that was expected based on models put forth from past investigations are noted. Often the surprising phenotype is due to the loss of a gene product that is downstream from the functions of the disrupted gene, whereas the lack of an expected phenotype may be due to compensatory roles filled by alternate mechanisms. As the descriptive studies of the knockout continue, use of the model is often shifted to the role as a unique research reagent, to be used in studies that 1) were not previously possible in a wild-type model; 2) aimed at finding related proteins or pathways whose existence or functions were previously masked; or 3) the subsequent effects of the gene disruption on related physiological and biochemical systems. The alpha ERKO mice continue to satisfy the confirmatory role of a knockout quite well. As summarized in Table 4, the phenotypes observed in the alpha ERKO due to estrogen insensitivity have definitively illustrated several roles that were previously believed to be dependent on functional ER alpha, including 1) the proliferative and differentiative actions critical to the function of the adult female reproductive tract and mammary gland; 2) as an obligatory component in growth factor signaling in the uterus and mammary gland; 3) as the principal steroid involved in negative regulation of gonadotropin gene transcription and LH levels in the hypothalamic-pituitary axis; 4) as a positive regulator of PR expression in several tissues; 5) in the positive regulation of PRL synthesis and secretion from the pituitary; 6) as a promotional factor in oncogene-induced mammary neoplasia; and 7) as a crucial component in the differentiation and activation of several behaviors in both the female and male. The list of unpredictable phenotypes in the alpha ERKO must begin with the observation that generation of an animal lacking a functional ER alpha gene was successful and produced animals of both sexes that exhibit a life span comparable to wild-type. The successful generation of beta ERKO mice suggests that this receptor is also not essential to survival and was most likely not a compensatory factor in the survival of the alpha ERKO. In support of this is our recent successful generation of double knockout, or alpha beta ERKO mice of both sexes. The precise defects in certain components of male reproduction, including the production of abnormal sperm and the loss of intromission and ejaculatory responses that were observed in the alpha ERKO, were quite surprising. In turn, certain estrogen pathways in the alpha ERKO female appear intact or unaffected, such as the ability of the uterus to successfully exhibit a progesterone-induced decidualization response, and the possible maintenance of an LH surge system in the hypothalamus. [ABSTRACT TRUNCATED]
Collapse
Affiliation(s)
- J F Couse
- Receptor Biology Section, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
46
|
Yokota H, Yamada K, Liu X, Kobayashi J, Abe Y, Mizunuma H, Ibuki Y. Paradoxical action of activin A on folliculogenesis in immature and adult mice. Endocrinology 1997; 138:4572-6. [PMID: 9348180 DOI: 10.1210/endo.138.11.5526] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Activin A is a gonadal protein originally isolated from follicular fluid and is recognized as a local regulator of granulosa cell differentiation. Whether activin A promotes folliculogenesis, however, still remains unclarified. The present study was designed to elucidate the effect of activin A on follicular growth in in vitro follicle culture systems. Preantral follicles, 100-120 microm in diameter, were mechanically isolated from BDF1 hybrid immature mice (11 days old) and adult mice (8 weeks old), then cultured for 4 days in a serum-free medium supplemented with activin A (100 ng/ml), FSH (100 mIU/ml), and a combination of both. Follicular diameter was measured daily, and the amount of estradiol and inhibin released at day 4 was determined by RIA. Preantral follicles collected from immature mice showed a significant increase in diameter when cultured with activin A or both activin A and FSH. FSH alone showed no significant effect on the diameter of follicles from immature mice. In contrast, the diameter of preantral follicles from adult mice significantly increased in response to FSH. Activin A did not stimulate growth of follicles from adult mice, and more interestingly, blocked the effect of FSH. The inhibitory action of activin A was in part restored by co-culture with follistatin (100 ng/ml). These results indicate that activin A is folliculogenetic in the prepubertal mouse, but not in adults.
Collapse
Affiliation(s)
- H Yokota
- Department of Obstetrics and Gynecology, Gunma University School of Medicine, Maebashi, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Evans AC, Fortune JE. Selection of the dominant follicle in cattle occurs in the absence of differences in the expression of messenger ribonucleic acid for gonadotropin receptors. Endocrinology 1997; 138:2963-71. [PMID: 9202241 DOI: 10.1210/endo.138.7.5245] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Mechanisms that allow selection of a dominant ovarian follicle from a cohort of growing follicles are unknown. Large healthy, estrogen-active follicles contain more LH receptors than atretic estrogen-inactive follicles, and levels of messenger RNA (mRNA) for LH receptor increase in the granulosa cells of dominant follicles as growth progresses. The aim of the present study was to test the hypothesis that changes in the temporal pattern of expression of mRNA for LH and FSH receptors are associated with selection of dominant follicles in cattle. Based on size, the dominant and two largest subordinate follicles were collected from the ovaries of heifers on days 2 (n = 3) or 3 (n = 3) of a follicular wave. On day 2, the dominant follicle was 1 mm larger than the largest subordinate follicle, but by day 3 of the wave the dominant follicle was 2-4 mm larger than the largest subordinate. Follicular fluid concentrations of estradiol and estradiol secretion in vitro by pieces of follicle wall (granulosa and theca cells) were greatest by the dominant compared with the subordinate follicles (P < 0.05). These data indicate that selection of a dominant follicle had occurred by the second day of the follicular wave. By in situ hybridization, mRNAs for LH and FSH receptors, P450 aromatase and P450 17alpha-hydroxylase (17alpha-OH) were localized in frozen sections from each follicle. The expression of mRNA for LH receptor in granulosa cells was always at or near background and was not different between days or follicle types (P = 0.63). In contrast, the expression of mRNA for LH receptor in theca cells of the same sections was readily detectable; there was no difference between follicle types on the second day of the follicular wave, but by the third day expression in the subordinate follicles had decreased (P < 0.05). The expression of mRNA for FSH receptor was highest in granulosa cells of dominant follicles collected on day 3 of the follicular wave (P < 0.05) and was not different between dominant and subordinate follicles on day 2 of the wave (P > 0.05). The expression of mRNA for aromatase in granulosa cells was similar (P > 0.05) between the dominant follicles on days 2 and 3 and the largest subordinate follicle on day 2 of the follicular wave and was much lower in the remaining follicles (P < 0.01). On day 2 of the wave, the expression of mRNA for 17alpha-OH was not different between the dominant and subordinate follicles, but by day 3 the dominant follicles had more mRNA for 17alpha-OH than the subordinate follicles (P < 0.05). These data show that the dominant follicle had been selected by the second day of the follicular wave (based on diameter and estradiol secretion) and that selection occurred in the absence of detectable levels of mRNA for LH receptor in the granulosa cells or differences between dominant and subordinate follicles in mRNA for LH receptor in theca cells or FSH receptor in granulosa cells. However, the divergent pattern of growth between dominant and subordinate follicles (after follicle selection) was associated with higher levels of mRNA for gonadotropin receptors and steroidogenic enzymes in dominant compared with subordinate follicles. Therefore, selection of the dominant follicle in cattle does not appear to involve the regulation of expression of mRNA for gonadotropin receptors, although such regulation may be important at other stages of differentiation of the dominant follicle.
Collapse
Affiliation(s)
- A C Evans
- Department of Physiology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, USA
| | | |
Collapse
|
48
|
McGee E, Spears N, Minami S, Hsu SY, Chun SY, Billig H, Hsueh AJ. Preantral ovarian follicles in serum-free culture: suppression of apoptosis after activation of the cyclic guanosine 3',5'-monophosphate pathway and stimulation of growth and differentiation by follicle-stimulating hormone. Endocrinology 1997; 138:2417-24. [PMID: 9165031 DOI: 10.1210/endo.138.6.5164] [Citation(s) in RCA: 82] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Progression of preantral follicle development is essential to further follicle maturation and ovulation, but there are few models for studying the regulation of preantral follicle survival and growth. We have evaluated preantral follicle survival in vivo and in vitro, and have developed a serum-free rat follicle culture system that can be used to characterize the regulation of preantral follicle growth and differentiation. Analysis of ovarian cell DNA fragmentation during the first wave of follicle growth in the infantile rat indicated negligible apoptosis up to day 16 of age. However, a major increase in apoptosis was found by day 18, a time point associated with the appearance of large antral follicles. In situ analysis confirmed that apoptotic DNA fragments were limited to antral follicles. Culture of individual preantral follicles mechanically dissected from ovaries of 12- or 14-day-old rats in serum-free conditions led to major increases in follicle cell apoptosis, similar to that seen in cultures of antral and preovulatory follicles. In contrast to antral and preovulatory follicles, treatment of preantral follicles with gonadotropins or cAMP analogs did not prevent apoptosis. However, treatment with 8-bromo-cGMP or 10% serum suppressed apoptosis by 75% in cultured preantral follicles. In situ analysis identified granulosa cells as the cell type susceptible to apoptosis regulation. Taking advantage of the ability of the cGMP analog to suppress apoptosis, we evaluated the potential of FSH as a growth factor. In the absence of serum, FSH treatment for 48 h did not affect follicle size compared to controls; however, treatment with the cGMP analog together with FSH increased follicle diameter (13%; P < 0.01) and viable cells (2.4-fold; P < 0.01) compared to control values. Immunoblot analysis further indicated that the inhibin-alpha content of the cultured follicles was increased by treatment with the combination of FSH and 8-bromo-cGMP, demonstrating the induction of follicle cell differentiation during culture. Therefore, we demonstrated that activation of the cGMP pathway promotes the survival of cultured preantral follicles and that in the presence of alpha cGMP analog, FSH is a growth and differentiation factor for preantral follicles. The present serum-free follicle culture model system will be useful in further evaluation of the regulation of growth and differentiation of preantral follicles.
Collapse
Affiliation(s)
- E McGee
- Department of Gynecology and Obstetrics, Stanford University School of Medicine, California 94305-5317, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Bley MA, Saragüeta PE, Barañao JL. Concerted stimulation of rat granulosa cell deoxyribonucleic acid synthesis by sex steroids and follicle-stimulating hormone. J Steroid Biochem Mol Biol 1997; 62:11-9. [PMID: 9366494 DOI: 10.1016/s0960-0760(97)00021-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Although follicle-stimulating hormone (FSH) and estrogens are known to be the main physiological stimuli for the development of the ovarian follicle in mammals, their growth-promoting activity has not been clearly established "in vitro". Furthermore, experimental evidence indicates that FSH and estradiol can independently inhibit granulosa cell proliferation. The present study was aimed at examining the effect of sex steroids in combination with FSH, on DNA synthesis in rat granulosa cells cultured in completely defined medium. Estradiol and FSH, when added separately, produced a significant inhibition of [3H] thymidine incorporation. In contrast, a combination of a low dose of FSH (20 ng/ml) with estradiol (100 ng/ml) produced a shift in the period of maximal DNA synthesis from 96 to 48 h after plating. Dose response studies showed that estradiol effects were produced at physiological intraovarian concentrations (1-100 ng/ml), whereas the effects of FSH were biphasic, with high doses (200 ng/ml) being inhibitory. A similar biphasic dose response curve was observed with increasing concentrations of a cAMP derivative in the presence of maximally effective doses of either an aromatizable steroid (androstenedione), insulin or insulin-like growth factor I. Non-aromatizable androgens (5alpha-dihydrotestosterone, 5alpha-androstane 3alpha-17beta diol and androsterone) showed a potency comparable to that of estradiol. The effect of 5alpha-dihydrotestosterone was completely blocked by a specific antiandrogen (hydroxy-flutamide), indicating that it was mediated by the androgen receptor. The effects of estradiol and androgens were not additive. The interaction between estradiol and FSH was further amplified in the presence of a maximally effective dose of insulin. Data presented herein indicate that both estrogens and androgens are able to elicit a mitogenic response in purified granulosa cells, cultured in a completely defined medium, provided the cells are stimulated by a physiological dose of FSH. These results suggest that, during follicular development, the stimulus for granulosa cell proliferation is given by the concerted action of steroid and peptide hormones acting through different signalling pathways.
Collapse
Affiliation(s)
- M A Bley
- Instituto de Biología y Medicina Experimental-CONICET and Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
| | | | | |
Collapse
|
50
|
Bodensteiner KJ, Wiltbank MC, Bergfelt DR, Ginther OJ. Alterations in follicular estradiol and gonadotropin receptors during development of bovine antral follicles. Theriogenology 1996; 45:499-512. [PMID: 16727813 DOI: 10.1016/0093-691x(95)00386-m] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/1995] [Accepted: 06/30/1995] [Indexed: 11/18/2022]
Abstract
It was hypothesized that growth divergence of dominant and subordinate follicles during Wave 1 and growth termination of the dominant follicle would be associated with changes in the number of gonadotropin receptors on granulosa cells and estradiol in follicular fluid. To test this hypothesis, follicular development of 16 Holstein heifers was monitored by ultrasound, and follicles were collected on Days 2,4,6 and 10 (Day 0 = ovulation). Dominant follicles were compared across days, whereas dominant and largest subordinate follicles were compared on Days 2 and 4 only. The numbers of LH and FSH receptors on the granulosa cells of dominant follicles did not differ significantly over Days 2, 4, 6 and 10. In contrast, concentrations of estradiol in follicular fluid decreased (P < 0.05) from Days 2 to 10 (373 +/- 150 to 42 +/- 12 ng/ml) and concentrations of progesterone in follicular fluid increased (P < 0.05) from Days 2 to 10 (12.2 +/- 2.3 to 24.4 +/- 4.8 ng/ml). Correspondingly, the ratio of estradiol:progesterone in the dominant follicles decreased (P < 0.05) from Days 2 to 10. Comparisons between dominant and subordinate follicles indicated greater (P < 0.05) estradiol concentrations in the dominant follicle on Day 2, but the number of gonadotropin receptors was not different until Day 4. Thus, differences in concentrations of follicular fluid estradiol, but not numbers of granulosa cell gonadotropin receptors, were associated with the early growth divergence of dominant and subordinate follicles (Day 2) and the eventual growth termination of the dominant follicle (Day 10). Late divergence (Day 4) was associated with higher gonadotropin receptor numbers and follicular estradiol concentrations in the dominant than in the subordinate follicles. These results indicate that an increase in estradiol productivity of the selected dominant follicle occurred before an increase in the number of gonadotropin receptors.
Collapse
Affiliation(s)
- K J Bodensteiner
- Department of Dairy Science, University of Wisconsin--Madison, Madison, WI 53706, USA
| | | | | | | |
Collapse
|