51
|
Fuentes A, Sequeira K, Tapia-Pizarro A, Muñoz A, Salinas A, Céspedes P, Escalona J, Godoy A. Androgens Profile in Blood Serum and Follicular Fluid of Women With Poor Ovarian Response During Controlled Ovarian Stimulation Reveals Differences Amongst POSEIDON Stratification Groups: A Pilot Study. Front Endocrinol (Lausanne) 2019; 10:458. [PMID: 31379738 PMCID: PMC6646462 DOI: 10.3389/fendo.2019.00458] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 06/25/2019] [Indexed: 12/31/2022] Open
Abstract
Patients with poor ovarian response (POR) to exogenous gonadotropins stimulation for assisted reproductive technology (ART) have decreased circulating androgens during spontaneous cycles. The Patient-Oriented Strategies Encompassing Individualized Oocyte Number (POSEIDON) is a 4-tier stratification of women with POR to controlled ovarian stimulation (COH) based on age and biomarkers of ovarian reserve has been proposed to maximize the clinical management of this group for ART. The aim of the present study was to characterize the levels of androgens during COH in follicular fluid (FF) and serum in POSEIDON subgroups and compared them with women of normal ovarian response. Sixty nine consecutive patients undergoing ART were included and testosterone, androstenedione, dehydroepiandrosterone sulfate (DHEA-S), estradiol, sex hormone-binding globulin (SHBG), and insulin-like growth factor 1 (IGF-1) were measured in serum and FF collected at the time of oocyte pick-up. The number of retrieved oocytes was registered for each patient for their allocation to the respective POSEIDON subgroup. The control group comprised 19 women and the POSEIDON group 1 (age < 35, normal ovarian reserve biomarkers) n = 14, group 2 (age ≥ 35, normal ovarian reserve biomarkers) n = 8, group 3 (age < 35, poor ovarian reserve biomarkers) n = 6 and group 4 (age ≥ 35, poor ovarian reserve biomarkers) n = 22. Serum levels of total testosterone, androstenedione and DHEA-S were not different in group 1 vs. control but significantly decreased in group 3 vs. control. DHEA-S in FF was also significantly decreased in group 3 vs. control. In addition, serum testosterone was decreased in groups 2 and 4 vs. control; and serum androstenedione and estradiol were reduced in group 4 vs. control. No differences were observed for estradiol, SHBG and IGF-1 in FF. Finally, a high correlation between serum and FF DHEA-S was observed when data from samples of all groups were pooled. Group 1 did not show hypoandrogenemia however group 3 had low levels of all measured androgens in serum and DHEA-S in FF. Such differences might help to better characterize and/or improve the clinical management of women with POR according to their respective POSEIDON stratification.
Collapse
|
52
|
Li J, Huang D, Sun X, Li X, Cheng CHK. Zinc mediates the action of androgen in acting as a downstream effector of luteinizing hormone on oocyte maturation in zebrafish†. Biol Reprod 2018; 100:468-478. [DOI: 10.1093/biolre/ioy224] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 08/01/2018] [Indexed: 12/18/2022] Open
Affiliation(s)
- Jianzhen Li
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| | - Duo Huang
- School of Biomedical Sciences, The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Xiao Sun
- School of Biomedical Sciences, The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| | - Xuehui Li
- College of Life Sciences, Northwest Normal University, Lanzhou, China
| | - Christopher H K Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong-Shandong University Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong, China
| |
Collapse
|
53
|
Chávez-Genaro R, Anesetti G. First ovarian response to gonadotrophin stimulation in rats exposed to neonatal androgen excess. J Mol Histol 2018; 49:631-637. [PMID: 30302594 DOI: 10.1007/s10735-018-9800-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 10/05/2018] [Indexed: 11/29/2022]
Abstract
This study analyzes the effects of neonatal androgenization on follicular growth and first ovulation in response to gonadotrophins, using a model of exogenous stimulation or the use of subcutaneous ovary grafts in castrated animals to replace the hypothalamus-pituitary signal. Neonatal rats (days 1-5) were treated with testosterone, dihydrotestosterone or vehicle. At juvenile period, rats were stimulated with PMSG, hCG (alone or combined) or used as ovarian donors to be grafted on castrated adult female rats. Ovulation and ovarian histology were analyzed in both groups. Animals treated with vehicle or dihydrotestosterone stimulated with gonadotrophins (pharmacological or by using an ovary graft) ovulated, showing a normal histological morphology whereas rats exposed to testosterone and injected with the same doses of gonadotrophins did not it. In this group, ovulation was reached using a higher dose of hCG. Ovaries in the testosterone group were characterized by the presence of follicles with atretic appearance and a larger size than those observed in control or dihydrotestosterone groups. A similar appearance was observed in testosterone ovary grafts although luteinization and some corpora lutea were also identified. Our findings suggest that neonatal exposure to aromatizable androgens induces a more drastic signalling on the ovarian tissue that those driven by non-aromatizable androgens in response to gonadotrophins.
Collapse
Affiliation(s)
- Rebeca Chávez-Genaro
- Histology and Embryology Department, School of Medicine, UdelaR, General Flores 2125, CP 11800, Montevideo, Uruguay.
| | - Gabriel Anesetti
- Histology and Embryology Department, School of Medicine, UdelaR, General Flores 2125, CP 11800, Montevideo, Uruguay
| |
Collapse
|
54
|
Dehydroepiandrosterone Ameliorates Abnormal Mitochondrial Dynamics and Mitophagy of Cumulus Cells in Poor Ovarian Responders. J Clin Med 2018; 7:jcm7100293. [PMID: 30241351 PMCID: PMC6210273 DOI: 10.3390/jcm7100293] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/19/2018] [Accepted: 09/19/2018] [Indexed: 12/30/2022] Open
Abstract
Mitochondrial dysfunction is related to reproductive decline in humans, with consequences for in vitro fertilization (IVF). We assessed whether dehydroepiandrosterone (DHEA) could regulate mitochondrial homeostasis and mitophagy of cumulus cells (CCs) in poor ovarian responders (PORs). A total of 66 women who underwent IVF treatment at the Reproductive Medicine Center of Kaohsiung Veterans General Hospital were included in this study. Twenty-eight normal ovarian responders (NOR) and 38 PORs were enrolled. PORs were assigned to receive DHEA supplementation (n = 19) or not (n = 19) before IVF cycles. DHEA prevents mitochondrial dysfunction by decreasing the activation of DNM1L and MFF, and increasing MFN1 expression. Downregulation of PINK1 and PRKN occurred after DHEA treatment, along with increased lysosome formation. DHEA not only promoted mitochondrial mass but also improved mitochondrial homeostasis and dynamics in the CCs of POR. We also observed effects of alterations in mRNAs known to regulate mitochondrial dynamics and mitophagy in the CCs of POR. DHEA may prevent mitochondrial dysfunction through regulating mitochondrial homeostasis and mitophagy.
Collapse
|
55
|
Chern CU, Tsui KH, Vitale SG, Chen SN, Wang PH, Cianci A, Tsai HW, Wen ZH, Lin LT. Dehydroepiandrosterone (DHEA) supplementation improves in vitro fertilization outcomes of poor ovarian responders, especially in women with low serum concentration of DHEA-S: a retrospective cohort study. Reprod Biol Endocrinol 2018; 16:90. [PMID: 30223902 PMCID: PMC6142344 DOI: 10.1186/s12958-018-0409-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 09/11/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Dehydroepiandrosterone (DHEA) is now widely used as an adjuvant for in vitro fertilization (IVF) cycles in poor ovarian responders (PORs). Several studies showed that DHEA supplementation could improve IVF outcomes of PORs. However, most of the PORs do not respond to DHEA clinically. Therefore, the aim of this study is to confirm the beneficial effects of DHEA on IVF outcomes of PORs and to investigate which subgroups of PORs can best benefit from DHEA supplementation. METHODS This retrospective cohort study was performed between January 2015 and December 2017. A total of 151 PORs who fulfilled the Bologna criteria and underwent IVF cycles with the gonadotropin-releasing hormone antagonist protocol were identified. The study group (n = 67) received 90 mg of DHEA daily for an average of 3 months before the IVF cycles. The control group (n = 84) underwent the IVF cycles without DHEA pretreatment. The basic and cycle characteristics and IVF outcomes between the two groups were compared using independent t-tests, Chi-Square tests and binary logistic regression. RESULTS The study and control groups did not show significant differences in terms of basic characteristics. The study group demonstrated a significantly greater number of retrieved oocytes, metaphase II oocytes, fertilized oocytes, day 3 embryos and top-quality embryos at day 3 and a higher clinical pregnancy rate, ongoing pregnancy rate and live birth rate than those measures in the control group. The multivariate analysis revealed that DHEA supplementation was positively associated with clinical pregnancy rate (OR = 4.93, 95% CI 1.68-14.43, p = 0.004). Additionally, in the study group, the multivariate analysis showed that serum dehydroepiandrosterone-sulfate (DHEA-S) levels < 180 μg/dl were significantly associated with a rate of retrieved oocytes > 3 (OR = 5.92, 95% CI 1.48-23.26, p = 0.012). CONCLUSIONS DHEA supplementation improves IVF outcomes of PORs. In PORs with DHEA pretreatment, women with lower DHEA-S level may have greater possibility of attaining more than 3 oocytes.
Collapse
Affiliation(s)
- Chyi-Uei Chern
- 0000 0004 0572 9992grid.415011.0Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist, Kaohsiung City, 81362 Taiwan
| | - Kuan-Hao Tsui
- 0000 0004 0572 9992grid.415011.0Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist, Kaohsiung City, 81362 Taiwan
- 0000 0001 0425 5914grid.260770.4Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, No. 155, Sec. 2, Li-Nong Street, Pei-Tou, Taipei, 112 Taiwan
- 0000 0004 0639 0943grid.412902.cDepartment of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, No.20, Weixin Rd, Yanpu, Township, Pingtung County 90741 Taiwan
| | - Salvatore Giovanni Vitale
- 0000 0004 1757 1969grid.8158.4Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123 Catania, Italy
| | - San-Nung Chen
- 0000 0004 0572 9992grid.415011.0Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist, Kaohsiung City, 81362 Taiwan
| | - Peng-Hui Wang
- 0000 0001 0425 5914grid.260770.4Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, No. 155, Sec. 2, Li-Nong Street, Pei-Tou, Taipei, 112 Taiwan
- 0000 0004 0604 5314grid.278247.cDepartment of Obstetrics and Gynecology, Taipei Veterans General Hospital, No. 201, Section 2, Shih-Pai Road, Taipei, 112 Taiwan
- 0000 0004 0572 9415grid.411508.9Department of Medical Research, China Medical University Hospital, No. 2, Yude Road, North District, Taichung City, 40447 Taiwan
| | - Antonio Cianci
- 0000 0004 1757 1969grid.8158.4Department of General Surgery and Medical Surgical Specialties, University of Catania, 95123 Catania, Italy
| | - Hsiao-Wen Tsai
- 0000 0004 0572 9992grid.415011.0Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist, Kaohsiung City, 81362 Taiwan
- 0000 0001 0425 5914grid.260770.4Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, No. 155, Sec. 2, Li-Nong Street, Pei-Tou, Taipei, 112 Taiwan
| | - Zhi-Hong Wen
- 0000 0004 0531 9758grid.412036.2Department of Marine Biotechnology and Resources, National Sun Yat-sen University, 70 Lienhai Rd, Kaohsiung City, 80424 Taiwan
| | - Li-Te Lin
- 0000 0004 0572 9992grid.415011.0Department of Obstetrics and Gynecology, Kaohsiung Veterans General Hospital, No.386, Dazhong 1st Rd., Zuoying Dist, Kaohsiung City, 81362 Taiwan
- 0000 0001 0425 5914grid.260770.4Department of Obstetrics and Gynecology, National Yang-Ming University School of Medicine, No. 155, Sec. 2, Li-Nong Street, Pei-Tou, Taipei, 112 Taiwan
- 0000 0004 0531 9758grid.412036.2Department of Biological Science, National Sun Yat-Sen University, 70 Lienhai Rd, Kaohsiung City, 80424 Taiwan
| |
Collapse
|
56
|
D'Amato G, Caringella AM, Stanziano A, Cantatore C, Palini S, Caroppo E. Mild ovarian stimulation with letrozole plus fixed dose human menopausal gonadotropin prior to IVF/ICSI for infertile non-obese women with polycystic ovarian syndrome being pre-treated with metformin: a pilot study. Reprod Biol Endocrinol 2018; 16:89. [PMID: 30217209 PMCID: PMC6137735 DOI: 10.1186/s12958-018-0405-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 08/28/2018] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Letrozole is widely employed as ovulation induction agent in women with PCOS, but its use in mild stimulation (MS) protocols for IVF is limited. Aim of the present study was to evaluate the feasibility of a MS protocol with letrozole plus hMG in non-obese PCOS women undergoing IVF after a metformin pre-treatment. METHODS We retrospectively evaluated the data of 125 non-obese PCOS undergoing MS with letrozole plus hMG, 150 IU as starting dose, (group 1, N = 80) compared to those undergoing a conventional IVF stimulation protocols (CS) (group 2, N = 45) prior to IVF. All patients had received metformin extended release 1200-2000 mg daily for three to six months before IVF. GnRH antagonist was administered in both groups when the leading follicles reached 14 mm. RESULTS Both groups were comparable for age, BMI and ovarian reserve markers. Both groups showed lower than expected AFC and AMH values as a consequence of metformin pre-treatment. Letrozole-treated patients required a significantly lower amount of gonadotropins units (p < 0.0001), and showed significantly lower day 5, day 8 and hCG day E2 levels compared to patients undergoing the CS protocol (p < 0.0001, p < 0.0001 and p = 0.001 respectively). The oocyte yield, in terms of total (6, IQR 3, vs 6, IQR 4 respectively,) and MII oocytes (5, IQR 3, vs 5, IQR 3, respectively) number, did not differ among groups; the number of total (3, IQR 2, vs 3, IQR 1 respectively) and good quality embryos (2, IQR1 vs 2, IQR 1,5 respectively) obtained was comparable as well in the two groups. The number of fresh transfers was significantly higher in group 1 compared to group 2 (80% vs 60%, p = 0.016). A trend for higher cumulative clinical pregnancy rate was found in women undergoing MS compared to CS (42.5%vs 24,4%, p = 0.044), but the study was not powered to detect this difference. CONCLUSIONS The present study suggests that the use of letrozole as adjuvant treatment to MS protocols for IVF may be an effective alternative to CS protocols for non-obese PCOS patients pre-treated with metformin, as it provides comparable IVF outcome without requiring high FSH dose, and avoiding supraphysiological estradiol levels.
Collapse
Affiliation(s)
- Giuseppe D'Amato
- Asl Bari, Department of Maternal and Child Health, Reproductive and IVF Unit, Conversano, BA, Italy
| | - Anna Maria Caringella
- Asl Bari, Department of Maternal and Child Health, Reproductive and IVF Unit, Conversano, BA, Italy
| | - Antonio Stanziano
- Asl Bari, Department of Maternal and Child Health, Reproductive and IVF Unit, Conversano, BA, Italy
| | - Clementina Cantatore
- Asl Bari, Department of Maternal and Child Health, Reproductive and IVF Unit, Conversano, BA, Italy
| | - Simone Palini
- Asl Bari, Department of Maternal and Child Health, Reproductive and IVF Unit, Conversano, BA, Italy
| | - Ettore Caroppo
- Asl Bari, Department of Maternal and Child Health, Reproductive and IVF Unit, Conversano, BA, Italy.
- ASL Bari, PTA "F Jaia", Fisiopatologia della Riproduzione Umana e P.M.A, via de Amicis 30, 70014, Conversano, BA, Italy.
| |
Collapse
|
57
|
Ciucci A, Ferrandina G, Mascilini F, Filippetti F, Scambia G, Zannoni GF, Gallo D. Estrogen receptor β: Potential target for therapy in adult granulosa cell tumors? Gynecol Oncol 2018; 150:158-165. [DOI: 10.1016/j.ygyno.2018.05.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 10/16/2022]
|
58
|
Gleicher N. Expected advances in human fertility treatments and their likely translational consequences. J Transl Med 2018; 16:149. [PMID: 29866181 PMCID: PMC5987489 DOI: 10.1186/s12967-018-1525-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/26/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Due to rapid research progress in reproductive biology and reproductive clinical endocrinology, many human infertility treatments are close to potential breakthroughs and translational applications. We here review current barriers, where such breakthroughs will likely come from, what they will entail, and their potential clinical applications. MAIN TEXT The radical nature of change will primarily benefit older women, reduce fertility treatment costs and thereby expand access to treatment. A still widely overlooked prerequisite for implantation and normal pregnancy maintenance is timely development of maternal immunological tolerance toward an implanting paternal semi-allograft, if malfunctioning associated with implantation failure and pregnancy loss, while premature termination of tolerance appears associated with premature labor, pre-eclampsia/eclampsia and gestoses of pregnancy. Common denominators between pregnancy and invasive malignancies have again been attracting attention, suggesting that, like in malignant tumors, degrees of embryo aneuploidy may affect invasiveness and ability to "disarm" the immune system's innate response against implanting embryos. Linking tolerance to implantation, we offer evidence that the so-called "implantation window" is likely immunological rather than hormonally defined. CONCLUSIONS Because many here outlined treatment changes will disproportionally benefit older women, they will exert a pronounced effect on society, as increasing numbers of women at grandparental ages will become mothers.
Collapse
Affiliation(s)
- Norbert Gleicher
- The CHR, 21 East 69th Street, New York, NY, 10021, USA.
- The Foundation for Reproductive Medicine, New York, NY, 10021, USA.
- Laboratory for Stem Cell Biology and Molecular Embryology, Rockefeller University, New York, NY, 10065, USA.
- Department of Obstetrics and Gynecology, Vienna Medical School, 1090, Vienna, Austria.
| |
Collapse
|
59
|
Gleicher N, Kushnir VA, Darmon S, Albertini DF, Barad DH. Older women using their own eggs? Issue framed with two oldest reported IVF pregnancies and a live birth. Reprod Biomed Online 2018; 37:172-177. [PMID: 29936089 DOI: 10.1016/j.rbmo.2018.05.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 05/15/2018] [Accepted: 05/18/2018] [Indexed: 10/16/2022]
Abstract
RESEARCH QUESTION What level of IVF pregnancy success is currently possible in women of extremely advanced age? DESIGN This study reports on outcomes in women aged 43-51 years at the Centre for Human Reproduction, an academically affiliated private clinical fertility and research centre in New York City. RESULTS During the study years of 2014-2016, 16 pregnancies were established, all through day 3 transfers. Based on 'intent to treat' (cycle start), clinical pregnancy rates were 4/190 (2.1%), 5/234 (2.1%) and 7/304 (2.3%) and live birth rates were 2/190 (1.1%), 1/234 (0.43%) and 4/304 (1.3%) in 2014, 2015 and 2016, respectively. With reference to embryo transfer, clinical pregnancy rates were 4/140 (2.9%), 5/159 (3.1%) and 7/167 (4.2%) and live birth rates were 2/140 (1.4%), 1/159 (0.63%) and 4/167 (2.4%) for the same years. The results for 2016 also included what are probably the two oldest autologous IVF pregnancies ever reported in the literature. These results were obtained with patient ages, percentage of cycle cancellations and other adverse outcome parameters steadily increasing year by year. CONCLUSIONS Female age above 42 is widely viewed as the ultimate barrier to conception with IVF. Data reported here, although small and preliminary, demonstrate that potential outcomes are better than widely perceived, while pregnancy and live birth rates remain significantly inferior to donor egg recipient cycles. However, for selected women at very advanced ages, especially with higher egg/embryo numbers, autologous oocyte IVF offers a better option than widely acknowledged, if they are given individualized age-specific care.
Collapse
Affiliation(s)
- Norbert Gleicher
- Centre for Human Reproduction, New York, NY, USA; The Foundation for Reproductive Medicine, New York, NY, USA; Laboratory for Stem Cell Biology and Molecular Embryology, Rockefeller University, New York, NY, USA; Department of Obstetrics and Gynecology, Vienna Medical School, Vienna, Austria.
| | - Vitaly A Kushnir
- Centre for Human Reproduction, New York, NY, USA; Department of Obstetrics and Gynecology, Wake Forest University, Winston Salem, NC, USA
| | - Sarah Darmon
- Centre for Human Reproduction, New York, NY, USA
| | - David F Albertini
- Centre for Human Reproduction, New York, NY, USA; Laboratory for Stem Cell Biology and Molecular Embryology, Rockefeller University, New York, NY, USA
| | - David H Barad
- Centre for Human Reproduction, New York, NY, USA; The Foundation for Reproductive Medicine, New York, NY, USA
| |
Collapse
|
60
|
Gleicher N, Kushnir VA, Barad DH. Impact of androgen supplementation on the follicular endocrine milieu in women with hypoandrogenism. Reprod Biomed Online 2018; 36:719-720. [PMID: 29598847 DOI: 10.1016/j.rbmo.2018.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/05/2018] [Indexed: 11/30/2022]
Affiliation(s)
- Norbert Gleicher
- The Center for Human Reproduction, New York, New York, 10021, USA; The Foundation for Reproductive Medicine, New York, New York 10020, USA; Stem Cell Biology and Molecular Embryology Laboratory, Rockefeller University, New York, New York 10016, USA; Department of Obstetrics and Gynecology, Vienna University School of Medicine, 1090 Vienna, Austria.
| | - Vitaly A Kushnir
- The Center for Human Reproduction, New York, New York, 10021, USA; Department of Obstetrics and Gynecology, Wake Forest University, Winston Salem VAK, North Carolina 27101, USA
| | - David H Barad
- The Center for Human Reproduction, New York, New York, 10021, USA; The Foundation for Reproductive Medicine, New York, New York 10020, USA
| |
Collapse
|
61
|
Histone demethylase KDM4A and KDM4B expression in granulosa cells from women undergoing in vitro fertilization. J Assist Reprod Genet 2018. [PMID: 29536385 DOI: 10.1007/s10815-018-1151-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE To assess expression of the histone demethylases KDM4A and KDM4B in granulosa collected from women undergoing oocyte retrieval and to determine if expression was related to pregnancy outcome. METHODS Cumulus and mural granulosa cells were obtained from women undergoing oocyte retrieval. KDM4A and KDM4B mRNA expression was determined by qRT-PCR. KDM4A and KDM4B proteins were immunohistochemically localized in ovarian tissue sections obtained from archival specimens. RESULTS KDM4A and KDM4B protein was localized to oocytes, granulosa cells, and theca and luteal cells in ovaries from reproductive-aged women. KDM4A and KDM4B mRNA expression was overall higher in cumulus compared to mural granulosa. When comparing granulosa demethylase gene expression, KDM4A and KDM4B mRNA expression was higher in both cumulus and mural granulosa from not pregnant patients compared to patients in the pregnant-live birth group. CONCLUSIONS Histone demethylases KDM4A and KDM4B mRNA are differentially expressed in cumulus and mural granulosa. Expression of both KDM4A and KDM4B mRNA was lower in cumulus granulosa and mural granulosa from pregnant compared to not pregnant patients. These findings suggest that altered expression of histone demethylases may impact epigenetic changes in granulosa cells associated with pregnancy.
Collapse
|
62
|
Zhou C, Flaws JA. Effects of an Environmentally Relevant Phthalate Mixture on Cultured Mouse Antral Follicles. Toxicol Sci 2018; 156:217-229. [PMID: 28013214 DOI: 10.1093/toxsci/kfw245] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Phthalates are used in building materials, medical devices, and personal care products. Most studies on phthalates have focused on single phthalates, but it is important to study mixtures of phthalates because humans are exposed to such mixtures daily. We tested the hypothesis that phthalate mixture exposure decreases antral follicle growth, compromises steroidogenic capacity, and induces atresia. Antral follicles from adult CD-1 mice were cultured with vehicle control or phthalate mixture (1-500 µg/ml) for 96 h. The mixture was made of 35% diethyl phthalate, 21% di(2-ethylhexyl) phthalate, 15% dibutyl phthalate, 15% diisononyl phthalate, 8% diisobutyl phthalate, and 5% benzylbutyl phthalate. During culture, antral follicle diameters were measured every 24 h to monitor growth. After culture, media were subjected to measurements of sex steroid hormones and follicles were subjected to evaluation of gene expression and atresia. The phthalate mixture (100 and 500 µg/ml) decreased antral follicle growth starting at 24 h compared to controls. The mixture at 10, 100, and 500 µg/ml also decreased androstenedione, testosterone, estrone, and estradiol levels compared to control. The mixture (10, 100, and 500 µg/ml) reduced atresia rating, but it induced more oocyte fragmentation compared to control. The phthalate mixture at different doses adversely affected cell cycle regulators, antioxidant enzymes, apoptotic factors, steroidogenic enzymes, and receptors. Collectively, these data indicate that exposure to an environmentally relevant phthalate mixture reduces antral follicle growth, induces oocyte fragmentation, and decreases hormone production by adversely affecting the expression of cell cycle regulators, apoptotic factors, steroidogenic enzymes, and receptors.
Collapse
Affiliation(s)
- Changqing Zhou
- Department of Comparative Biosciences, University of Illinois, Urbana, Illinois 61802
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois, Urbana, Illinois 61802
| |
Collapse
|
63
|
Gleicher N, Kushnir VA, Darmon SK, Wang Q, Zhang L, Albertini DF, Barad DH. Suspected ontogeny of a recently described hypo-androgenic PCOS-like phenotype with advancing age. Endocrine 2018; 59:661-676. [PMID: 29305800 DOI: 10.1007/s12020-017-1498-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 12/11/2017] [Indexed: 11/29/2022]
Abstract
BACKGROUND A recent report described a new PCOS-like phenotype in lean older infertile women, and was characterized by high age-specific anti-Müllerian hormone (AMH) but hypo- rather than the expected hyper-androgenism. The hypo-androgenism was, furthermore, characterized of, likely, adrenal origin and autoimmune etiology. PATIENTS AND METHODS We extracted data on 708 consecutive infertility patients, and separated them into three age-strata, <35, 36-42, and >42 years. In each stratum, we investigated how levels of anti-Müllerian hormone (AMH) and testosterone (T) interrelate between high-AMH (AMH ≥ 75th quantile) and normal AMH (25th-75th quantile) and low-T (total testosterone ≤19.0 ng/dL), normal-T (19.0-29.0 ng/dL) and high-T (>29.0 ng/dL). High-AMH cycles were presumed to reflect PCOS-like patients. Routine in vitro fertilization (IVF) cycle outcomes and clinical phenotypes of patients were then compared between groups with AMH and T as statistical variables. RESULTS This hypo-androgenic PCOS-like phenotype already exists in age stratum <35 years. It appears to arise from a lean, at very young ages hyper-androgenic PCOS phenotype that develops in comparison to controls (likely autoimmune-induced) insufficiency of the adrenal zona reticularis (low-T and low-DHEAS) and zona fasciculata (low-C), and is characterized by frequent evidence of autoimmunity. A degree of adrenal insufficiency, thus, concomitantly appears to affect adrenal androgen and, to lesser degrees, glucocorticoid production (mineralocorticoids were not investigated). CONCLUSIONS Here investigated new PCOS-like phenotype demonstrates features compatible with what under Rotterdam criteria has been referred to as PCOS phenotype-D. If confirmed, the observation that the ontogeny of this phenotype already at young ages is, likely, driven by adrenal autoimmunity, supports the position of the androgen excess and PCOS society that the etiology of phenotype-D differs from that of classical hyper-androgenic PCOS of mostly ovarian etiology.
Collapse
Affiliation(s)
- Norbert Gleicher
- The Center for Human Reproduction, New York, NY, 10021, USA.
- The Foundation for Reproductive Medicine, New York, NY, 10020, USA.
- Stem Cell Biology and Molecular Embryology Laboratory, Rockefeller University, New York, NY, 10016, USA.
- Department of Obstetrics and Gynecology, Vienna University School of Medicine, 1090, Vienna, Austria.
| | - Vitaly A Kushnir
- The Center for Human Reproduction, New York, NY, 10021, USA
- Department of Obstetrics and Gynecology, Wake Forest University, Winston Salem, Winston Salem, NC, 27101, USA
| | - Sarah K Darmon
- The Center for Human Reproduction, New York, NY, 10021, USA
| | - Qi Wang
- The Center for Human Reproduction, New York, NY, 10021, USA
| | - Lin Zhang
- The Center for Human Reproduction, New York, NY, 10021, USA
| | - David F Albertini
- The Center for Human Reproduction, New York, NY, 10021, USA
- Stem Cell Biology and Molecular Embryology Laboratory, Rockefeller University, New York, NY, 10016, USA
| | - David H Barad
- The Center for Human Reproduction, New York, NY, 10021, USA
- The Foundation for Reproductive Medicine, New York, NY, 10020, USA
| |
Collapse
|
64
|
Zebrafish androgen receptor is required for spermatogenesis and maintenance of ovarian function. Oncotarget 2018; 9:24320-24334. [PMID: 29849943 PMCID: PMC5966271 DOI: 10.18632/oncotarget.24407] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 01/24/2018] [Indexed: 11/25/2022] Open
Abstract
The androgen receptor (AR) is a nuclear receptor protein family member and inducible transcription factor that modulates androgen target gene expression. Studies using a mouse model confirmed the need for ar in reproductive development, particularly spermatogenesis. Here, we investigated the role of ar in zebrafish using CRISPR/Cas9 gene targeting technology. Targeted disruption of ar in zebrafish increases the number of female offspring and increases offspring weight. In addition, ar-null male zebrafish have female secondary sex characteristics. More importantly, targeted disruption of ar in zebrafish causes male infertility via defective spermatogenesis and female premature ovarian failure during growth. Mechanistic assays suggest that these effects are caused by fewer proliferated cells and more apoptotic cells in ar-null testes. Moreover, genes involved in reproductive development, estradiol induction and hormone synthesis were dys-regulated in testes and ovaries and the reproductive-endocrine axis was disordered. Our data thus suggest that the zebrafish ar is required for spermatogenesis and maintenance of ovarian function, which confirms evolutionarily conserved functions of ar in vertebrates, as well as indicates that ar-null zebrafish are a suitable model for studying pathologic mechanisms related to androgen disorders.
Collapse
|
65
|
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
|
66
|
Monson C, Forsgren K, Goetz G, Harding L, Swanson P, Young G. A teleost androgen promotes development of primary ovarian follicles in coho salmon and rapidly alters the ovarian transcriptome†. Biol Reprod 2017; 97:731-745. [DOI: 10.1093/biolre/iox124] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/12/2017] [Indexed: 12/30/2022] Open
|
67
|
Duda M, Wartalski K, Tabarowski Z, Gorczyca G. The Role of Androgens in Ovarian Follicular Development: From Fertility to Ovarian Cancer. Theriogenology 2017. [DOI: 10.5772/intechopen.68881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
68
|
Gleicher N, Kushnir VA, Albertini DF, Barad DH. Letter to the Editor: Including the Zona Reticularis in the Definition of Hypoadrenalism and Hyperadrenalism. J Clin Endocrinol Metab 2017; 102:3569-3570. [PMID: 28911156 DOI: 10.1210/jc.2017-00671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/27/2017] [Indexed: 02/13/2023]
Affiliation(s)
- Norbert Gleicher
- The Center for Human Reproduction, New York, New York 10021
- Foundation for Reproductive Medicine, New York, New York 10021
- Laboratory of Stem Cell Biology and Molecular Embryology, Rockefeller University, New York, New York 10065
- Department of Obstetrics and Gynecology, University of Vienna School of Medicine, 1090 Vienna, Austria
| | - Vitaly A Kushnir
- The Center for Human Reproduction, New York, New York 10021
- Department of Obstetrics and Gynecology, Wake Forest University, Winston Salem, North Carolina 27109
| | - David F Albertini
- The Center for Human Reproduction, New York, New York 10021
- Laboratory of Stem Cell Biology and Molecular Embryology, Rockefeller University, New York, New York 10065
| | - David H Barad
- The Center for Human Reproduction, New York, New York 10021
- Foundation for Reproductive Medicine, New York, New York 10021
| |
Collapse
|
69
|
Converse A, Zhang C, Thomas P. Membrane Androgen Receptor ZIP9 Induces Croaker Ovarian Cell Apoptosis via Stimulatory G Protein Alpha Subunit and MAP Kinase Signaling. Endocrinology 2017. [PMID: 28633436 DOI: 10.1210/en.2017-00087] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent studies show that androgen-induced apoptosis in Atlantic croaker primary granulosa and theca (G/T) cells and in human breast and prostate cancer cell lines is mediated by the membrane androgen receptor ZIP9, which belongs to the SLC39A zinc transporter family. However, the apoptotic signaling pathways remain unclear because ZIP9 activates an inhibitory G protein in human cancer cells, whereas recombinant croaker ZIP9 activates a stimulatory G protein (Gs) in transfected cancer cells. We investigated androgen-dependent apoptotic pathways to identify the signaling pathways regulated through wild-type croaker ZIP9 in ovarian follicle cells. We show that the ZIP9-mediated apoptotic signaling pathway in croaker G/T cells shares several proapoptotic members with those in human cancer cells, but is activated through a Gsα subunit-dependent pathway. Testosterone treatment of croaker G/T cells increased intracellular zinc levels, mitogen-activated protein (MAP) kinase activity, caspase 3 activity, messenger RNA levels of proapoptotic members Bax, p53, and c-Jun N-terminal kinase, and the incidence of apoptosis, similar to findings in mammalian cancer cells, but also increased cyclic adenosine monophosphate concentrations. Transfection with small interfering RNA targeting croaker ZIP9 blocked testosterone-induced increase in bax, p53, and jnk expression. Testosterone-induced apoptosis and caspase 3 activation depended on the presence of extracellular zinc and were effectively blocked with cotreatment of inhibitors of the Gsα subunit, adenylyl cyclase, protein kinase A, and MAP kinase (Erk1/2) activation. These results indicate that ZIP9-mediated testosterone signaling in croaker G/T cells involves multiple pathways, some of which differ from those activated through ZIP9 in human cancer cells even though a similar apoptotic response is observed.
Collapse
Affiliation(s)
- Aubrey Converse
- Marine Science Institute, The University of Texas at Austin, Port Aransas, Texas 78373
| | - Chenan Zhang
- Marine Science Institute, The University of Texas at Austin, Port Aransas, Texas 78373
| | - Peter Thomas
- Marine Science Institute, The University of Texas at Austin, Port Aransas, Texas 78373
| |
Collapse
|
70
|
Ma X, Hayes E, Biswas A, Seger C, Prizant H, Hammes SR, Sen A. Androgens Regulate Ovarian Gene Expression Through Modulation of Ezh2 Expression and Activity. Endocrinology 2017; 158:2944-2954. [PMID: 28666321 PMCID: PMC5659665 DOI: 10.1210/en.2017-00145] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/23/2017] [Indexed: 02/07/2023]
Abstract
A substantial amount of evidence suggests that androgen signaling through classical androgen receptors is critical for both normal and pathologic ovarian physiology. Specifically, we and others have shown that, in mouse granulosa cells, androgen actions through both extranuclear and nuclear androgen receptor signaling are critical for normal follicle development and ovulation. Here, we show that androgens through the PI3K/Akt pathway rapidly (within minutes) phosphorylate and inhibit activity of the Polycomb group protein enhancer of zeste homolog 2 (Ezh2). Over the course of 24 to 48 hours, androgens then induce expression of the microRNA miR-101, which targets Ezh2 messenger RNA (mRNA), leading to a nearly complete loss of Ezh2 protein expression. This long-term androgen-induced loss of Ezh2 actions ultimately results in sustained reduction of the H3K27me3-repressive mark in the promoter region of the Runt-related transcription factor-1 (Runx1) gene, a luteinizing hormone (LH)-induced transcription factor essential for ovulation, leading to increased Runx1 mRNA expression. Accordingly, blocking androgen-induced inhibition of Ezh2 in vivo adversely affects LH-induced Runx1 mRNA expression and subsequent ovulation. Importantly, although estrogen treatment of granulosa cells similarly causes rapid activation of the PI3K/Akt pathway and short-term phosphorylation of Ezh2, it does not induce miR-101 expression and thereby does not reduce overall Ezh2 expression, demonstrating the androgen specificity of long-term Ezh2 suppression. Thus, this study provides insight regarding how androgen-induced extranuclear kinase signaling and intranuclear transcription through Ezh2 modifications may influence the expression pattern of genes, ultimately affecting various downstream physiological processes.
Collapse
Affiliation(s)
- Xiaoting Ma
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Emily Hayes
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Anindita Biswas
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Christina Seger
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Hen Prizant
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Stephen R. Hammes
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Aritro Sen
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
71
|
Torres-Ortiz MC, Gutiérrez-Ospina G, Gómez-Chavarín M, Murcia C, Alonso-Morales RA, Perera-Marín G. The presence of VEGF and Notch2 during preantral-antral follicular transition in infantile rats: Anatomical evidence and its implications. Gen Comp Endocrinol 2017; 249:82-92. [PMID: 28495268 DOI: 10.1016/j.ygcen.2017.05.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/25/2017] [Accepted: 05/06/2017] [Indexed: 12/25/2022]
Abstract
Folliculogenesis is a process that depends on angiogenesis, in which VEGF and Notch signaling pathway members are involved. Although this pathway is present in preantral and antral follicular structures during the second stage of folliculogenesis, this association has not been described. Therefore, this study aimed to identify VEGF and Notch2 in ovary structures of infantile rats after induction of follicular development with a gonadotropin stimulus. In order to explore this possibility we analyzed rat ovary morphology from days 10-25 after birth; subsequently, the transition from preantral follicle to an antral stage was analyzed by the induction of follicular development with equine chorionic gonadotropin (eCG) and VEGF and Notch were identified in the rat ovary by fluorescence. The histological analysis revealed that the ovary of a 10-day-old rat has the highest percentage of preantral follicles and based on this a 10IU eCG dose promoted an increase in the number of antral follicles, as well as a decrease in the number of preantral follicles, related to which there was an increase in ovary weight and size. In addition, a higher concentration of circulating estradiol was observed, proliferation of granulosa cells in both follicle groups was stimulated, and the accumulation of VEGF in granulosa and theca cells and in the antral follicle oocyte was increased (p<0.05), whereas the presence of Notch2 was limited to mural granulosa cells, in granulosa cells that formed the cumulus oophorus and in the oocyte of both groups of follicles. The multiple correspondence analysis allowed us to support an association between VEGF and Notch2 during the transition from preantral to antral follicles in the ovary of an infantile rat.
Collapse
Affiliation(s)
- Minerva Carolina Torres-Ortiz
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico; Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico
| | - Gabriel Gutiérrez-Ospina
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico; Coordinación de Psicobiología y Neurociencias, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico
| | - Margarita Gómez-Chavarín
- Departamento de Fisiología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico; Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico
| | - Clara Murcia
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico
| | - Rogelio A Alonso-Morales
- Departamento de Genética y Bioestadística, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico
| | - Gerardo Perera-Marín
- Departamento de Reproducción, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico; Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico.
| |
Collapse
|
72
|
Morales-Ledesma L, Díaz Ramos JA, Trujillo Hernández A. Polycystic ovary syndrome induced by exposure to testosterone propionate and effects of sympathectomy on the persistence of the syndrome. Reprod Biol Endocrinol 2017; 15:50. [PMID: 28693534 PMCID: PMC5504549 DOI: 10.1186/s12958-017-0267-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/20/2017] [Indexed: 11/10/2022] Open
Affiliation(s)
- Leticia Morales-Ledesma
- 0000 0001 2159 0001grid.9486.3Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM, AP 9-020, CP 15000 México, D. F, México
| | - Juan Antonio Díaz Ramos
- 0000 0001 2159 0001grid.9486.3Biology of Reproduction Research Unit, Physiology of Reproduction Laboratory, Facultad de Estudios Superiores Zaragoza, UNAM, AP 9-020, CP 15000 México, D. F, México
| | - Angélica Trujillo Hernández
- 0000 0001 2112 2750grid.411659.eBenemérita Universidad Autónoma de Puebla, Facultad de Ciencias Biológicas, Edificio BIO1, Ciudad Universitaria, Boulevard Valsequillo y Avenida San Claudio S/N. C.P, 72570 Puebla, Puebla México
| |
Collapse
|
73
|
Hsu JY, Sikora MJ. CRISPR Fish Reel in Novel Roles for Estrogen Receptors in Reproduction. Endocrinology 2017; 158:2082-2083. [PMID: 28881866 PMCID: PMC5505223 DOI: 10.1210/en.2017-00417] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 05/08/2017] [Indexed: 11/19/2022]
Affiliation(s)
- Jessica Y Hsu
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| | - Matthew J Sikora
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045
| |
Collapse
|
74
|
Santamaría CG, Rodriguez HA, Abud JE, Rivera OE, Muñoz-de-Toro M, Luque EH. Impaired ovarian response to exogenous gonadotropins in female rat offspring born to mothers perinatally exposed to Bisphenol A. Reprod Toxicol 2017. [PMID: 28648634 DOI: 10.1016/j.reprotox.2017.06.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The ovary is sensitive to disruption by the environmental estrogen Bisphenol A (BPA). Our aim was to investigate whether perinatal exposure to BPA (50μg/kgday), orally administered, affects ovarian response to exogenous gonadotrophins (PMSG or PMSG+hCG) in prepubertal female offspring. An altered response to gonadotrophins was observed in BPA-exposed rats. Increased proportion of antral follicles, altered levels of ovarian steroidogenic enzymes, gonadotropin receptors, AR and ERβ were observed in PMSG group. Besides that, in response to PMSG+hCG, a persistent high Fshr mRNA expression and a decreased number of follicles with high expression of PR before ovulation were observed. After ovulation, there was an increase in antral atretic follicles, reduced Lhcgr mRNA expression and high serum levels of E2. Therefore, an early exposure to a low dose of BPA during perinatal period induces ovarian changes leading to an altered response to exogenous gonadotropin treatment later in life.
Collapse
Affiliation(s)
- C G Santamaría
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina
| | - H A Rodriguez
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina.
| | - J E Abud
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina
| | - O E Rivera
- Instituto de Investigación Agropecuaria, Ambiente y Salud (IIPAAS), Facultad de Ciencias Agrarias, Universidad Nacional de Lomas de Zamora, Buenos Aires, Argentina
| | - M Muñoz-de-Toro
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina
| | - E H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina
| |
Collapse
|
75
|
Shapiro AJ, Darmon SK, Barad DH, Albertini DF, Gleicher N, Kushnir VA. Effect of race and ethnicity on utilization and outcomes of assisted reproductive technology in the USA. Reprod Biol Endocrinol 2017; 15:44. [PMID: 28595591 PMCID: PMC5465464 DOI: 10.1186/s12958-017-0262-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 05/30/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The purpose of this study was to determine the utilization and live birth rates of assisted reproductive technology (ART) modalities among various racial and ethnic groups in recent years. METHODS We reviewed ART data reported to the Society for Assisted Reproductive Technologies Clinic Outcome Reporting System (SART CORS) for autologous ART and third-party ART (3ART) cycles which involved donor oocytes, sperm, embryos and gestational carrier, performed in the U.S. between 2004 and 2013. To gauge demand by various racial/ethnic groups for ART services, we examined fertility rates and demographics of the entire U.S. birth cohort over the same time interval. RESULTS Of 1,132,844 autologous ART cycles 335,462 resulted in a live birth (29.6%). An additional, 217,030 3ART cycles resulted in 86,063 live births (39.7%). Hispanic and Black women demonstrated high fertility and lower utilization rates of autologous ART and 3ART. Caucasian and Asian women exhibited lower fertility rates and higher autologous ART and 3ART utilization. Autologous ART resulted in higher live birth rates among Caucasian and Hispanic women and lower rates among Asian and especially Black women. 3ART improved live birth rates in all races/ethnicities, though Black women experienced lower live birth rates with most modalities. Spontaneous abortion rates were higher among Black women following autologous ART and some 3ART modalities than those among Caucasian women. CONCLUSION Utilization of ART is inversely related to fertility rates. Autologous ART produces lower live birth rates among Asian and Black women. 3ART results in relatively low live birth rates among Black women. TRIAL REGISTRATION SART CORS #57 , Registered 5/14/2015.
Collapse
Affiliation(s)
- Alice J. Shapiro
- Center for Human Reproduction, 21 East 69th Street, New York, NY 10021 USA
- Department of Obstetrics, Gynecology & Women’s Health, Rutgers New Jersey Medical School, Newark, NJ USA
| | - Sarah K. Darmon
- Center for Human Reproduction, 21 East 69th Street, New York, NY 10021 USA
| | - David H. Barad
- Center for Human Reproduction, 21 East 69th Street, New York, NY 10021 USA
- Foundation for Reproductive Medicine, New York, NY USA
| | - David F. Albertini
- Center for Human Reproduction, 21 East 69th Street, New York, NY 10021 USA
- Stem Cell Biology and Molecular Embryology Laboratory, The Rockefeller University, New York, NY USA
| | - Norbert Gleicher
- Center for Human Reproduction, 21 East 69th Street, New York, NY 10021 USA
- Foundation for Reproductive Medicine, New York, NY USA
- Stem Cell Biology and Molecular Embryology Laboratory, The Rockefeller University, New York, NY USA
- Department of Obstetrics and Gynecology, University of Vienna School of Medicine, Vienna, Austria
| | - Vitaly A. Kushnir
- Center for Human Reproduction, 21 East 69th Street, New York, NY 10021 USA
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC USA
| |
Collapse
|
76
|
Lebbe M, Taylor AE, Visser JA, Kirkman-Brown JC, Woodruff TK, Arlt W. The Steroid Metabolome in the Isolated Ovarian Follicle and Its Response to Androgen Exposure and Antagonism. Endocrinology 2017; 158:1474-1485. [PMID: 28323936 PMCID: PMC5460835 DOI: 10.1210/en.2016-1851] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 02/17/2017] [Indexed: 11/29/2022]
Abstract
The ovarian follicle is a major site of steroidogenesis, crucially required for normal ovarian function and female reproduction. Our understanding of androgen synthesis and metabolism in the developing follicle has been limited by the sensitivity and specificity issues of previously used assays. Here we used liquid chromatography-tandem mass spectrometry to map the stage-dependent endogenous steroid metabolome in an encapsulated in vitro follicle growth system, from murine secondary through antral follicles. Furthermore, follicles were cultured in the presence of androgen precursors, nonaromatizable active androgen, and androgen receptor (AR) antagonists to assess effects on steroidogenesis and follicle development. Cultured follicles showed a stage-dependent increase in endogenous androgen, estrogen, and progesterone production, and incubations with the sex steroid precursor dehydroepiandrosterone revealed the follicle as capable of active androgen synthesis at early developmental stages. Androgen exposure and antagonism demonstrated AR-mediated effects on follicle growth and antrum formation that followed a biphasic pattern, with low levels of androgens inducing more rapid follicle maturation and high doses inhibiting oocyte maturation and follicle growth. Crucially, our study provides evidence for an intrafollicular feedback circuit regulating steroidogenesis, with decreased follicle androgen synthesis after exogenous androgen exposure and increased androgen output after additional AR antagonist treatment. We propose that this feedback circuit helps maintain an equilibrium of androgen exposure in the developing follicle. The observed biphasic response of follicle growth and function in increasing androgen supplementations has implications for our understanding of polycystic ovary syndrome pathophysiology and the dose-dependent utility of androgens in in vitro fertilization settings.
Collapse
Affiliation(s)
- Marie Lebbe
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Angela E. Taylor
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Jenny A. Visser
- Department of Internal Medicine, Erasmus MC, 3015 CN Rotterdam, The Netherlands
| | - Jackson C. Kirkman-Brown
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Tommy’s National Centre for Miscarriage Research, Birmingham Women’s Hospital NHS Foundation Trust, Birmingham B15 2TH, United Kingdom
| | - Teresa K. Woodruff
- Division of Reproductive Science in Medicine, Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Wiebke Arlt
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, United Kingdom
| |
Collapse
|
77
|
Lim JJ, Han CY, Lee DR, Tsang BK. Ring Finger Protein 6 Mediates Androgen-Induced Granulosa Cell Proliferation and Follicle Growth via Modulation of Androgen Receptor Signaling. Endocrinology 2017; 158:993-1004. [PMID: 28324045 DOI: 10.1210/en.2016-1866] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 01/19/2017] [Indexed: 01/18/2023]
Abstract
The destiny of the ovarian follicle (growth or atresia) is tightly regulated by the actions and interactions of endocrine, paracrine, and autocrine factors. Although androgens are known to be important in the regulation of folliculogenesis, whether they facilitate or suppress follicular growth has been controversial, and the mechanisms involved are not fully understood. Moreover, the role and regulation of androgen receptor (AR) in mediating androgen signaling during follicular development is not clear. Here, we report that the active androgen dihydrotestosterone upregulates the expression of AR and its E3 ligase ring finger protein 6 (RNF6), increasing site-specific AR polyubiquitination and AR transcriptional activity for soluble Kit ligand (sKit-L) expression in preantral follicle growth. RNF6 silencing suppressed dihydrotestosterone-induced AR ubiquitination (lysine residue 63) and proliferation and suppressed apoptosis in preantral granulosa cells, with these responses being overcome by the presence of exogenous sKit-L. Taken together, our findings support the notion that RNF6 plays an important role in androgen-induced, follicle-stage-dependent follicle growth and that it acts by facilitating AR-mediated granulosa cell sKit-L expression and proliferation. Our findings offer insights into the regulatory mechanism of androgen action in ovarian follicular growth.
Collapse
Affiliation(s)
- Jung Jin Lim
- Department of Obstetrics and Gynecology and Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Chae Young Han
- Department of Obstetrics and Gynecology and Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Dong Ryul Lee
- Fertility Center of CHA Gangnam Medical Center, College of Medicine, CHA University, Seoul, Korea
- Department of Biomedical Science, College of Life Science, CHA University, Seoul, Korea
| | - Benjamin K Tsang
- Department of Obstetrics and Gynecology and Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macao, China
| |
Collapse
|
78
|
Sellix MT, Sen A. Finding the Right Balance: Androgens at the Tipping Point of Fertility and Metabolism in Women. Endocrinology 2017; 158:467-469. [PMID: 28430914 PMCID: PMC5460785 DOI: 10.1210/en.2016-1959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 11/19/2022]
Affiliation(s)
- Michael T Sellix
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| | - Aritro Sen
- Division of Endocrinology and Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642
| |
Collapse
|
79
|
Gleicher N, Kushnir VA, Darmon SK, Wang Q, Zhang L, Albertini DF, Barad DH. New PCOS-like phenotype in older infertile women of likely autoimmune adrenal etiology with high AMH but low androgens. J Steroid Biochem Mol Biol 2017; 167:144-152. [PMID: 27993552 DOI: 10.1016/j.jsbmb.2016.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 12/13/2016] [Accepted: 12/14/2016] [Indexed: 01/02/2023]
Abstract
How anti-Müllerian hormone (AMH) and testosterone (T) interrelate in infertile women is currently largely unknown. We, therefore, in a retrospective cohort study investigated how infertile women with high-AMH (AMH ≥75th quantile; n=144) and with normal-AMH (25th-75th quantile; n=313), stratified for low-T (total testosterone ≤19.0ng/dL), normal-T (19.0-29.0ng/dL) and high-T (>29.0ng/dL) phenotypically behaved. Patient age, follicle stimulating hormone (FSH), dehyroepiandrosterone (DHEA), DHEA sulphate (DHEAS), cortisol (C), adrenocorticotrophic hormone (ACTH), IVF outcomes, as well as inflammatory and immune panels were then compared between groups, with AMH and T as variables. We identified a previously unknown infertile PCOS-like phenotype, characterized by high-AMH but, atypically, low-T, with predisposition toward autoimmunity. It presents with incompatible high-AMH and low-T (<19.0ng/dL), is restricted to lean PCOS-like patients, presenting delayed for tertiary fertility services. Since also characterized by low DHEAS, low-T is likely of adrenal origina, and consequence of autoimmune adrenal insufficiency since also accompanied by low-C and evidence of autoimmunity. DHEA supplementation in such patients equalizes low- to normal-T and normalizes IVF cycle outcomes. Once recognized, this high-AMH/low-T phenotype is surprisingly common in tertiary fertility centers but, currently, goes unrecognized. Its likely adrenal autoimmune etiology offers interesting new directions for investigations of adrenals control over ovarian function via adrenal androgen production.
Collapse
Affiliation(s)
- Norbert Gleicher
- The Center for Human Reproduction, New York, NY, 10021, United States; The Foundation for Reproductive Medicine, New York, NY 10020, United States; Stem Cell Biology and Molecular Embryology Laboratory, Rockefeller University, New York, NY 10016, United States; Department of Obstetrics and Gynecology, Vienna University School of Medicine, 1090 Vienna, Austria.
| | - Vitaly A Kushnir
- The Center for Human Reproduction, New York, NY, 10021, United States; Department of Obstetrics and Gynecology, Wake Forest University, Winston Salem, NC 27101, United States
| | - Sarah K Darmon
- The Center for Human Reproduction, New York, NY, 10021, United States
| | - Qi Wang
- The Center for Human Reproduction, New York, NY, 10021, United States
| | - Lin Zhang
- The Center for Human Reproduction, New York, NY, 10021, United States
| | - David F Albertini
- The Center for Human Reproduction, New York, NY, 10021, United States; Stem Cell Biology and Molecular Embryology Laboratory, Rockefeller University, New York, NY 10016, United States
| | - David H Barad
- The Center for Human Reproduction, New York, NY, 10021, United States; The Foundation for Reproductive Medicine, New York, NY 10020, United States
| |
Collapse
|
80
|
Sharma RP, Schuhmacher M, Kumar V. Review on crosstalk and common mechanisms of endocrine disruptors: Scaffolding to improve PBPK/PD model of EDC mixture. ENVIRONMENT INTERNATIONAL 2017; 99:1-14. [PMID: 27697394 DOI: 10.1016/j.envint.2016.09.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 09/18/2016] [Accepted: 09/19/2016] [Indexed: 06/06/2023]
Abstract
Endocrine disruptor compounds (EDCs) are environment chemicals that cause harmful effects through multiple mechanisms, interfering with hormone system resulting in alteration of homeostasis, reproduction and developmental effect. Many of these EDCs have concurrent exposure with crosstalk and common mechanisms which may lead to dynamic interactions. To carry out risk assessment of EDCs' mixture, it is important to know the detailed toxic pathway, crosstalk of receptor and other factors like critical window of exposure. In this review, we summarize the major mechanism of actions of EDCs with the different/same target organs interfering with the same/different class of hormone by altering their synthesis, metabolism, binding and cellular action. To show the impact of EDCs on life stage development, a case study on female fertility affecting germ cell is illustrated. Based on this summarized discussion, major groups of EDCs are classified based on their target organ, mode of action and potential risk. Finally, a conceptual model of pharmacodynamic interaction is proposed to integrate the crosstalk and common mechanisms that modulate estrogen into the predictive mixture dosimetry model with dynamic interaction of mixture. This review will provide new insight for EDCs' risk assessment and can be used to develop next generation PBPK/PD models for EDCs' mixture analysis.
Collapse
Affiliation(s)
- Raju Prasad Sharma
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
| | - Marta Schuhmacher
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain
| | - Vikas Kumar
- Center of Environmental Food and Toxicological Technology (TecnATox), Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, Catalonia, Spain.
| |
Collapse
|
81
|
A model of ovulatory regulation examining the effects of insulin-mediated testosterone production on ovulatory function. J Theor Biol 2017; 416:149-160. [PMID: 28069449 DOI: 10.1016/j.jtbi.2017.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/20/2016] [Accepted: 01/04/2017] [Indexed: 12/16/2022]
Abstract
Polycystic ovary syndrome (PCOS), a common cause of infertility in women, is often accompanied by abnormal reproductive and metabolic hormone levels. Specifically, androgens such as testosterone are elevated in many PCOS women, and the syndrome itself is frequently associated with insulin resistance, which leads to hyperinsulinemia, i.e., elevated insulin. Although the precise role of insulin in ovulatory function is unclear, its role in ovulatory dysfunction is often linked to the effects of increased ovarian androgen production. We present a mathematical model of the menstrual cycle that incorporates regulation by the pituitary-ovarian axis and mechanisms of ovarian testosterone production. We determine a physiological role for testosterone in the normal ovulatory cycle and study the role of hyperinsulinemia in pathological regulation of the cycle. Model results indicate increased ovulatory disruption with elevated insulin-mediated testosterone production and suggest that variations in the response of ovarian follicles to essential signals can alter the degree to which hyperinsulinemia disrupts the ovulatory cycle. The model also provides insight into the various PCOS phenotypes and the severity of ovulatory dysfunction.
Collapse
|
82
|
Komatsu K, Masubuchi S. Observation of the dynamics of follicular development in the ovary. Reprod Med Biol 2016; 16:21-27. [PMID: 29259446 PMCID: PMC5715870 DOI: 10.1002/rmb2.12010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/09/2016] [Indexed: 12/26/2022] Open
Abstract
The number of ovulated oocytes is different among mammals but does not vary much within the same species. In order to sustain periodic ovulation, follicular development must be coordinated at the tissue level. Elucidating the regulatory mechanisms of follicular development is difficult because the ovary has a complicated structure and it takes a long time for primordial follicles to develop into Graafian follicles. Therefore, it is not possible to observe follicular development by conventional experiments. The authors previously developed a new ovarian tissue culture method that enabled the observation of follicular development from the early follicle stage. These findings indicated that follicular interactions are important in regulating follicular development and ovulation. This review describes the current methods of observing follicular development in the ovary and the regulatory mechanisms of follicular development.
Collapse
Affiliation(s)
- Kouji Komatsu
- Department of PhysiologyAichi Medical UniversityAichiJapan
| | | |
Collapse
|
83
|
Braakhekke M, Mol F, Mastenbroek S, Mol BWJ, van der Veen F. Equipoise and the RCT. Hum Reprod 2016; 32:257-260. [DOI: 10.1093/humrep/dew286] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 10/13/2016] [Accepted: 10/26/2016] [Indexed: 11/13/2022] Open
|
84
|
Rosenfield RL, Ehrmann DA. The Pathogenesis of Polycystic Ovary Syndrome (PCOS): The Hypothesis of PCOS as Functional Ovarian Hyperandrogenism Revisited. Endocr Rev 2016; 37:467-520. [PMID: 27459230 PMCID: PMC5045492 DOI: 10.1210/er.2015-1104] [Citation(s) in RCA: 734] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 07/20/2016] [Indexed: 02/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) was hypothesized to result from functional ovarian hyperandrogenism (FOH) due to dysregulation of androgen secretion in 1989-1995. Subsequent studies have supported and amplified this hypothesis. When defined as otherwise unexplained hyperandrogenic oligoanovulation, two-thirds of PCOS cases have functionally typical FOH, characterized by 17-hydroxyprogesterone hyperresponsiveness to gonadotropin stimulation. Two-thirds of the remaining PCOS have FOH detectable by testosterone elevation after suppression of adrenal androgen production. About 3% of PCOS have a related isolated functional adrenal hyperandrogenism. The remaining PCOS cases are mild and lack evidence of steroid secretory abnormalities; most of these are obese, which we postulate to account for their atypical PCOS. Approximately half of normal women with polycystic ovarian morphology (PCOM) have subclinical FOH-related steroidogenic defects. Theca cells from polycystic ovaries of classic PCOS patients in long-term culture have an intrinsic steroidogenic dysregulation that can account for the steroidogenic abnormalities typical of FOH. These cells overexpress most steroidogenic enzymes, particularly cytochrome P450c17. Overexpression of a protein identified by genome-wide association screening, differentially expressed in normal and neoplastic development 1A.V2, in normal theca cells has reproduced this PCOS phenotype in vitro. A metabolic syndrome of obesity-related and/or intrinsic insulin resistance occurs in about half of PCOS patients, and the compensatory hyperinsulinism has tissue-selective effects, which include aggravation of hyperandrogenism. PCOS seems to arise as a complex trait that results from the interaction of diverse genetic and environmental factors. Heritable factors include PCOM, hyperandrogenemia, insulin resistance, and insulin secretory defects. Environmental factors include prenatal androgen exposure and poor fetal growth, whereas acquired obesity is a major postnatal factor. The variety of pathways involved and lack of a common thread attests to the multifactorial nature and heterogeneity of the syndrome. Further research into the fundamental basis of the disorder will be necessary to optimally correct androgen levels, ovulation, and metabolic homeostasis.
Collapse
Affiliation(s)
- Robert L Rosenfield
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago Pritzker School of Medicine, Chicago, Illinois 60637
| | - David A Ehrmann
- Section of Adult and Pediatric Endocrinology, Diabetes, and Metabolism, The University of Chicago Pritzker School of Medicine, Chicago, Illinois 60637
| |
Collapse
|
85
|
Hayes E, Kushnir V, Ma X, Biswas A, Prizant H, Gleicher N, Sen A. Intra-cellular mechanism of Anti-Müllerian hormone (AMH) in regulation of follicular development. Mol Cell Endocrinol 2016; 433:56-65. [PMID: 27235859 DOI: 10.1016/j.mce.2016.05.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 05/24/2016] [Accepted: 05/24/2016] [Indexed: 11/21/2022]
Abstract
Anti-Müllerian hormone (AMH) is a member of the transforming growth factor-β superfamily and plays a crucial role in testicular and ovarian functions. In clinical practice, AMH is used as a diagnostic and/or prognostic marker in women in association with ovulation induction and in various pathophysiological conditions. Despite widespread clinical use of AMH, our mechanistic understanding of AMH actions in regulating follicular development is limited. Using a mouse model, we in this study report that in vivo AMH treatment while stalls follicular development and inhibits ovulation, also prevents follicular atresia. We further show that these AMH actions are mediated through induction of two miRNAs, miR-181a and miR-181b, which regulate various aspects of FSH signaling and follicular growth, ultimately affecting downstream gene expression and folliculogenesis. We also report that in this mouse model AMH pre-treatment prior to superovulation improves oocyte yield. These studies, therefore, offer new mechanistic insight into AMH actions in folliculogenesis and point toward potential utilization of AMH as a therapeutic agent.
Collapse
Affiliation(s)
- Emily Hayes
- Division of Endocrinology & Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Vitaly Kushnir
- Center for Human Reproduction, New York, NY 10021, USA; Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Xiaoting Ma
- Division of Endocrinology & Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Anindita Biswas
- Division of Endocrinology & Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Hen Prizant
- Division of Endocrinology & Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Norbert Gleicher
- Center for Human Reproduction, New York, NY 10021, USA; The Rockefeller University, New York, NY 10065, USA
| | - Aritro Sen
- Division of Endocrinology & Metabolism, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Center for Human Reproduction, New York, NY 10021, USA.
| |
Collapse
|
86
|
Gleicher N, Kushnir VA, Albertini DF, Barad DH. Improvements in IVF in women of advanced age. J Endocrinol 2016; 230:F1-6. [PMID: 27154334 DOI: 10.1530/joe-16-0105] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 05/06/2016] [Indexed: 11/08/2022]
Abstract
Women above age 40 years in the US now represent the most rapidly growing age group having children. Patients undergoing in vitro fertilization (IVF) are rapidly aging in parallel. Especially where egg donations are legal, donation cycles, therefore, multiply more rapidly than autologous IVF cycles. The donor oocytes, however, are hardly ever a preferred patient choice. Since with use of own eggs, live birth rates decline with advancing age but remain stable (and higher) with donor eggs, older patients always face the difficult and very personal choice between poorer chances with own and better chances with donor oocytes. Physician contribution to this decision should in our opinion be restricted to accurate outcome information for both options. Achievable pregnancy and live birth rates in older women are, however, frequently underestimated, thereby mistakenly biasing fertility providers, private insurance companies and even regulatory government agencies. Restriction on access to IVF for older women is then often the consequence. In this review, we summarize the limited published data on best treatments of 'older' ovaries, while also addressing treatment approaches that should be avoided in older women. This focused review, therefore, to a degree is subjective. Research addressing aging ovaries in IVF has been disappointingly sparse, and has in our opinion too heavily concentrated on methods of embryo selection (ES), which, especially in older women, not only fail to improve IVF outcomes, but actually, negatively affect live birth chances. We conclude that, aside from breakthroughs in gamete creation, only pharmacological interventions into early (small growing follicle stages) follicle maturation will offer new potential to positively impact oocyte and embryo quality and, therefore, IVF outcomes. Research, therefore, should be accordingly redirected.
Collapse
Affiliation(s)
- Norbert Gleicher
- The Center for Human ReproductionNew York, New York, USA The Foundation for Reproductive MedicineNew York, New York, USA The Brivanlou Stem Cell Biology and Molecular Embryology LaboratoryThe Rockefeller University, New York, New York, USA
| | - Vitaly A Kushnir
- The Center for Human ReproductionNew York, New York, USA Department of Obstetrics and GynecologyWake Forest University, Winston Salem, North Carolina, USA
| | - David F Albertini
- The Center for Human ReproductionNew York, New York, USA Department of Molecular and Integrative PhysiologyThe University of Kansas Medical Center, Kansas City, Kansas, USA
| | - David H Barad
- The Center for Human ReproductionNew York, New York, USA The Foundation for Reproductive MedicineNew York, New York, USA
| |
Collapse
|
87
|
Gibson DA, Simitsidellis I, Saunders PTK. Regulation of androgen action during establishment of pregnancy. J Mol Endocrinol 2016; 57:R35-47. [PMID: 27067639 DOI: 10.1530/jme-16-0027] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 04/11/2016] [Indexed: 01/02/2023]
Abstract
During the establishment of pregnancy, the ovarian-derived hormones progesterone and oestradiol regulate remodelling of the endometrium to promote an environment that is able to support and maintain a successful pregnancy. Decidualisation is characterised by differentiation of endometrial stromal cells that secrete growth factors and cytokines that regulate vascular remodelling and immune cell influx. This differentiation process is critical for reproduction, and inadequate decidualisation is implicated in the aetiology of pregnancy disorders such as foetal growth restriction and preeclampsia. In contrast to progesterone and oestradiol, the role of androgens in regulating endometrial function is poorly understood. Androgen receptors are expressed in the endometrium, and androgens are reported to regulate both the transcriptome and the secretome of endometrial stromal cells. In androgen-target tissues, circulating precursors are activated to mediate local effects, and recent studies report that steroid concentrations detected in endometrial tissue are distinct to those detected in the peripheral circulation. New evidence suggests that decidualisation results in dynamic changes in the expression of androgen biosynthetic enzymes, highlighting a role for pre-receptor regulation of androgen action during the establishment of pregnancy. These results suggest that such enzymes could be future therapeutic targets for the treatment of infertility associated with endometrial dysfunction. In conclusion, these data support the hypothesis that androgens play a beneficial role in regulating the establishment and maintenance of pregnancy. Future studies should be focussed on investigating the safety and efficacy of androgen supplementation with the potential for utilisation of novel therapeutics, such as selective androgen receptor modulators, to improve reproductive outcomes in women.
Collapse
Affiliation(s)
- Douglas A Gibson
- Medical Research Council Centre for Inflammation ResearchQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ioannis Simitsidellis
- Medical Research Council Centre for Inflammation ResearchQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Philippa T K Saunders
- Medical Research Council Centre for Inflammation ResearchQueen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
88
|
Early ovarian follicular development in prepubertal Wistar rats acutely exposed to androgens. J Dev Orig Health Dis 2016; 7:384-90. [DOI: 10.1017/s2040174416000222] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Androgens may directly modulate early ovarian follicular development in preantral stages and androgen excess before puberty may disrupt this physiological process. Therefore, the aim of this study was to investigate the dynamics of follicular morphology and circulating androgen and estradiol levels in prepubertal Wistar rats acutely exposed to androgens. Prepubertal female Wistar rats were distributed into three groups: control, equine chorionic gonadotropin (eCG) intervention and eCG plus dehydroepiandrosterone (DHEA) intervention (eCG+DHEA). Serum DHEA, testosterone and estradiol levels were determined, and ovarian morphology and morphometry were assessed. The eCG+DHEA group presented increased serum estradiol and testosterone levels as compared with the control group (P<0.01), and higher serum DHEA concentration v. the eCG-only and control groups (P<0.01). In addition, the eCG+DHEA group had a higher number of, and larger-sized, primary and secondary follicles as compared with the control group (P<0.05). The eCG group presented intermediate values for number and size of primary and secondary follicles, without significant differences as compared with the other two groups. The number of antral follicles was higher in the eCG+DHEA and eCG groups v. controls (P<0.05). The number of primordial, atretic and cystic follicles were similar in all groups. In conclusion, the present experimental model using an acute eCG+DHEA intervention was useful to investigate events involved in initial follicular development under hyperandrogenic conditions, and could provide a reliable tool to study defective follicular development with possible deleterious reproductive consequences later in life.
Collapse
|
89
|
Abstract
Dicer1 is an RNase III enzyme necessary for microRNA (miRNA) biogenesis, as it cleaves pre-miRNAs into mature miRNAs. miRNAs are important regulators of gene expression. In recent years, several miRNA-independent roles of Dicer1 have been identified. They include the production of endogenous small interfering RNAs, detoxifying retrotransposon-derived transcripts, and binding to new targets; messenger RNAs and long noncoding RNAs. Further, in this review, the functional significance of Dicer1 in the male reproductive tract is discussed. Conditional Dicer1 knock-out mouse models have demonstrated a requisite role for Dicer in male fertility. Deletion of Dicer1 from somatic or germ cells in the testis cause spermatogenic problems rendering male mice infertile. The lack of Dicer1 in the proximal epididymis causes dedifferentiation of the epithelium, with unbalanced sex steroid receptor expression, defects in epithelial lipid homeostasis, and subsequent male infertility. In addition, Dicer1 ablation from the prostate leads to increased apoptosis of the differentiated luminal cells, followed by epithelial hypotrophy of the ventral prostate. However, further studies are needed to clarify which functions of Dicer1 are responsible for the observed phenotypes in the male reproductive tract.
Collapse
Affiliation(s)
| | - Petra Sipilä
- Department of Physiology, Institute of Biomedicine; Laboratory Animal Centre, University of Helsinki, Helsinki, Finland
| |
Collapse
|
90
|
Gleicher N, Kushnir VA, Weghofer A, Barad DH. The importance of adrenal hypoandrogenism in infertile women with low functional ovarian reserve: a case study of associated adrenal insufficiency. Reprod Biol Endocrinol 2016; 14:23. [PMID: 27112552 PMCID: PMC4845439 DOI: 10.1186/s12958-016-0158-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 04/20/2016] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Low testosterone (T), whether due to ovarian and/or adrenal insufficiency, usually results in poor follicle maturation at small growing follicle stages. The consequence is a phenotype of low functional ovarian reserve (LFOR), characterized by poor granulosa cell mass, low anti-Müllerian hormone and estradiol but rising follicle stimulating hormone. Such hypoandrogenism can be of ovarian and/or adrenal origin. Dehydroepiandrosterone sulfate (DHEAS) is exclusively produced by adrenals and, therefore, reflects adrenal androgen production in the zona reticularis. We here determined in a case study of infertile women with LFOR the presence of adrenal hypoandrogenism, its effects on ovarian function, and the possibility of presence of concomitant adrenal insufficiency (AI), thus reflecting insufficiency of all three adrenal cortical zonae. METHODS We searched our center's anonymized electronic research database for women with LFOR, who were also characterized by peripheral adrenal hypoandrogenemia (total testosterone < 16.9 ng/dL) and low DHEAS (<76.0 μg/dL). Among 225 women with LFOR, we identified 29 (12.9 %). The adrenal function of so identified women were further investigated with morning cortisol and ACTH levels and/or standard ACTH stimulation tests. We also determined the prevalence of classical AI (insufficiency glucocorticoid production by zona fasciculata) in hypoandrogenic women with LFOR, and impact of adrenal hypoandrogenism on ovaries. RESULTS Among 14/28 women with adrenal hypoandrogenism due to insufficiency of the zona reticularis available for follow up, 4 (28.6 %) also demonstrated previously unrecognized classical primary, secondary or tertiary AI due to insufficiency of the zona fasciculata. An additional patient with presenting diagnosis of seemingly primary ovarian insufficiency (POI), demonstrated extremely low T and DHEAS levels, a diagnosis of Addison's disease, and was on glucocorticoid but not androgen supplementation. As her dramatic improvement in ovarian function criteria after androgen supplementation confirmed, her correct diagnosis, therefore, was actually secondary ovarian insufficiency (SOI) due to adrenal hypoandrogenism. CONCLUSIONS Women with LFOR, characterized by low T and DHEAS, are also at risk for AI, while women with AI may be at risk for adrenal induced hypoandrogenism and, therefore, SOI. A currently undetermined percentage of POI patients actually are, likely, affected by SOI, a for prognostic reasons highly significant difference in diagnosis.
Collapse
Affiliation(s)
- Norbert Gleicher
- The Center for Human Reproduction, 21 East 69th Street, New York, NY 10021 USA
- The Foundation for Reproductive Medicine, New York, NY USA
- Stem Cell Biology and Molecular Embryology Laboratory, The Rockefeller University, New York, NY USA
| | - Vitaly A. Kushnir
- The Center for Human Reproduction, 21 East 69th Street, New York, NY 10021 USA
- Department of Obstetrics and Gynecology, Wake Forest University, Winston-Salem, NC 27106 USA
| | - Andrea Weghofer
- The Center for Human Reproduction, 21 East 69th Street, New York, NY 10021 USA
- Vienna University School of Medicine, Vienna, Austria
| | - David H. Barad
- The Center for Human Reproduction, 21 East 69th Street, New York, NY 10021 USA
- The Foundation for Reproductive Medicine, New York, NY USA
- Department of Obstetrics and Gynecology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| |
Collapse
|
91
|
Johansson HKL, Jacobsen PR, Hass U, Svingen T, Vinggaard AM, Isling LK, Axelstad M, Christiansen S, Boberg J. Perinatal exposure to mixtures of endocrine disrupting chemicals reduces female rat follicle reserves and accelerates reproductive aging. Reprod Toxicol 2016; 61:186-94. [PMID: 27049580 DOI: 10.1016/j.reprotox.2016.03.045] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/03/2016] [Accepted: 03/28/2016] [Indexed: 12/27/2022]
Abstract
Exposure to endocrine disrupting chemicals (EDCs) during development can have negative consequences later in life. In this study we investigated the effect of perinatal exposure to mixtures of human relevant EDCs on the female reproductive system. Rat dams were exposed to a mixture of phthalates, pesticides, UV-filters, bisphenol A, butylparaben, as well as paracetamol. The compounds were tested together (Totalmix) or in subgroups with anti-androgenic (AAmix) or estrogenic (Emix) potentials. Paracetamol was tested separately. In pre-pubertal rats, a significant reduction in primordial follicle numbers was seen in AAmix and PM groups, and reduced plasma levels of prolactin was seen in AAmix. In one-year-old animals, the incidence of irregular estrous cycles was higher after Totalmix-exposure and reduced ovary weights were seen in Totalmix, AAmix, and PM groups. These findings resemble premature ovarian insufficiency in humans, and raises concern regarding potential effects of mixtures of EDCs on female reproductive function.
Collapse
Affiliation(s)
- Hanna Katarina Lilith Johansson
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark
| | - Pernille Rosenskjold Jacobsen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark
| | - Ulla Hass
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark
| | - Terje Svingen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark
| | - Anne Marie Vinggaard
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark
| | - Louise Krag Isling
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark
| | - Marta Axelstad
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark
| | - Sofie Christiansen
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark
| | - Julie Boberg
- Division of Diet, Disease Prevention and Toxicology, National Food Institute, Technical University of Denmark, Søborg DK-2860, Denmark.
| |
Collapse
|
92
|
Gleicher N, Seier K, Kushnir VA, Weghofer A, Wu YG, Wang Q, Albertini DF, Barad DH. Associations between peripheral androgens and cortisol in infertile women. J Steroid Biochem Mol Biol 2016; 158:82-89. [PMID: 26804970 DOI: 10.1016/j.jsbmb.2016.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/14/2015] [Accepted: 01/09/2016] [Indexed: 11/16/2022]
Abstract
Testosterone has in recent years been proven essential for normal growth and maturation of small growing follicles. Concomitantly, low functional ovarian reserve (LFOR), characterized by a small growing follicle pool, has been associated with low testosterone levels, which can be of ovarian and/or adrenal origin. In this study we, therefore, investigated whether peripheral sex steroid precursors and testosterone levels potentially reflect on adrenal function. In a retrospective cohort study of 355 consecutive infertile women, who presented to an academically affiliated fertility center in New York City, we investigated in a series of statistical models whether low peripheral sex steroid precursors and testosterone are associated with peripheral cortisol (C) levels, reflecting adrenal function. To determine potential correlations, we investigated the dehydroepiandrosterone (DHEA), DHEA sulfate (DHEAS), androstenedione (AD), total testosterone (TT), free testosterone (FT); sex hormone binding globulin (SHBG), anti-Müllerian hormone (AMH), thyroid stimulating hormone (TSH) and C in a series of multivariate and logistic regression analyses, utilizing C either as a continuous variable or with cut off <5.0μg/dL, and TT only as a continuous variable. Practically all models demonstrated significant predictability of peripheral sex hormone precursors for C levels, with DHEA demonstrating the strongest and most consistent predictability as an individual parameter and as part of the DHEAS/DHEA ratio. We conclude that in infertile women peripheral sex hormone precursors, especially DHEA, reflect C levels and, therefore, adrenal function. In infertile women, at all ages low levels of sex hormone precursors, therefore, should be considered indications for further adrenal assessments.
Collapse
Affiliation(s)
- Norbert Gleicher
- The Center for Human Reproduction (CHR), New York, NY, United States; The Foundation for Reproductive Medicine, New York, NY, United States; Stem Cell and Molecular Embryology Laboratory, The Rockefeller University, New York, NY, United States.
| | - Kenneth Seier
- The Center for Human Reproduction (CHR), New York, NY, United States
| | - Vitaly A Kushnir
- The Center for Human Reproduction (CHR), New York, NY, United States; Department of Obstetrics and Gynecology, Wake Forest University, Winston Salem, NC, United States
| | - Andrea Weghofer
- The Center for Human Reproduction (CHR), New York, NY, United States; Department of Obstetrics and Gynecology, Vienna University School of Medicine, Vienna, Austria
| | - Yan-Guang Wu
- The Center for Human Reproduction (CHR), New York, NY, United States
| | - Qi Wang
- The Center for Human Reproduction (CHR), New York, NY, United States
| | - David F Albertini
- The Center for Human Reproduction (CHR), New York, NY, United States; University of Kansas Medical Center, Kansas City, KS, United States
| | - David H Barad
- The Center for Human Reproduction (CHR), New York, NY, United States; The Foundation for Reproductive Medicine, New York, NY, United States; Department of Obstetrics and Gynecology, Albert Einstein College of Medicine, Bronx, New York, NY, United States
| |
Collapse
|
93
|
Santamaría C, Durando M, Muñoz de Toro M, Luque EH, Rodriguez HA. Ovarian dysfunctions in adult female rat offspring born to mothers perinatally exposed to low doses of bisphenol A. J Steroid Biochem Mol Biol 2016; 158:220-230. [PMID: 26658420 DOI: 10.1016/j.jsbmb.2015.11.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/17/2015] [Accepted: 11/25/2015] [Indexed: 01/08/2023]
Abstract
The study of oral exposure to the environmental estrogen bisphenol A (BPA) during the perinatal period and its effects on ovarian functionality in adulthood has generated special interest. Thus, our objective was to investigate ovarian folliculogenesis and steroidogenesis in adult female rat offspring born to mothers exposed to low doses of BPA (BPA50: 50μg/kgday; BPA0.5: 0.5μg/kgday) by the oral route during gestation and breastfeeding. Ovaries from both BPA-treated groups showed reduced primordial follicle recruitment and a greater number of corpora lutea, indicating an increased number of ovulated oocytes, coupled with higher levels of mRNA expression of 3β-hydroxysteroid dehydrogenase and serum progesterone. BPA50-treated animals had lower expression of androgen receptor (AR) at different stages of the growing follicle population. BPA0.5-treated rats evidenced an imbalance of AR expression between primordial/primary follicles, with higher mRNA-follicle-stimulating hormone receptor expression. These results add to the growing evidence that folliculogenesis and steroidogenesis are targets of BPA within the ovary.
Collapse
Affiliation(s)
- Clarisa Santamaría
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina
| | - Milena Durando
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina
| | - Mónica Muñoz de Toro
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina
| | - Enrique H Luque
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina
| | - Horacio A Rodriguez
- Instituto de Salud y Ambiente del Litoral (ISAL, CONICET-UNL), Ciudad Universitaria, Paraje El Pozo s/n, Facultad de Bioquímica y Ciencias Biológicas, Universidad Nacional del Litoral, CP3000 Santa Fe, Argentina.
| |
Collapse
|
94
|
Grzesiak M, Knapczyk-Stwora K, Luck MR, Mobasheri A, Slomczynska M. Effect of Prenatal and Neonatal Anti-Androgen Flutamide Treatment on Aquaporin 5 Expression in the Adult Porcine Ovary. Reprod Domest Anim 2015; 51:105-13. [PMID: 26661749 DOI: 10.1111/rda.12652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 11/12/2015] [Indexed: 02/06/2023]
Abstract
The growth of ovarian follicles is accompanied by fluid-filled antrum formation. Water movement within the follicular wall is predominantly transcellular via membranous water channels named aquaporins (AQPs). Androgens are important regulators of mammalian folliculogenesis, and their prenatal and/or neonatal deficiency affects female fertility in adulthood. Therefore, this study was performed to determine whether gestational or neonatal exposure to the anti-androgen flutamide influences androgen-dependent AQP5 expression in pre-antral and large antral follicles of adult pigs. Flutamide was injected into pregnant gilts between days 80 and 88 of gestation and into female piglets between days 2 and 10 post-natally. The ovaries were collected from flutamide-treated and non-treated (control) sexually mature pigs. In pre-antral follicles, AQP5 mRNA and protein levels were both downregulated following maternal (p < 0.01 and p < 0.01, respectively) and neonatal (p < 0.01 and p < 0.01, respectively) flutamide exposure. Likewise, the expression of mRNA (p < 0.01 and p < 0.001, respectively) and protein (p < 0.05 and p < 0.01, respectively) for AQP5 were diminished in large antral follicles in both groups. Immunohistochemistry showed decreased intensity of AQP5 immunoreaction in pre-antral (p < 0.01) and large antral (p < 0.001) follicles following flutamide treatment. Moreover, radioimmunological analysis revealed that changes observed in AQP5 expression corresponded with diminished follicular androgens production after both maternal (p < 0.05 and p < 0.05, respectively) and neonatal (p < 0.05 and p < 0.01, respectively) flutamide administration. Therefore, AQP5 appears to be a potential regulator of follicular fluid accumulation, under androgen control, and may be a key factor in antral follicle growth.
Collapse
Affiliation(s)
- M Grzesiak
- Department of Endocrinology, Institute of Zoology, Jagiellonian University in Krakow, Krakow, Poland.,Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Krakow, Poland
| | - K Knapczyk-Stwora
- Department of Endocrinology, Institute of Zoology, Jagiellonian University in Krakow, Krakow, Poland
| | - M R Luck
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, UK
| | - A Mobasheri
- Department of Veterinary Preclinical Sciences, School of Veterinary Medicine, Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK
| | - M Slomczynska
- Department of Endocrinology, Institute of Zoology, Jagiellonian University in Krakow, Krakow, Poland
| |
Collapse
|
95
|
Kushnir VA, Halevy N, Barad DH, Albertini DF, Gleicher N. Relative importance of AMH and androgens changes with aging among non-obese women with polycystic ovary syndrome. J Ovarian Res 2015; 8:45. [PMID: 26156856 PMCID: PMC4496928 DOI: 10.1186/s13048-015-0175-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 07/01/2015] [Indexed: 12/05/2022] Open
Abstract
Background To assess the changes in phenotypes and endocrine profiles of women with polycystic ovary syndrome (PCOS) with advancing age. Methods In a cross-sectional study conducted at a private tertiary fertility clinical and research center we identified anonymized electronic records of 37 women who had presented with a prior diagnosis of PCOS. They were stratified as younger (<35 years) and older (≥40 years). As controls, we identified 43 women with age-specific low functional ovarian reserve and 14 young women with normal functional ovarian reserve. Endocrine profiles for each group were evaluated based on total (TT) and free testosterone (FT), anti-Müllerian hormone (AMH) and sex hormone binding globulin (SHBG). Results Patients including those with PCOS were mostly non-obese, evidenced by normal BMIs (21.6 ± 6.0) with no differences between study groups. Young PCOS patients presented with a typical pattern of significant hyperandrogenemia and elevated AMH in comparison to young women with normal functional ovarian reserve [TT 44.0 (32.9–58.7) vs. 23.9 (20.3–28.1) ng/dL, (P<0.05); and AMH 7.7 (6.2–9.1) vs. 2.5 (2.0–3.0) ng/mL, (P<0.05)]. With advancing age, hyperandrogenemia in PCOS diminished in comparison to young women with normal functional ovarian reserve, resulting in similar TT levels [28.6 (19.7–37.5) vs. 23.9 (20.3–28.1) ng/dL]. Though also declining, AMH remained significantly elevated in older PCOS women in comparison to young women with normal functional ovarian reserve [4.0 (2.7–5.2) vs. 2.5 (2.0–3.0) ng/mL, (P<0.05)]. Patients with low functional ovarian reserve demonstrated significantly lower AMH at both young and older ages compared to women with normal functional ovarian reserve (P<0.05 for both). However, among patients with low functional ovarian reserve no differences were observed at young compared to older ages in TT [17.6 (12.9–24.1) vs. 18.1 (13.6–24.1) ng/dL)] and AMH [0.4 (0.3–0.6) vs. 0.3 (0.2–0.5) ng/mL]. SHBG did not differ significantly between groups but trended opposite to testosterone. Conclusions The PCOS population predominantly consisted of non-obese phenotype at both young and advanced ages. This suggests that patients with “classical” obese PCOS phenotype rarely reach tertiary infertility care, while non-obese PCOS patients may be more resistant to lower levels of infertility treatments. PCOS patients also demonstrate more precipitous declines in testosterone then AMH with advancing age. These data support incorporation of AMH as diagnostic criterion for PCOS regardless of age, and imply that testosterone should not be relied upon in the diagnosis of PCOS in older women.
Collapse
Affiliation(s)
- Vitaly A Kushnir
- Center for Human Reproduction, 21 East 69th Street, New York, NY, 10021, USA. .,Wake Forest School of Medicine, Winston-Salem, NC, USA.
| | - Noy Halevy
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - David H Barad
- Center for Human Reproduction, 21 East 69th Street, New York, NY, 10021, USA. .,Foundation for Reproductive Medicine, New York, NY, USA.
| | - David F Albertini
- Center for Human Reproduction, 21 East 69th Street, New York, NY, 10021, USA. .,University of Kansas, Lawrence, KS, USA.
| | - Norbert Gleicher
- Center for Human Reproduction, 21 East 69th Street, New York, NY, 10021, USA. .,Foundation for Reproductive Medicine, New York, NY, USA. .,Rockefeller University, New York, NY, USA.
| |
Collapse
|
96
|
Yang M, Li J, An Y, Zhang S. Effects of androgen on immunohistochemical localization of androgen receptor and Connexin 43 in mouse ovary. Tissue Cell 2015. [PMID: 26206424 DOI: 10.1016/j.tice.2015.06.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Androgens have essential roles in the regulation of follicular development and female fertility. Androgen excess is the leading defect in polycystic ovary syndrome (PCOS) patients and involved in the ovarian dysfunction. The aim of this study was to elucidate the regarding regulatory role of androgen in the follicular development of female mouse. Immunohistochemical staining and Western blot analyses were performed to detect androgen receptor (AR) and Connexin 43 (Cx43) expression in ovaries from both control and testosterone-treated group mice. In this study, localizations of AR and Cx43 were dramatically altered in testosterone-treated mouse ovaries. In addition, AR expression was significantly increased, whereas Cx43 expression was markedly decreased after testosterone treatment. Alterations of AR and Cx43 expression by testosterone with concomitant reduction of MII oocytes. Overall, these results suggest the involvement of androgen in the regulation of AR and Cx43 localizations in mouse ovary. Alterations of AR and Cx43 expression by testosterone may affect normal folliculogenesis. Together these findings will enable us to begin understanding the important roles of AR and Cx43 actions in the regulation of follicular development, as well as providing insights into the role of AR and Cx43 actions in the androgen-associated reproductive diseases such as PCOS.
Collapse
Affiliation(s)
- Mei Yang
- Center of Reproductive Medicine and Genetics, General Hospital of Beijing Army, 5 Nanmencang, Dongcheng District, Beijing 100700, China.
| | - Jianhua Li
- Center of Reproductive Medicine and Genetics, General Hospital of Beijing Army, 5 Nanmencang, Dongcheng District, Beijing 100700, China.
| | - Yulin An
- Center of Reproductive Medicine and Genetics, General Hospital of Beijing Army, 5 Nanmencang, Dongcheng District, Beijing 100700, China.
| | - Shuiwen Zhang
- Center of Reproductive Medicine and Genetics, General Hospital of Beijing Army, 5 Nanmencang, Dongcheng District, Beijing 100700, China.
| |
Collapse
|
97
|
Shohat-Tal A, Sen A, Barad DH, Kushnir V, Gleicher N. Genetics of androgen metabolism in women with infertility and hypoandrogenism. Nat Rev Endocrinol 2015; 11:429-41. [PMID: 25942654 DOI: 10.1038/nrendo.2015.64] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hypoandrogenism in women with low functional ovarian reserve (LFOR, defined as an abnormally low number of small growing follicles) adversely affects fertility. The androgen precursor dehydroepiandrosterone (DHEA) is increasingly used to supplement treatment protocols in women with LFOR undergoing in vitro fertilization. Due to differences in androgen metabolism, however, responses to DHEA supplementation vary between patients. In addition to overall declines in steroidogenic capacity with advancing age, genetic factors, which result in altered expression or enzymatic function of key steroidogenic proteins or their upstream regulators, might further exacerbate variations in the conversion of DHEA to testosterone. In this Review, we discuss in vitro studies and animal models of polymorphisms and gene mutations that affect the conversion of DHEA to testosterone and attempt to elucidate how these variations affect female hormone profiles. We also discuss treatment options that modulate levels of testosterone by targeting the expression of steroidogenic genes. Common variants in genes encoding DHEA sulphotransferase, aromatase, steroid 5α-reductase, androgen receptor, sex-hormone binding globulin, fragile X mental retardation protein and breast cancer type 1 susceptibility protein have been implicated in androgen metabolism and, therefore, can affect levels of androgens in women. Short of screening for all potential genetic variants, hormonal assessments of patients with low testosterone levels after DHEA supplementation facilitate identification of underlying genetic defects. The genetic predisposition of patients can then be used to design individualized fertility treatments.
Collapse
Affiliation(s)
- Aya Shohat-Tal
- Center for Human Reproduction, 21 E. 69th Street, New York, NY 10021, USA
| | - Aritro Sen
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - David H Barad
- Center for Human Reproduction, 21 E. 69th Street, New York, NY 10021, USA
| | - Vitaly Kushnir
- Center for Human Reproduction, 21 E. 69th Street, New York, NY 10021, USA
| | - Norbert Gleicher
- Center for Human Reproduction, 21 E. 69th Street, New York, NY 10021, USA
| |
Collapse
|
98
|
Anesetti G, Chávez-Genaro R. Neonatal testosterone exposure induces early development of follicular cysts followed by sympathetic ovarian hyperinnervation. Reprod Fertil Dev 2015; 28:RD14460. [PMID: 25989716 DOI: 10.1071/rd14460] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 04/05/2015] [Indexed: 01/22/2023] Open
Abstract
This study analysed the temporal association between ovarian cyst development induced by neonatal androgenisation and sympathetic innervation. Neonatal rats (postnatal Days 1 to 5) were treated with testosterone or dihydrotestosterone and the effects were evaluated at postnatal Days 20, 40, 90 or 180. Ovulation rate, number of cystic follicles and density of sympathetic fibres were analysed. The effects of surgical denervation or gonadotrophin stimulation were also assessed. Rats exposed to testosterone showed no oestrous cycle activity and did not ovulate, maintaining a polycystic ovarian morphology at all ages studied. Also, a significant increase in ovarian density of noradrenergic fibres was detected at postnatal Days 90 and 180. Sympathectomy was unable to re-establish ovarian activity; however, human chorionic gonadotrophin stimulation was enough to induce ovulation. The impact of dihydrotestosterone on ovarian function was less noticeable, showing the coexistence of corpora lutea and cystic structures without changes in sympathetic innervation. Our findings suggest that a remodelling of ovarian sympathetic innervation occurs as a response to modifications in the pattern of follicular growth induced by testosterone. A role of sympathetic innervation in the maintenance of the polycystic condition is suggested.
Collapse
|
99
|
Rivera OE, Varayoud J, Rodríguez HA, Santamaría CG, Bosquiazzo VL, Osti M, Belmonte NM, Muñoz-de-Toro M, Luque EH. Neonatal exposure to xenoestrogens impairs the ovarian response to gonadotropin treatment in lambs. Reproduction 2015; 149:645-55. [PMID: 25778539 DOI: 10.1530/rep-14-0567] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 03/16/2015] [Indexed: 11/08/2022]
Abstract
Bisphenol A (BPA) and diethylstilbestrol (DES) are xenoestrogens, which have been associated with altered effects on reproduction. We hypothesized that neonatal xenoestrogen exposure affects the ovarian functionality in lambs. Thus, we evaluated the ovarian response to exogenous ovine FSH (oFSH) administered from postnatal day 30 (PND30) to PND32 in female lambs previously exposed to low doses of DES or BPA (BPA50: 50 μg/kg per day, BPA0.5: 0.5 μg/kg per day) from PND1 to PND14. We determined: i) follicular growth, ii) circulating levels of 17β-estradiol (E2), iii) steroid receptors (estrogen receptor alpha, estrogen receptor beta, and androgen receptor (AR)) and atresia, and iv) mRNA expression levels of the ovarian bone morphogenetic protein (BMPs) system (BMP6, BMP15, BMPR1B, and GDF9) and FSH receptor (FSHR). Lambs neonatally exposed to DES or BPA showed an impaired ovarian response to oFSH with a lower number of follicles ≥2 mm in diameter together with a lower number of atretic follicles and no increase in E2 serum levels in response to oFSH treatment. In addition, AR induction by oFSH was disrupted in granulosa and theca cells of lambs exposed to DES or BPA. An increase in GDF9 mRNA expression levels was observed in oFSH-primed lambs previously treated with DES or BPA50. In contrast, a decrease in BMPR1B was observed in BPA0.5-postnatally exposed lambs. The modifications in AR, GDF9, and BMPR1B may be associated with the altered ovarian function due to neonatal xenoestrogen exposure in response to an exogenous gonadotropin stimulus. These alterations may be the pathophysiological basis of subfertility syndrome in adulthood.
Collapse
Affiliation(s)
- Oscar E Rivera
- Facultad de Ciencias AgrariasUniversidad Nacional de Lomas de Zamora, Buenos Aires, ArgentinaFacultad de Bioquímica y Ciencias BiológicasInstituto de Salud y Ambiente del Litoral (ISAL), Universidad Nacional del Litoral, Casilla de Correo 242, 3000 Santa Fe, Argentina
| | - Jorgelina Varayoud
- Facultad de Ciencias AgrariasUniversidad Nacional de Lomas de Zamora, Buenos Aires, ArgentinaFacultad de Bioquímica y Ciencias BiológicasInstituto de Salud y Ambiente del Litoral (ISAL), Universidad Nacional del Litoral, Casilla de Correo 242, 3000 Santa Fe, Argentina
| | - Horacio A Rodríguez
- Facultad de Ciencias AgrariasUniversidad Nacional de Lomas de Zamora, Buenos Aires, ArgentinaFacultad de Bioquímica y Ciencias BiológicasInstituto de Salud y Ambiente del Litoral (ISAL), Universidad Nacional del Litoral, Casilla de Correo 242, 3000 Santa Fe, Argentina
| | - Clarisa G Santamaría
- Facultad de Ciencias AgrariasUniversidad Nacional de Lomas de Zamora, Buenos Aires, ArgentinaFacultad de Bioquímica y Ciencias BiológicasInstituto de Salud y Ambiente del Litoral (ISAL), Universidad Nacional del Litoral, Casilla de Correo 242, 3000 Santa Fe, Argentina
| | - Verónica L Bosquiazzo
- Facultad de Ciencias AgrariasUniversidad Nacional de Lomas de Zamora, Buenos Aires, ArgentinaFacultad de Bioquímica y Ciencias BiológicasInstituto de Salud y Ambiente del Litoral (ISAL), Universidad Nacional del Litoral, Casilla de Correo 242, 3000 Santa Fe, Argentina
| | - Mario Osti
- Facultad de Ciencias AgrariasUniversidad Nacional de Lomas de Zamora, Buenos Aires, ArgentinaFacultad de Bioquímica y Ciencias BiológicasInstituto de Salud y Ambiente del Litoral (ISAL), Universidad Nacional del Litoral, Casilla de Correo 242, 3000 Santa Fe, Argentina
| | - Norberto M Belmonte
- Facultad de Ciencias AgrariasUniversidad Nacional de Lomas de Zamora, Buenos Aires, ArgentinaFacultad de Bioquímica y Ciencias BiológicasInstituto de Salud y Ambiente del Litoral (ISAL), Universidad Nacional del Litoral, Casilla de Correo 242, 3000 Santa Fe, Argentina
| | - Mónica Muñoz-de-Toro
- Facultad de Ciencias AgrariasUniversidad Nacional de Lomas de Zamora, Buenos Aires, ArgentinaFacultad de Bioquímica y Ciencias BiológicasInstituto de Salud y Ambiente del Litoral (ISAL), Universidad Nacional del Litoral, Casilla de Correo 242, 3000 Santa Fe, Argentina
| | - Enrique H Luque
- Facultad de Ciencias AgrariasUniversidad Nacional de Lomas de Zamora, Buenos Aires, ArgentinaFacultad de Bioquímica y Ciencias BiológicasInstituto de Salud y Ambiente del Litoral (ISAL), Universidad Nacional del Litoral, Casilla de Correo 242, 3000 Santa Fe, Argentina
| |
Collapse
|