1
|
Follicular Atresia, Cell Proliferation, and Anti-Mullerian Hormone in Two Neotropical Primates (Aotus nancymae and Sapajus macrocephalus). Animals (Basel) 2023; 13:ani13061051. [PMID: 36978591 PMCID: PMC10044352 DOI: 10.3390/ani13061051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/29/2022] [Accepted: 02/07/2023] [Indexed: 03/17/2023] Open
Abstract
This study evaluated the follicular atresia, cell proliferation, and anti-Mullerian hormone action in Aotus nancymae and Sapajus macrocephalus during three sexual phases (follicular, luteal, and gestational). Follicular quantification and immunolocalization of Caspase-3 protein, B-cell lymphoma 2 (BCL-2), proliferating cell nuclear antigen (PCNA), and anti-Mullerian hormone (AMH) were performed. A significant difference in the quantification between preantral and antral follicles, with a progressive decrease in the antrals, was identified. Protein and hormonal markers varied significantly between follicle cell types (A. nancymae p = 0.001; S. macrocephalus, p = 0.002). Immunostaining in the preantral and antral follicles was present in all sexual phases; for Caspase-3, in granulosa cells, oocytes, and stroma; for BCL-2, in granulosa cells, oocytes, and theca; and for PCNA and AMH, in oocytes and granulosa cells. The immunostaining for Caspase-3 was more expressive in the preantral follicles (follicular phase, p < 0.05), while that for BCL-2 and PCNA was more expressive in the antral follicles of the follicular phase. The AMH was more expressive in the primary and antral follicles of nonpregnant females, in both the follicular and luteal phases. Our results contribute to understanding the ovarian follicular selection, recruitment, and degeneration of these species.
Collapse
|
2
|
Performance of Marmoset Monkeys as Embryo Donors Is Reflected by Different Stress-Related Parameters. Animals (Basel) 2022; 12:ani12182414. [PMID: 36139275 PMCID: PMC9494952 DOI: 10.3390/ani12182414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/28/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Non-human primates (NHPs) serve as embryo donors for embryo collection in order to mimic genetic diseases in humans by genetic modification. Reproductive health of the embryo donors is crucial, and chronic distress needs to be avoided. Embryo retrieval rates (ERR), anti-Müllerian hormone (AMH) concentrations, cortisol levels, and body weight fluctuations were assessed as markers for fertility and distress. With regard to successful embryo retrievals (total n = 667), the animals were either used for extended periods (long-term group; LTG) or only for short periods (short-term group; STG). Retrospective evaluation expectedly showed that animals in the LTG had a higher ERR than animals in the STG (p < 0.0001). Importantly, ERR in the LTG remained stable throughout the experimental period, and high embryo rates were already encountered during the first year of experimental use (p = 0.0002). High ERR were associated with high AMH and low cortisol levels, and minimal body weight fluctuations following anesthesia, indicating a superior ability of the LTG animals to handle distress. We conclude that the long-term experimental use of marmosets does not impair their fertility or health status per se, supporting the view that animal reuse can be in accordance with the 3R-principle, implying reduction, replacement, and refinement in animal experimentation.
Collapse
|
3
|
di Clemente N, Racine C, Pierre A, Taieb J. Anti-Müllerian Hormone in Female Reproduction. Endocr Rev 2021; 42:753-782. [PMID: 33851994 DOI: 10.1210/endrev/bnab012] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Indexed: 12/26/2022]
Abstract
Anti-Müllerian hormone (AMH), also called Müllerian inhibiting substance, was shown to be synthesized by the ovary in the 1980s. This article reviews the main findings of the past 20 years on the regulation of the expression of AMH and its specific receptor AMHR2 by granulosa cells, the mechanism of action of AMH, the different roles it plays in the reproductive organs, its clinical utility, and its involvement in the principal pathological conditions affecting women. The findings in respect of regulation tell us that AMH and AMHR2 expression is mainly regulated by bone morphogenetic proteins, gonadotropins, and estrogens. It has now been established that AMH regulates the different steps of folliculogenesis and that it has neuroendocrine effects. On the other hand, the importance of serum AMH as a reliable marker of ovarian reserve and as a useful tool in the prediction of the polycystic ovary syndrome (PCOS) and primary ovarian failure has also been acknowledged. Last but not least, a large body of evidence points to the involvement of AMH in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Nathalie di Clemente
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France
| | - Chrystèle Racine
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine (CRSA), Paris, France.,Institut Hospitalo-Universitaire ICAN, Paris, France.,Sorbonne Paris Cité, Paris-Diderot Université, Paris, France
| | - Alice Pierre
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| | - Joëlle Taieb
- Sorbonne Paris Cité, Université Paris-Diderot, CNRS, INSERM, Biologie Fonctionnelle et Adaptative UMR 8251, Physiologie de l'Axe Gonadotrope U1133, Paris, France
| |
Collapse
|
4
|
Madrazo I, Vélez MF, Hidalgo JJ, Ortiz G, Suárez JJ, Porchia LM, Gonzalez-Mejia ME, López-Bayghen E. Prediction of severe ovarian hyperstimulation syndrome in women undergoing in vitro fertilization using estradiol levels, collected ova, and number of follicles. J Int Med Res 2021; 48:300060520945551. [PMID: 32790579 PMCID: PMC7427026 DOI: 10.1177/0300060520945551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Objective Our objective was to determine whether estradiol (E2) levels (Day 3 and fold change to Day 10), antral follicle count (AFC), and number of ova collected could predict ovarian hyperstimulation syndrome (OHSS) and culdocentesis intervention. Methods We conducted a retrospective review of patient charts between January 2008 and December 2017. OHSS was defined using American Society for Reproductive Medicine criteria. Predictability was evaluated by measuring the area under the receiver operating characteristic curve (AUC). Results The cohort included 319 women (166 controls, 153 OHSS, of whom 54 had severe OHSS). The OHSS group had higher E2Day 3 (249 ± 177 vs. 150 ± 230 ng/L), E2FoldChange (32.2 ± 29.1 vs. 20.1 ± 23.8), AFC (18.2 ± 9.1 vs. 11.6 ± 8.3), and number of ova collected (21.1 ± 9.0 vs. 10.1 ± 6.5). E2Day 3 (AUC = 0.76, 95%CI: 0.71–0.82), E2FoldChange (AUC = 0.71, 95%CI: 0.65–0.77), AFC (AUC = 0.75, 95%CI: 0.70–0.81), and number of ova collected (AUC = 0.85, 95%CI: 0.81–0.89) were predictive for OHSS. All variables were predictive for culdocentesis intervention (E2Day 3: AUC = 0.63, 95%CI: 0.55–0.70; E2FoldChange: AUC = 0.63, 95%CI: 0.55–0.71; AFC: AUC = 0.74, 95%CI: 0.68–0.80; number of ova collected: AUC = 0.80, 95%CI: 0.75–0.85). Conclusions Day 3 E2 levels and number of ova collected predict patients who could develop OHSS and may require culdocentesis.
Collapse
Affiliation(s)
- Ivan Madrazo
- Instituto de Infertilidad y Genética, Ingenes, México City, México
| | | | | | - Ginna Ortiz
- Instituto de Infertilidad y Genética, Ingenes, México City, México
| | - Juan José Suárez
- Instituto de Infertilidad y Genética, Ingenes, México City, México
| | - Leonardo M Porchia
- Departamento de Toxicología, Centro de Investigación de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| | - M Elba Gonzalez-Mejia
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, Puebla, Puebla, México
| | - Esther López-Bayghen
- Departamento de Toxicología, Centro de Investigación de Estudios Avanzados del Instituto Politécnico Nacional, México City, México
| |
Collapse
|
5
|
Xu F, Lawson MS, Campbell SP, Tkachenko OY, Park BS, Bishop CV, Xu J. Stage-dependent actions of antimüllerian hormone in regulating granulosa cell proliferation and follicular function in the primate ovary. F&S SCIENCE 2020; 1:161-171. [PMID: 34355206 PMCID: PMC8329754 DOI: 10.1016/j.xfss.2020.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To study the direct action and physiological role of antimüllerian hormone (AMH) in regulating ovarian follicular development and function in vivo in primates. DESIGN Animals were assigned to six treatment sequences in a crossover design study. Intraovarian infusion was performed during the follicular phase of the menstrual cycle with agents including: control vehicle; recombinant human AMH (rhAMH); and neutralizing anti-human AMH antibody (AMHAb). Before ovariectomy after the final treatment, the animals received intravenous injections of bromodeoxyuridine (BrdU). SETTING National primate research center. ANIMALS Adult female rhesus macaques (Macaca mulatta). INTERVENTIONS None. MAIN OUTCOME MEASURES Cycle length, follicle cohorts, and serum steroid levels were assessed. Ovarian histology, as well as granulosa cell (GC) proliferation and oocyte viability, were evaluated. RESULTS In vehicle-infused ovaries, a dominant follicle was observed at midcycle E2 peak. However, rhAMH-treated ovaries exhibited an increased number of small antral follicles, whereas AMHAb-treated ovaries developed multiple large antral follicles. Serum E2 levels in the follicular phase decreased after rhAMH infusion and increased after AMHAb infusion. The rhAMH infusion increased serum T levels. Whereas early-growing follicles of rhAMH-treated ovaries contained BrdU-positive GCs, antral follicles containing BrdU-positive GCs were identified in AMHAb-treated ovaries. Autophagy was observed in oocytes of early-growing and antral follicles exposed to AMHAb and rhAMH, respectively. CONCLUSIONS AMH enhanced early-stage follicle growth, but prevented antral follicle development and function via its stage-dependent regulation of GC proliferation and oocyte viability. This study provides information relevant to the pathophysiology of ovarian dysfunction and the treatment of infertility.
Collapse
Affiliation(s)
- Fuhua Xu
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland
| | - Maralee S. Lawson
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton
| | - Shawn P. Campbell
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland
| | - Olena Y. Tkachenko
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton
| | - Byung S. Park
- OHSU-PSU School of Public Health, Oregon Health & Science University, Portland
| | - Cecily V. Bishop
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton
- Department of Animal and Rangeland Sciences, College of Agriculture, Oregon State University, Corvallis, Oregon
| | - Jing Xu
- Division of Reproductive Endocrinology, Department of Obstetrics and Gynecology, School of Medicine, Oregon Health & Science University, Portland
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton
| |
Collapse
|
6
|
Grootenhuis A, van den Hoogen A, Broekmans F, Torrance H, van Os-Medendorp H, Ockhuijsen H. Young women's opinions on the use of a blood test to predict the possibility of premature ovarian failure: a qualitative study. HUM FERTIL 2019; 24:304-314. [PMID: 31530237 DOI: 10.1080/14647273.2019.1665198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The anti-Müllerian hormone (AMH) test is increasingly being used to predict the age when women will enter menopause, signalling the end of their reproductive life. However, the accuracy of the AMH test varies widely. In this qualitative study, we asked young women of three differing education levels about the possibility of predicting premature ovarian failure (POF), defined as menopause that occurs before the age of 40. A total of 15 face-to-face semi-structured interviews were conducted. The overarching theme was 'considering the possibility of predicting POF', and the three interpretive themes were (i) limited fertility knowledge, (ii) preconditions for pregnancy and (iii) desire to have children. The interview was their first awareness of the concept of POF. All the women who participated in this study wished to have children at some point later in life and tended to think more about fulfilling preconditions before getting pregnant than about their actual fertility. Most participants (n = 8) were interested in the AMH test regardless of their education level. Five participants wanted to take the test in the future. This research provides some insight into young women's current thoughts about the possibility of predicting POF with an AMH test.
Collapse
Affiliation(s)
- Anouk Grootenhuis
- Clinical Health Sciences at the University Utrecht, Utrecht, The Netherlands
| | - Agnes van den Hoogen
- Department Women and Baby, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Frank Broekmans
- Department Women and Baby, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Helen Torrance
- Department Women and Baby, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | - Henrietta Ockhuijsen
- Department Women and Baby, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
7
|
Treatment of female rhesus macaques with a somatostatin receptor antagonist that increases oocyte fertilization rates without affecting post-fertilization development outcomes. J Assist Reprod Genet 2018; 36:229-239. [PMID: 30430314 DOI: 10.1007/s10815-018-1369-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/05/2018] [Indexed: 10/27/2022] Open
Abstract
PURPOSE To determine the effects of PGL1001, a somatostatin receptor isoform-2 (SSTR-2) antagonist, on ovarian follicle development, oocyte fertilization, and subsequent embryo developmental potential in the rhesus macaque. METHODS Cycling female rhesus macaques (N = 8) received vehicle through one menstrual (control) cycle, followed by daily injections of PGL1001, a SSTR-2 antagonist, for three menstrual (treatment) cycles. Main endpoints include overall animal health and ovarian hormones (e.g., estradiol [E2], progesterone [P4], and anti-Müllerian hormone [AMH]), ovarian circumference, numbers of oocytes and their maturation status following controlled ovarian stimulation (COS), as well as oocyte fertilization and subsequent blastocyst rates that were assessed in control and PGL1001 treatment cycles. Circulating PGL1001 levels were assessed at baseline as well as 6, 60, and 90 days during treatment. RESULTS PGL1001 treatment did not impact overall animal health, menstrual cycle length, or circulating levels of ovarian hormones (E2, P4, and AMH) in comparison to vehicle treatment during natural cycles. PGL1001 treatment increased (p ˂ 0.05) ovarian circumference and the day 8 to day 1 ratio of AMH levels (p ˂ 0.05) during a COS protocol, as well as oocyte fertilization rates compared to the vehicle treatment interval. Blastocyst development rates were not significantly different between vehicle and PGL1001 treatment groups. CONCLUSION Prolonged treatment with PGL1001 appears to be safe and does not affect rhesus macaque general health, menstrual cycle length, or ovarian hormone production. Interestingly, PGL1001 treatment increased the fertilization rate of rhesus macaque oocytes collected following ovarian stimulation.
Collapse
|
8
|
Almeida FRCL, Costermans NGJ, Soede NM, Bunschoten A, Keijer J, Kemp B, Teerds KJ. Presence of anti-Müllerian hormone (AMH) during follicular development in the porcine ovary. PLoS One 2018; 13:e0197894. [PMID: 30063719 PMCID: PMC6067700 DOI: 10.1371/journal.pone.0197894] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 04/03/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Anti-Müllerian hormone (AMH) is expressed by granulosa cells of developing follicles and plays an inhibiting role in the cyclic process of follicular recruitment by determining follicle-stimulating hormone threshold levels. Knowledge of AMH expression in the porcine ovary is important to understand the reproductive efficiency in female pigs. RESEARCH AIM In the present study we investigated the expression of AMH during follicular development in prepubertal and adult female pigs by immunohistochemistry, laser capture micro-dissection and RT-qPCR. RESULTS AND CONCLUSION Although in many aspects the immunohistochemical localization of AMH in the porcine ovary does not differ from other species, there are also some striking differences. As in most species, AMH appears for the first time during porcine follicular development in the fusiform granulosa cells of recruited primordial follicles and continues to be present in granulosa cells up to the antral stage. By the time follicles reach the pre-ovulatory stage, AMH staining intensity increases significantly, and both protein and gene expression is not restricted to granulosa cells; theca cells now also express AMH. AMH continues to be expressed after ovulation in the luteal cells of the corpus luteum, a phenomenon unique to the porcine ovary. The physiological function of AMH in the corpus luteum is at present not clear. One can speculate that it may contribute to the regulation of the cyclic recruitment of small antral follicles. By avoiding premature exhaustion of the ovarian follicular reserve, AMH may contribute to optimization of reproductive performance in female pigs.
Collapse
Affiliation(s)
- Fernanda R. C. L. Almeida
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Antônio Carlos, Belo Horizonte, Minas Gerais, Brazil
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | - Natasja G. J. Costermans
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
- Human and Animal Physiology, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | - Nicoline M. Soede
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | - Annelies Bunschoten
- Human and Animal Physiology, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | - Jaap Keijer
- Human and Animal Physiology, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | - Bas Kemp
- Adaptation Physiology Group, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
| | - Katja J. Teerds
- Human and Animal Physiology, Department of Animal Sciences, Wageningen University, Wageningen, Netherlands
- * E-mail:
| |
Collapse
|
9
|
Rajabi Z, Khokhar Z, Yazdekhasti H. The Growth of Preantral Follicles and the Impact of Different Supplementations and Circumstances: A Review Study with Focus on Bovine and Human Preantral Follicles. Cell Reprogram 2018; 20:164-177. [PMID: 29782184 DOI: 10.1089/cell.2017.0068] [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: 12/31/2022] Open
Abstract
One of the most important concerns cancer survivors face is fertility. Current treatment modalities often result in damage to the reproductive system. Different options have been proposed to preserve the fertility of affected women, and many attempts have been made to improve their chance of childbearing after therapy. Cryopreservation of ovarian tissue and follicles before the onset of cancer treatment and then either transplantation of ovarian tissue or culture of ovarian tissue and individual follicles in vitro is a commonly cited approach. Extensive research is being done to design an optimal condition for the culture of ovarian follicles. Improving follicle culture systems by understanding their actual growth needs might be a crucial step toward fertility preservation in cancer patients. This review article will try to provide a summary of the role of different factors and conditions on growth of human and bovine preantral follicles in vitro.
Collapse
Affiliation(s)
- Zahra Rajabi
- 1 Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences , Tehran, Iran .,2 Department of Biomedical Engineering, University of Virginia , Charlottesville, Virginia
| | - Zunair Khokhar
- 3 Department of Cell Biology, University of Virginia , Charlottesville, Virginia
| | - Hossein Yazdekhasti
- 4 Center for Research in Contraception and Reproductive Health, University of Virginia , Charlottesville, Virginia.,5 Center for Membrane & Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia , Charlottesville, Virginia
| |
Collapse
|
10
|
Kim YY, Tamadon A, Ku SY. Potential Use of Antiapoptotic Proteins and Noncoding RNAs for EfficientIn VitroFollicular Maturation and Ovarian Bioengineering. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:142-158. [PMID: 27763207 DOI: 10.1089/ten.teb.2016.0156] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea
| | - Amin Tamadon
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
11
|
Anti-Müllerian hormone is produced heterogeneously in primate preantral follicles and is a potential biomarker for follicle growth and oocyte maturation in vitro. J Assist Reprod Genet 2016; 33:1665-1675. [PMID: 27638727 DOI: 10.1007/s10815-016-0804-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/23/2016] [Indexed: 10/21/2022] Open
Abstract
PURPOSE The main goals of this study were to investigate the expression of anti-Müllerian hormone (AMH) and its receptor (AMHR2) during follicular development in primates, and to evaluate the potential of AMH as a biomarker for follicle growth and oocyte maturation in vitro. METHODS The mRNA and protein expression of AMH and AMHR2 were determined using isolated follicles and ovarian sections from rhesus macaques (n = 4) by real-time PCR and immunohistochemistry, respectively. Isolated secondary follicles were cultured individually. Follicle growth and media AMH concentrations were assessed by ELISA. The mRNA expression profiles, obtained from RNA sequencing, of in vitro- and in vivo-developed antral follicles were compared. Secondary follicles from additional animals (n = 35) were cultured. Follicle growth, oocyte maturation, and media AMH concentrations were evaluated for forecasting follicular development in vitro by AMH levels. RESULTS AMH immunostaining was heterogeneous in the population of preantral follicles that were also stained for AMHR2. The mRNA expression profiles were comparable between in vivo- and in vitro-developed follicles. AMH levels produced by growing follicles were higher than those of nongrowing follicles in culture. With a cutoff value of 1.40 ng/ml, 85 % of nongrowing follicles could be identified while eliminating only 5 % of growing follicles. Growing follicles that generated metaphase II-stage oocytes secreted greater amounts of AMH than did those yielding immature germinal vesicle-stage oocytes. CONCLUSIONS AMH, co-expressed with AMHR2, was produced heterogeneously by preantral follicles in macaques with levels correlated positively with follicle growth and oocyte maturation. AMH may serve as a biomarker for primate follicular development in vitro.
Collapse
|
12
|
Dewailly D, Robin G, Peigne M, Decanter C, Pigny P, Catteau-Jonard S. Interactions between androgens, FSH, anti-Müllerian hormone and estradiol during folliculogenesis in the human normal and polycystic ovary. Hum Reprod Update 2016; 22:709-724. [PMID: 27566840 DOI: 10.1093/humupd/dmw027] [Citation(s) in RCA: 312] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 07/12/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Androgens, FSH, anti-Müllerian hormone (AMH) and estradiol (E2) are essential in human ovarian folliculogenesis. However, the interactions between these four players is not fully understood. OBJECTIVES AND RATIONALE The purpose of this review is to highlight the chronological sequence of the appearance and function of androgens, FSH, AMH and E2 and to discuss controversies in the relationship between FSH and AMH. A better understanding of this interaction could supplement our current knowledge about the pathophysiology of the polycystic ovary syndrome (PCOS). SEARCH METHODS A literature review was performed using the following search terms: androgens, FSH, FSH receptor, anti-Mullerian hormone, AMHRII, estradiol, follicle, ovary, PCOS, aromatase, granulosa cell, oocyte. The time period searched was 1980-2015 and the databases interrogated were PubMed and Web of Science. OUTCOMES During the pre-antral ('gonadotropin-independent') follicle growth, FSH is already active and promotes follicle growth in synergy with theca cell-derived androgens. Conversely, AMH is inhibitory by counteracting FSH. We challenge the hypothesis that AMH is regulated by androgens and propose rather an indirect effect through an androgen-dependent amplification of FSH action on granulosa cells (GCs) from small growing follicles. This hypothesis implies that FSH stimulates AMH expression. During the antral ('gonadotropin-dependent') follicle growth, E2 production results from FSH-dependent activation of aromatase. Conversely, AMH is inhibitory but the decline of its expression, amplified by E2, allows full expression of aromatase, characteristic of the large antral follicles. We propose a theoretical scheme made up of two triangles that follow each other chronologically. In PCOS, pre-antral follicle growth is excessive (triangle 1) because of intrinsic androgen excess that renders GCs hypersensitive to FSH, with consequently excessive AMH expression. Antral follicle growth and differentiation are disturbed (triangle 2) because of the abnormally persisting inhibition of FSH effects by AMH that blocks aromatase. Beside anovulation, this scenario may also serve to explain the higher receptiveness to gonadotropin therapy and the increased risk of ovarian hyperstimulation syndrome (OHSS) in patients with PCOS. WIDER IMPLICATIONS Within GCs, the balance between FSH and AMH effects is pivotal in the shift from androgen- to oestrogen-driven follicles. Our two triangles hypothesis, based on updated data from the literature, offers a pedagogic template for the understanding of folliculogenesis in the normal and polycystic ovary. It opens new avenues for the treatment of anovulation due to PCOS.
Collapse
Affiliation(s)
- Didier Dewailly
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France .,Faculté de Médecine, Université Lille Nord de France, 59000 Lille, France
| | - Geoffroy Robin
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France
| | - Maëliss Peigne
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France
| | - Christine Decanter
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France
| | - Pascal Pigny
- Faculté de Médecine, Université Lille Nord de France, 59000 Lille, France.,CHU Lille, Laboratoire de Biochimie & Hormonologie, Centre de Biologie Pathologie, F-59037 Lille, France
| | - Sophie Catteau-Jonard
- CHU Lille, Service de Gynécologie Endocrinienne et Médecine de la Reproduction, Hôpital Jeanne de Flandre, F-59037, Lille, France.,Faculté de Médecine, Université Lille Nord de France, 59000 Lille, France
| |
Collapse
|
13
|
Visser JA. Shaping up the function of anti-Müllerian hormone in ovaries of mono-ovulatory species. Hum Reprod 2016; 31:1403-5. [PMID: 27165619 DOI: 10.1093/humrep/dew101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 04/01/2016] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jenny A Visser
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| |
Collapse
|
14
|
Xu J, Bishop CV, Lawson MS, Park BS, Xu F. Anti-Müllerian hormone promotes pre-antral follicle growth, but inhibits antral follicle maturation and dominant follicle selection in primates. Hum Reprod 2016; 31:1522-30. [PMID: 27165618 DOI: 10.1093/humrep/dew100] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/18/2016] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION What are the direct effects and physiological role of anti-Müllerian hormone (AMH) during primate follicular development and function at specific stages of folliculogenesis? SUMMARY ANSWER AMH actions in the primate ovary may be stage-dependent, directly promoting pre-antral follicle growth while inhibiting antral follicle maturation and dominant follicle selection. WHAT IS KNOWN ALREADY AMH is expressed in the adult ovary, particularly in developing follicles. Studies in mice suggest that AMH suppresses pre-antral follicle growth in vitro, and inhibits primordial follicle recruitment and FSH-stimulated antral follicle steroidogenesis. STUDY DESIGN, SIZE, DURATION For in vitro study, secondary follicles were isolated from ovaries of 12 rhesus macaques and cultured for 5 weeks. For in vivo study, intraovarian infusion was conducted on five monkeys for the entire follicular phase during two spontaneous menstrual cycles. PARTICIPANTS/MATERIALS, SETTING, METHODS For in vitro study, individual follicles were cultured in a 5% O2 environment, in alpha minimum essential medium supplemented with recombinant human FSH. Follicles were randomly assigned to treatments of recombinant human AMH protein or neutralizing anti-human AMH antibody (AMH-Ab). Follicle survival, growth, steroid production, steroidogenic enzyme expression, and oocyte maturation were assessed. For in vivo study, ovaries were infused with control vehicle or AMH-Ab during the follicular phase of the menstrual cycle. Cycle length, serum steroid levels, and antral follicle growth were evaluated. MAIN RESULTS AND THE ROLE OF CHANCE AMH exposure during culture weeks 0-3 (pre-antral stage) promoted, while AMH-Ab delayed, antrum formation of growing follicles compared with controls. AMH treatment during culture weeks 3-5 (antral stage) decreased (P < 0.05) estradiol (E2) production, as well as the mRNA expression of cytochrome P450 family 19 subfamily A polypeptide 1, by antral follicles relative to controls, whereas AMH-Ab increased (P < 0.05) follicular mRNA levels of the enzyme. Intraovarian infusion of AMH-Ab during the follicular phase of the menstrual cycle increased (P < 0.05) the average levels of serum E2 compared with those of the control cycles. Three of the five AMH-Ab-treated ovaries displayed multiple (n = 2-9) medium-to-large (2-8 mm) antral follicles at the mid-cycle E2 peak, whereas only one large (4-7 mm) antral follicle was observed in all monkeys during their control cycles. The average levels of serum progesterone were higher (P < 0.05) during the luteal phase of cycles following the AMH-Ab infusion relative to the vehicle infusion. LIMITATIONS, REASONS FOR CAUTION The in vitro study of AMH actions on cultured individual macaque follicles was limited to the interval from the secondary to small antral stage. A sequential study design was used for in vivo experiments, which may limit the power of the study. WIDER IMPLICATIONS OF THE FINDINGS The current study provides novel information on direct actions and role of AMH during primate follicular development, and selection of a dominant follicle by the late follicular phase of the menstrual cycle. We hypothesize that AMH acts positively on follicular growth during the pre-antral stage in primates, but negatively impacts antral follicle maturation, which is different from what is reported in the mouse model. STUDY FUNDING/COMPETING INTERESTS NIH NICHD R01HD082208, NIH ORWH/NICHD K12HD043488 (BIRCWH), NIH OD P51OD011092 (ONPRC), Collins Medical Trust. There are no conflicts of interest. TRIAL REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- J Xu
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - C V Bishop
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - M S Lawson
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - B S Park
- OHSU-PSU School of Public Health, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - F Xu
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| |
Collapse
|
15
|
FANG YANQIU, LU XIAODAN, LIU LEI, LIN XIUYING, SUN MUNAN, FU JIANHUA, XU SHUFEN, TAN YAN. Vascular endothelial growth factor induces anti-Müllerian hormone receptor 2 overexpression in ovarian granulosa cells of in vitro fertilization/intracytoplasmic sperm injection patients. Mol Med Rep 2016; 13:5157-62. [DOI: 10.3892/mmr.2016.5173] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 03/08/2016] [Indexed: 11/06/2022] Open
|
16
|
Morphometric characteristics of preantral and antral follicles and expression of factors involved in folliculogenesis in ovaries of adult baboons (Papio anubis). J Assist Reprod Genet 2016; 33:617-626. [PMID: 26945754 DOI: 10.1007/s10815-016-0681-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 02/22/2016] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Baboons are commonly utilized as an animal model for studies of human reproduction. However, folliculogenesis in this species has not been fully documented. The aim of this study was to assess follicle morphometry and expression of essential proteins involved in folliculogenesis in baboons. METHODS Ovaries were recovered from four adult baboons and processed for histological evaluation and immunohistochemical analyses. Follicle proportion, follicle and oocyte diameter, theca layer thickness, number of granulosa cells, and follicle density were calculated. Immunohistochemical staining was also carried out for connexin 43 (Cx43), aromatase, and zona pellucida 3 (ZP3). RESULTS A total of 2221 follicles were counted and measured. Proportions of primordial, primary, secondary, small antral, and large antral follicles were 49, 26, 23, 1, and 1 %, respectively. The increase in follicle diameter was due not only to the increase in oocyte diameter but also to granulosa cell proliferation. Almost all antral follicles were positive for Cx43 (89.8 %), aromatase (84.8 %), and ZP3 (100 %). Most secondary follicles were positive for Cx43 (65 %) and ZP3 (64.5 %), and some primary follicles were positive only for Cx43. No primordial follicles stained positive in any of these immunohistochemical analyses. Only antral follicles showed aromatase activity. CONCLUSIONS On the basis of these results, we can conclude that folliculogenesis in baboons appears to be similar to that in humans, and this animal therefore constitutes a valuable model.
Collapse
|
17
|
HUANG JILIANG, WANG XIAOYAN, LI ZHILING, MA RUOWU, XIAO WANFEN. Effects of GnRH agonists on the expression of developmental follicular anti-mullerian hormone in varying follicular stages in cyclic mice in vivo. Mol Med Rep 2015; 12:4305-4313. [PMID: 26126720 PMCID: PMC4526049 DOI: 10.3892/mmr.2015.3993] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 06/05/2015] [Indexed: 02/05/2023] Open
Abstract
Gonadotrophin‑releasing hormone (GnRH) agonists (GnRHa) have been widely used to induce a state of downregulation for in vitro fertilization, and its direct effects on the pituitary are well known. However, the effects of GnRHa on the expression of anti‑mullerian hormone (AMH) by follicles in varying stages in vivo remain to be fully elucidated. In the present study 84 cyclic mice were randomly divided equally into four GnRHa groups and three cyclic mice were used as a control group. The expression levels of AMH in follicles of varying stages between days 0 and 7 following GnRHa administration were quantified using immunohistochemistry. The expression of AMH in follicles at various stages revealed dynamic changes during the process of downregulation. AMH in primary follicles initially increased and then decreased gradually. In small and large preantral follicles and in granulosa cells (GCs) surrounding the oocyte of small antral follicles, the expression of AMH began to increase on day 1, was attenuated on day 2, and then increased to a peak. The expression levels of AMH in the GCs surrounding the basement membrane, in contrast to the GCs surrounding the oocyte, were significantly lower and did not increase on day 1. In all stages of follicles, the expression of AMH declined gradually between the peak level and last day of downregulation. On day 7, the varying follicular stages all expressed lower levels of AMH than on day 0. This decrease was more prominent in the higher dose groups, compared with the lower dose groups. In conclusion, GnRHa was observed to induce time‑dependent changes in the expression of AMH at varying follicular stages, which occurred in a dose‑dependent manner.
Collapse
Affiliation(s)
| | | | - ZHILING LI
- Correspondence to: Professor Zhiling Li, Reproductive Center, First Affiliated Hospital of Shantou University Medical College, Shantou University, 54 Changping Road, Jinping, Shantou, Guangdong 515041, P.R. China, E-mail:
| | | | | |
Collapse
|
18
|
Ting AY, Xu J, Stouffer RL. Differential effects of estrogen and progesterone on development of primate secondary follicles in a steroid-depleted milieu in vitro. Hum Reprod 2015; 30:1907-17. [PMID: 26040480 PMCID: PMC4507328 DOI: 10.1093/humrep/dev119] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 03/19/2015] [Accepted: 05/01/2015] [Indexed: 12/22/2022] Open
Abstract
STUDY QUESTION What are the direct effects of progesterone (P4) and estradiol (E2) on the development and function of primate follicles in vitro from the pre-antral to early antral stage? SUMMARY ANSWER In a steroid-depleted milieu, E2 improved follicle survival, growth, antrum formation and oocyte health, whereas P4 exerted minimal beneficial effects on follicle survival and reduced oocyte health. WHAT IS KNOWN ALREADY Effects of P4 and E2 on follicle development have been studied primarily in large antral and pre-ovulatory follicles. Chronic P4 exposure suppresses antral follicle growth, but acute P4 exposure promotes oocyte maturation in pre-ovulatory follicles. Effects of E2 can be stimulatory or inhibitory depending upon species, dose and duration of exposure. STUDY DESIGN, SIZE, DURATION Non-human primate model, randomized, control versus treatment. Macaque (n = 6) secondary follicles (n = 24 per animal per treatment group) were cultured for 5 weeks. PARTICIPANTS/MATERIALS, SETTING, METHODS Adult rhesus macaque secondary follicles were encapsulated in 0.25% alginate and cultured individually in media containing follicle stimulating hormone plus (i) vehicle, (ii) a steroid-synthesis inhibitor, trilostane (TRL, 250 ng/ml), (iii) TRL + low E2 (100 pg/ml) or progestin (P, 10 ng/ml R5020) and (iv) TRL + high E2 (1 ng/ml E2) or P (100 ng/ml R5020). Follicles reaching the antral stage (≥750 µm) were treated with human chorionic gonadotrophin for 34 h. End-points included follicle survival, antrum formation, growth pattern, plus oocyte health and maturation status, as well as media concentrations of P4, E2 and anti-Müllerian hormone (AMH). MAIN RESULTS AND THE ROLE OF CHANCE In a steroid-depleted milieu, low dose, but not high dose, P improved (P < 0.05) follicle survival, but had no effect (P > 0.05) on antrum formation and AMH production. Low-dose P increased (P < 0.05) P4 production in fast-grow follicles, and both doses of P elevated (P < 0.05) E2 production in slow-grow follicles. Additionally, low-dose P increased (P < 0.05) the percentage of no-grow follicles, and high-dose P promoted oocyte degeneration. In contrast, E2, in a steroid-depleted milieu, improved (P < 0.05) follicle survival, growth, antrum formation and oocyte health. E2 had no effect on P4 or E2 production. Follicles exposed to E2 yielded mature oocytes capable of fertilization and early cleavage, at a rate similar to untreated control follicles. LIMITATIONS, REASONS FOR CAUTION This study is limited to in vitro effects of P and E2 during the interval from the secondary to small antral stage of macaque follicles. WIDER IMPLICATIONS OF THE FINDINGS This study provides novel information on the direct actions of P4 and E2 on primate pre-antral follicle development. Combined with our previous report on the actions of androgens, our findings suggest that androgens appear to be a survival factor but hinder antral follicle differentiation, E2 appears to be a survival and growth factor at the pre-antral and early antral stage, whereas P4 may not be essential during early folliculogenesis in primates. STUDY FUNDING/COMPETING INTERESTS NIH P50 HD071836 (NCTRI), NIH ORWH/NICHD 2K12HD043488 (BIRCWH), ONPRC 8P51OD011092. There are no conflicts of interest.
Collapse
Affiliation(s)
- A Y Ting
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - J Xu
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - R L Stouffer
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA Department of Obstetrics and Gynecology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
19
|
Hasky N, Uri-Belapolsky S, Goldberg K, Miller I, Grossman H, Stemmer SM, Ben-Aharon I, Shalgi R. Gonadotrophin-releasing hormone agonists for fertility preservation: unraveling the enigma? Hum Reprod 2015; 30:1089-101. [PMID: 25796551 DOI: 10.1093/humrep/dev037] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 02/03/2015] [Indexed: 12/27/2022] Open
Abstract
STUDY QUESTION Can gonadotrophin-releasing hormone agonists (GnRH-a) preserve long-term fertility when administered prior to and concomitantly with chemotherapy? SUMMARY ANSWER GnRH-a display a differential protective effect on fertility, depending upon the specific chemotherapy-induced mechanism of ovarian injury. WHAT IS KNOWN ALREADY The role of GnRH-a in fertility preservation has been constantly debated and their use is considered experimental due to conflicting clinical evidence and paucity of data regarding their mechanism for ovarian protection. STUDY DESIGN, SIZE, DURATION In vivo model: 7-8 weeks old imprinting control region (ICR) mice were injected with GnRH-a (Leuprolide-acetate) or saline prior to and concomitantly with cyclophosphamide, doxorubicin or saline and sacrificed at various time-points on a longitudinal follow-up; 24 h (n = 36), 1 week (n = 40), 1 month (n = 36) and 9 months (n = 66) post chemotherapy treatment. Blood samples were drawn on Day 0 and on a monthly basis after chemotherapy treatment. On the day of sacrifice, blood samples were drawn and ovaries excised and processed for either immunohistochemistry (IHC), protein or RNA extraction. In vitro model: 21-23 days old Wistar-derived rats were sacrificed, their ovaries excised and primary granulosa cells (PGC) were either isolated for in vitro culture, or processed for immunofluorescence (IF) as well as for protein or RNA extraction. MATERIALS, SETTING, METHODS Ovarian reserve was estimated by serial measurements of serum anti-mullerian hormone (AMH), quantified by the AMH Gen II ELISA assay. Ovarian AMH and phosphorylated Akt (pAkt) were detected by immunoblotting. Vascular endothelial growth factor (VEGF) was measured by quantitative PCR. Ovarian GnRH receptor (GnRHR), AMH and CD34 were visualized by IHC, and apoptosis was evaluated using TdT (terminal deoxynucleotidyl transferase)-mediated dUDP nick-end labeling (TUNEL). MAIN RESULTS AND THE ROLE OF CHANCE Cyclophosphamide-induced ovarian injury caused a prompt decrease in AMH level (P < 0.01) and a further long-term decline in serum AMH (P = 0.017), indicating damage to the ovarian reserve. Pretreatment with GnRH-a diminished AMH-decrease (P < 0.05) and maintained serum AMH level in the long run (P < 0.05). Doxorubicin-exerted ovarian-vascular-injury is also displayed by an acute increase in ovarian VEGF level (P < 0.05) and a sustained decrease in serum AMH level (P < 0.001). This was followed by ovarian recovery manifested by increased neovascularization. GnRH-a delayed the recovery in AMH level and decreased the level of VEGF (P < 0.001), thus interfering with the vascular recovery subsequent to doxorubicin-induced vascular damage. LIMITATIONS, REASONS FOR CAUTION To portray the differential mechanism of each chemotherapy, cyclophosphamide and doxorubicin were given separately, whereas most of the clinical protocols include several types of chemotherapies. Thus, future study should explore a prospective evaluation of various chemotherapies, as well as combined chemotherapeutic protocols. WIDER IMPLICATIONS OF THE FINDINGS Our study demonstrates that different chemotherapy agents affect the ovary via diverse mechanisms and thus the administration of GnRH-a concomitantly, could be beneficial to a subpopulation of patients treated with cyclophosphamide-based protocols. STUDY FUNDING/COMPETING INTERESTS This work was partially supported by a grant from the Israel Science Foundation (ISF) to I.B.-A. The authors have no conflict of interest to disclose.
Collapse
Affiliation(s)
- Noa Hasky
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shiri Uri-Belapolsky
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Keren Goldberg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Irit Miller
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hadas Grossman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Salomon M Stemmer
- Institute of Oncology, Davidoff Center and Rabin Medical Center, Petah-Tiqva and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Irit Ben-Aharon
- Institute of Oncology, Davidoff Center and Rabin Medical Center, Petah-Tiqva and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ruth Shalgi
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
20
|
Scalercio SRRA, Brito AB, Domingues SFS, Santos RR, Amorim CA. Immunolocalization of growth, inhibitory, and proliferative factors involved in initial ovarian folliculogenesis from adult common squirrel monkey (Saimiri collinsi). Reprod Sci 2014; 22:68-74. [PMID: 24784715 DOI: 10.1177/1933719114532842] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We performed an immunohistochemical (IHC) study to determine the follicular expression of growth differentiation factor 9 (GDF-9), anti-Müllerian hormone (AMH), Kit Ligand (KL), and c-Kit in squirrel monkey ovary. Ovarian tissue fragments from 4 squirrel monkeys were collected by laparotomy and processed for classical histology and IHC. Additionally, follicle development was assessed by Ki67 immunostaining to evaluate proliferative status of granulosa cells. A total of 4025 follicles were examined (1475 for classical histology and 2550 for immunohistochemistry). More than 80% of the evaluated follicles were morphologically normal. The GDF-9 protein was detectable in oocyte cytoplasm from primordial (100%), primary (99.1%), and secondary (100%) follicles. The AMH was not expressed in primordial follicles but just in few primary follicles (13.8%). On the other hand, it was highly expressed in granulosa cells from secondary follicles (67.9%). c-Kit, KL receptor, was found in the oolemma of primordial (100%), primary (100%), and secondary (100%) follicles. The KL expression was observed in oocytes and granulosa cells from primordial (94.9%), primary (91.6%) and secondary follicles (100%). Ki67 immunostaining was observed in granulosa cells from primary (5.7%) and secondary (54.8%) follicles but not in primordial follicles. In conclusion, we described the localization of GDF-9, KL, c-Kit, and Ki67 proteins and confirmed the presence of AMH protein in preantral follicles from squirrel monkey. Our results offer contribution for understanding of folliculogenesis in neotropical nonhuman primates. Moreover, these markers can be used to assess follicular viability and functionality after cryopreservation, transplantation, or in vitro culture of ovarian tissue.
Collapse
Affiliation(s)
- S R R A Scalercio
- Laboratory of Wild Animal Biology and Medicine, Federal University of Pará, Castanhal, Pará, Brazil Animal Sciences PhD Program, Federal University of Pará, Belém, Pará, Brazil National Primate Centre, Secretary of Health Policy, Ministry of Health, Ananindeua, Pará, Brazil
| | - A B Brito
- Laboratory of Wild Animal Biology and Medicine, Federal University of Pará, Castanhal, Pará, Brazil Animal Sciences PhD Program, Federal University of Pará, Belém, Pará, Brazil
| | - S F S Domingues
- Laboratory of Wild Animal Biology and Medicine, Federal University of Pará, Castanhal, Pará, Brazil Animal Sciences PhD Program, Federal University of Pará, Belém, Pará, Brazil
| | - R R Santos
- Laboratory of Wild Animal Biology and Medicine, Federal University of Pará, Castanhal, Pará, Brazil Animal Sciences PhD Program, Federal University of Pará, Belém, Pará, Brazil Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - C A Amorim
- Pôle de Recherche en Gynécologie, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
21
|
Xu J, Lawson MS, Yeoman RR, Molskness TA, Ting AY, Stouffer RL, Zelinski MB. Fibrin promotes development and function of macaque primary follicles during encapsulated three-dimensional culture. Hum Reprod 2013; 28:2187-200. [PMID: 23608357 DOI: 10.1093/humrep/det093] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
STUDY QUESTION Does fibrin introduced into the extracellular matrix affect the growth and maturation of individual primate follicles during encapsulated three-dimensional (3D) culture? SUMMARY ANSWER While not altering follicle survival, fibrin-alginate (FIBRIN) improves macaque primary, but not secondary, follicle development during encapsulated 3D culture in terms of growth, steroidogenesis, anti-Müllerian hormone (AMH)/vascular endothelial growth factor (VEGF) production and oocyte maturation. WHAT IS KNOWN ALREADY Efforts to grow non-human primate ovarian follicles from the secondary to the antral stage during encapsulated 3D culture have been successful. However, the growth and maturation of primary follicles in vitro has not been reported in primates, especially in chemically defined conditions. STUDY DESIGN, SIZE, DURATION In vitro follicle maturation was investigated using the rhesus macaque (Macaca mulatta). Ovaries (n = 7 pairs) were obtained during the early follicular phase of the menstrual cycle (cycle day 1-4). Primary (80-120 µm diameter) and secondary (125-225 µm diameter) follicles were isolated mechanically, randomly assigned to experimental groups, encapsulated into alginate (0.25% w/v) or FIBRIN (25 mg/ml fibrinogen-0.25% alginate) and cultured for 13 and 5 weeks, respectively. MATERIALS, SETTING, METHODS Individual follicles were cultured in alpha minimum essential medium supplemented with FSH. Follicle survival and growth were assessed by microscopy. Follicles that reached the antral stage were treated with recombinant hCG. Metaphase II (MII) oocytes were inseminated via ICSI. Follicle morphology was evaluated by hematoxylin and eosin (H&E) staining. Immunohistochemistry was performed for cytochrome P450 family 17 subfamily A polypeptide 1 (CYP17A1) and 19 subfamily A polypeptide 1 (CYP19A1). Culture medium was analyzed for estradiol (E2) and progesterone by chemiluminescence, androstenedione (A4) by radioimmunoassay, as well as anti-Müllerian hormone (AMH) and vascular endothelial growth factor (VEGF) by enzyme-linked immunosorbent assay. MAIN RESULTS AND THE ROLE OF CHANCE A total of 105 primary and 133 secondary follicles were collected. The presence of fibrin in the alginate matrix had no effect on either primary or secondary follicle survival. Growing primary and secondary follicles formed an antrum at Weeks 9 and 3, respectively. The percentage of growing follicles was higher (P < 0.05) for primary follicles cultured in FIBRIN than alginate at Week 13. The diameters were larger for the growing secondary follicles cultured in alginate than FIBRIN at Week 5 (P < 0.05). H&E staining revealed the typical morphology for small antral follicles. CPY17A1 immunostaining was detected in theca cells, while CYP19A1 was observed in granulosa cells. E2 increased (P < 0.05) during antrum formation in growing follicles at Week 9 for primary and Week 3 for secondary follicles. AMH levels in medium from growing primary follicles increased (P < 0.05) after Week 4 with peak levels at Weeks 9-11. AMH increased (P < 0.05) in growing secondary follicles at Weeks 3-5. VEGF levels in medium were elevated (P < 0.05) in growing primary follicles at Week 9. VEGF increased (P < 0.05) in medium from growing secondary follicles at Weeks 3-5. E2, AMH and VEGF production was higher (P < 0.05) in primary follicle culture with FIBRIN than alginate alone. One primary follicle cultured in FIBRIN (1 of 5 follicles harvested) and a secondary follicle cultured in alginate alone (1 of 15 follicles harvested) yielded an MII oocyte. The fertilized oocyte from primary follicle culture arrested without cell division after fertilization, while the oocyte from secondary follicle culture cleaved and reached the morula stage. LIMITATIONS, REASONS FOR CAUTION The study reports on in vitro development and function of individual macaque follicles, that is limited to the interval from the primary and secondary stage to the small antral stage. The findings await translation to human ovarian follicles. WIDER IMPLICATIONS OF THE FINDINGS The 3D model for primate follicle development offers a unique opportunity to investigate the growth and regulation of primate primary, as well as secondary follicles, and their enclosed oocytes, as they grow to the antral stage by monitoring and manipulating factors or signaling pathways in vitro. Since primate primary follicles, in addition to secondary follicles, can be cultured to the antral stage to provide mature oocytes, they represent an additional source of pre-antral follicles for in vitro follicle maturation with the potential to provide gametes for assisted reproductive technology as an option for fertility preservation in women, including patients with cancer. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by The Oncofertility Consortium (NIH U54 RR024347-HD058294, PL1-EB008542), NIH U54-HD18185 (Eunice Kennedy Shriver Specialized Cooperative Centers Program in Reproduction and Infertility Research), NIH ORWH/NICHD 2K12HD043488 (BIRCWH), Oregon National Primate Research Center 8P51OD011092. There are no conflicts of interest.
Collapse
Affiliation(s)
- J Xu
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, 505 NW 185th Avenue, Beaverton, OR 97006, USA.
| | | | | | | | | | | | | |
Collapse
|
22
|
Amorim CA, Jacobs S, Devireddy RV, Van Langendonckt A, Vanacker J, Jaeger J, Luyckx V, Donnez J, Dolmans MM. Successful vitrification and autografting of baboon (Papio anubis) ovarian tissue. Hum Reprod 2013; 28:2146-56. [DOI: 10.1093/humrep/det103] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
23
|
Xu J, Xu M, Bernuci MP, Fisher TE, Shea LD, Woodruff TK, Zelinski MB, Stouffer RL. Primate follicular development and oocyte maturation in vitro. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 761:43-67. [PMID: 24097381 PMCID: PMC4007769 DOI: 10.1007/978-1-4614-8214-7_5] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The factors and processes involved in primate follicular development are complex and not fully understood. An encapsulated three-dimensional (3D) follicle culture system could be a valuable in vitro model to study the dynamics and regulation of folliculogenesis in intact individual follicles in primates. Besides the research relevance, in vitro follicle maturation (IFM) is emerging as a promising approach to offer options for fertility preservation in female patients with cancer. This review summarizes the current published data on in vitro follicular development from the preantral to small antral stage in nonhuman primates, including follicle survival and growth, endocrine (ovarian steroid hormone) and paracrine/autocrine (local factor) function, as well as oocyte maturation and fertilization. Future directions include major challenges and strategies to further improve follicular growth and differentiation with oocytes competent for in vitro fertilization and subsequent embryonic development, as well as opportunities to investigate primate folliculogenesis by utilizing this 3D culture system. The information may be valuable in identifying optimal conditions for human follicle culture, with the ultimate goal of translating the experimental results and products to patients, thereby facilitating diagnostic and therapeutic approaches for female fertility.
Collapse
Affiliation(s)
- Jing Xu
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA
| | - Min Xu
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Chicago, Illinois 61611, USA
| | - Marcelo P Bernuci
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA
| | - Thomas E Fisher
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA
- Department of Obstetrics & Gynecology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA
| | - Lonnie D Shea
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Chicago, Illinois 61611, USA
| | - Teresa K Woodruff
- Department of Obstetrics & Gynecology, Feinberg School of Medicine, Northwestern University, 303 E Superior Street, Chicago, Illinois 61611, USA
| | - Mary B Zelinski
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA
- Department of Obstetrics & Gynecology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA
| | - Richard L Stouffer
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA
- Department of Obstetrics & Gynecology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA
| |
Collapse
|
24
|
Grynberg M, Pierre A, Rey R, Leclerc A, Arouche N, Hesters L, Catteau-Jonard S, Frydman R, Picard JY, Fanchin R, Veitia R, di Clemente N, Taieb J. Differential regulation of ovarian anti-müllerian hormone (AMH) by estradiol through α- and β-estrogen receptors. J Clin Endocrinol Metab 2012; 97:E1649-57. [PMID: 22689696 DOI: 10.1210/jc.2011-3133] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
BACKGROUND Anti-müllerian hormone (AMH) is a member of the TGF-β family, which limits follicle maturation. Recently serum AMH has been recognized as a useful diagnostic and prognostic tool in human reproductive endocrinology. OBJECTIVE The aim of this study was to investigate the regulation of human ovarian AMH by estradiol and FSH. METHODS AMH mRNA were quantified by real time RT-PCR in human granulosa cells (GC). AMH transcription was studied in KK1 GC cotransfected with estrogen receptors (ER)-β or ERα, and normal human AMH promoter-luciferase construct (hAMH-luc) or mutated AMH promoter reporter constructs. Binding sites for estradiol (estrogen response element half-site) and steroidogenic factor 1 were disrupted by targeted mutagenesis. The level of ER in GC was determined by quantitative RT-PCR and Western blotting. RESULTS In KK1 cells, estradiol up-regulated and inhibited hAMH-luc in the presence of ERα and ERβ respectively. Disruption of estrogen response element half-site and/or steroidogenic factor 1 binding sites did not modify ERβ-mediated effect of estradiol on hAMH-luc, whereas it affected that conveyed by ERα. The FSH enhancement of hAMH-luc was abolished by estradiol in cells overexpressing ERβ. When both ER were transfected, estradiol inhibited hAMH-luc or had no effect. Estradiol repressed AMH mRNAs in human GC, which express a little more ERα than ERβ mRNA. CONCLUSIONS Our results show that AMH expression can be differentially regulated by estradiol depending on the ER and suggest that its decrease in GC of growing follicles, which mainly express ERβ, and during controlled ovarian hyperstimulation is due to the effect of estradiol.
Collapse
Affiliation(s)
- Michaël Grynberg
- Institut National de la Santé et de la Recherche Médicale Unité 782, 32 Rue des Carnets, Clamart F-92140, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rico C, Médigue C, Fabre S, Jarrier P, Bontoux M, Clément F, Monniaux D. Regulation of Anti-Müllerian Hormone Production in the Cow: A Multiscale Study at Endocrine, Ovarian, Follicular, and Granulosa Cell Levels1. Biol Reprod 2011; 84:560-71. [DOI: 10.1095/biolreprod.110.088187] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
26
|
Xu J, Lawson MS, Yeoman RR, Pau KY, Barrett SL, Zelinski MB, Stouffer RL. Secondary follicle growth and oocyte maturation during encapsulated three-dimensional culture in rhesus monkeys: effects of gonadotrophins, oxygen and fetuin. Hum Reprod 2011; 26:1061-72. [PMID: 21362681 DOI: 10.1093/humrep/der049] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND An alginate-based matrix supports the three-dimensional (3D) architecture of non-human primate follicles and, in the presence of FSH, permits the in vitro development of pre-antral follicles to the small antral stage, including the production of ovarian steroids and paracrine factors. The current study investigated the ability of gonadotrophins, fetuin and oxygen (O₂) to improve primate follicle growth and oocyte maturation in vitro. METHODS Macaque secondary follicles were isolated from the early follicular phase ovaries, encapsulated in a sodium alginate matrix and cultured individually for 40 days in supplemented medium. The effects of recombinant human (rh) FSH (15, 3 and 0.3 ng/ml for high, medium and low FSH, respectively), bovine fetuin (1 or 0 mg/ml) and O₂ (5 or 20% v/v) were examined. Half of the follicles in each culture condition received rhLH on Day 30-40. Follicles that reached antral stage were treated with rh chorionic gonadotrophin for 34 h to initiate oocyte meiotic maturation. Media were analyzed for ovarian steroids and anti-müllerian hormone (AMH). RESULTS Improved culture conditions supported non-human primate, secondary follicle growth to the antral stage and, for the first time, promoted oocyte maturation to the MII stage. In the presence of fetuin at 5% O₂, follicles had the highest survival rate if cultured with high or medium FSH, whereas follicles grew to larger diameters at Week 5 in low FSH. Oocyte health and maturation were promoted under 5% O₂. High FSH stimulated steroid production by growing follicles, and steroidogenesis by follicles cultured with low FSH was promoted by LH. AMH biosynthesis was elevated with high compared with low FSH and for longer under 5% O₂ than under 20% O₂. CONCLUSIONS This encapsulated 3D culture model permits further studies on the endocrine and local factors that influence primate follicle growth and oocyte maturation, with relevance to enhancing fertility preservation options in women.
Collapse
Affiliation(s)
- J Xu
- Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Taieb J, Grynberg M, Pierre A, Arouche N, Massart P, Belville C, Hesters L, Frydman R, Catteau-Jonard S, Fanchin R, Picard JY, Josso N, Rey RA, di Clemente N. FSH and its second messenger cAMP stimulate the transcription of human anti-Müllerian hormone in cultured granulosa cells. Mol Endocrinol 2011; 25:645-55. [PMID: 21330407 DOI: 10.1210/me.2010-0297] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Anti-Müllerian hormone (AMH), also called Müllerian-inhibiting substance, a member of the TGF-ß family, is responsible for the regression of Müllerian ducts in the male fetus. In females, AMH is synthesized by granulosa cells of preantral and small antral follicles, and production wanes at later stages of follicle maturation. Using RT-PCR in luteal granulosa cells in primary culture and reporter gene techniques in the KK1 granulosa cell line, we show that FSH and cAMP enhance AMH transcription, and LH has an additive effect. Gonadotropins and cAMP act through protein kinase A and p38 MAPK signaling pathways and involve the GATA binding factor-4 and steroidogenic factor-1 transcription factors, among others. The expression profile of AMH and the dynamics of serum AMH after gonadotropin stimulation have been interpreted as a down-regulating effect of FSH upon AMH production by granulosa cells. The specific effect of gonadotropins upon granulosa cells may be obscured in vivo by the effect of FSH upon follicular maturation and by the presence of other hormones and growth factors, acting individually or in concert.
Collapse
Affiliation(s)
- Joëlle Taieb
- Institut National de la Santé et de la Recherche Médicale, Unité 782, 32 rue des Carnets, 92120 Clamart, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
von Schönfeldt V, Chandolia R, Kiesel L, Nieschlag E, Schlatt S, Sonntag B. Advanced follicle development in xenografted prepubertal ovarian tissue: the common marmoset as a nonhuman primate model for ovarian tissue transplantation. Fertil Steril 2010; 95:1428-34. [PMID: 21122840 DOI: 10.1016/j.fertnstert.2010.11.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 10/28/2010] [Accepted: 11/02/2010] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To establish a nonhuman primate model addressing follicular development in cryopreserved prepubertal ovarian tissue after xenografting. DESIGN Experimental study. SETTING Academic research center. ANIMAL(S) Ovarian tissue from female prepubertal common marmoset (Callytrix jacchus jacchus) grafted into immunodeficient nude mice (Crl:NU-FoxnI(nu)). INTERVENTION(S) Removal and subsequent cryopreservation of ovarian tissues with dimethyl sulfoxide, followed by grafting to subcutaneous sites of ovariectomized and intact nude mice. MAIN OUTCOME MEASURE(S) Histologic evaluation for the mean number of total and morphologically normal follicles in each class. RESULT(S) The mean number of unadvanced follicles in frozen-thawed grafted ovarian tissues was reduced compared with pregraft controls, but the prevalence of normal follicular morphology was either slightly increased (primordial follicles) or unchanged (primary follicles). Previous ovariectomy in graft recipients increased total follicle numbers without effect on normal follicular morphology and shifted the ratio of primordial to primary follicles toward an increase in primary follicles, indicating activation of follicular maturation. CONCLUSION(S) The marmoset is a suitable primate model for studies on the subsequent use of cryopreserved ovarian tissue, demonstrating graft sustainment and the development of follicles from prepubertal ovarian tissue in immunodeficient hosts up to secondary and preantral stages.
Collapse
Affiliation(s)
- Viktoria von Schönfeldt
- Department of Obstetrics and Gynecology, Campus Grosshadern, Ludwig-Maximilians-Universität Munich, Munich, Germany
| | | | | | | | | | | |
Collapse
|
29
|
Garside SA, Henkin J, Morris KD, Norvell SM, Thomas FH, Fraser HM. A thrombospondin-mimetic peptide, ABT-898, suppresses angiogenesis and promotes follicular atresia in pre- and early-antral follicles in vivo. Endocrinology 2010; 151:5905-15. [PMID: 20881256 DOI: 10.1210/en.2010-0283] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Using a novel in vitro angiogenesis assay, we previously showed that thrombospondin (TSP)-1 has antiangiogenic effects on rat follicles and induces apoptosis in granulosa cells in vitro. ABT-898 is an octapeptide mimetic of TSP-1 closely related to ABT-510. Here, we demonstrate the inhibitory effects of ABT-898 on follicular angiogenesis and its proapoptotic effect on granulosa cells. To investigate the potential of this peptide to inhibit follicular angiogenesis in vivo, marmoset monkeys were treated with 2.5 mg/kg ABT-898 twice daily throughout the follicular phase of the cycle. Although treatment did not block emergence of dominant follicles, angiogenesis was reduced in preantral and early-antral follicles. Furthermore, the incidence of atresia at these follicle stages was increased. To investigate whether treatment with ABT-898 would interfere with the timing or duration of the normal ovulatory rise in plasma progesterone, marmosets were treated with a depot formulation containing 25 mg ABT-898 at the start of the follicular phase, with a second injection after 2 wk. Despite active concentrations of peptide being maintained in the circulation, no apparent effects on the ovulatory cycle were observed. Taken together, these results indicate that ABT-898 is capable of having a dual effect by inhibiting follicular angiogenesis and promoting atresia of antral follicles in vivo but does not prevent ovulation or induce luteolysis, as has been observed with direct vascular endothelial growth factor inhibitors. These results suggest that ABT-898 could be a novel therapeutic to inhibit abnormal angiogenesis and induce atresia of accumulated follicles in polycystic ovary syndrome.
Collapse
Affiliation(s)
- Samantha A Garside
- Medical Research Council Human Reproductive Sciences Unit, Centre for Reproductive Biology, Queens Medical Research Institute, Edinburgh, UK.
| | | | | | | | | | | |
Collapse
|
30
|
Lee JR, Kim SH, Kim SM, Jee BC, Ku SY, Suh CS, Choi YM, Kim JG, Moon SY. Anti-Mullerian hormone dynamics during controlled ovarian hyperstimulation and optimal timing of measurement for outcome prediction. Hum Reprod 2010; 25:2597-2604. [PMID: 20729237 DOI: 10.1093/humrep/deq204] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2024] Open
Abstract
BACKGROUND Anti-Müllerian hormone (AMH) has been suggested as a marker of ovarian reserve and predictor of ovarian response to controlled ovarian hyperstimulation (COH). Several studies have demonstrated AMH changes during follicular and luteal phases during COH, but not after human chorionic gonadotrophin (hCG) administration. The objectives of this study were to investigate changes in AMH levels during the entire COH cycle and to clarify the regulatory mechanism of AMH secretion. In addition, we analyzed the COH outcome parameters to determine the optimal timing for AMH measurement to predict outcome. METHODS The study included 74 women who underwent in vitro fertilization (IVF) cycles with a GnRH agonist or antagonist protocol. Serum AMH and inhibin B levels were measured at baseline, Day 5 of stimulation (d5), day of hCG administration (dhCG), day of oocyte retrieval (dOPU) and 14 days after oocyte retrieval (dPO14). Follicular fluid (FF) from dominant follicles upon oocyte retrieval were also analyzed for AMH and inhibin B concentrations. AMH levels were analyzed for changes during the cycle and for correlations with COH outcome parameters. RESULTS Serum AMH levels decreased progressively during COH until dhCG, then increased on dOPU and further increased on dPO14. Serum and FF AMH levels and dynamic changes were not different between the GnRH agonist and antagonist cycles. Serum AMH levels on every sample day and the FF AMH levels were significantly correlated with outcomes of COH, such as dose of gonadotrophins used, estradiol level on dhCG and number of retrieved oocytes; the strength of the relationship was highest for baseline AMH. CONCLUSIONS The results of the present study suggest that changes in the hormonal milieu during stimulation and after the LH surge may affect AMH secretion. Serum AMH levels during COH are good markers to predict ovarian response, but the baseline serum level seems to be the most predictive marker.
Collapse
Affiliation(s)
- Jung Ryeol Lee
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, Seongnam 463-707, Republic of Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Xu J, Bernuci MP, Lawson MS, Yeoman RR, Fisher TE, Zelinski MB, Stouffer RL. Survival, growth, and maturation of secondary follicles from prepubertal, young, and older adult rhesus monkeys during encapsulated three-dimensional culture: effects of gonadotropins and insulin. Reproduction 2010; 140:685-97. [PMID: 20729335 DOI: 10.1530/rep-10-0284] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A three-dimensional culture system supports the development of primate preantral follicles to the antral stage with appreciable steroid production. This study assessed i) whether in vitro developmental competence of follicles is age dependent, ii) the role of gonadotropins and insulin in supporting folliculogenesis, and iii) anti-Müllerian hormone (AMH) and vascular endothelial growth factor (VEGF) production by growing follicles. Ovaries were obtained from prepubertal, young, and older adult rhesus macaques. Secondary follicles were encapsulated into alginate beads and cultured individually for 40 days in media containing 0.05 or 5 μg/ml insulin, with or without recombinant human (rh) FSH (500 mIU/ml). No follicles survived in the culture without rhFSH. In the presence of rhFSH, survival was lower for follicles from older animals, whereas growth, i.e. follicle diameter, was less by day 40 for follicles from prepubertal animals. The surviving follicles were categorized as no-grow (NG; ≤ 250 μm), slow-grow (SG; 250-500 μm), and fast-grow (FG; ≥ 500 μm) according to their diameters. SG follicles cultured with 5 μg/ml insulin produced more ovarian steroids than those cultured with 0.05 μg/ml insulin by week 5. SG and FG follicles produced more AMH and VEGF than the NG, and levels peaked at weeks 2 and 5 respectively. After 100 ng/ml rh chorionic gonadotropin treatment for 34 h, more healthy oocytes were retrieved from young adults whose follicles were cultured with 5 μg/ml insulin. This culture system offers an opportunity to characterize the endocrine and paracrine function of primate follicles that influence follicle growth and oocyte maturation.
Collapse
Affiliation(s)
- Jing Xu
- Division of Reproductive Sciences, Oregon National Primate Research Center, Oregon Health & Science University, 505 Northwest 185th Avenue, Beaverton, Oregon 97006, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
McEwan IJ, McGuinness D, Hay CW, Millar RP, Saunders PTK, Fraser HM. Identification of androgen receptor phosphorylation in the primate ovary in vivo. Reproduction 2010; 140:93-104. [PMID: 20406952 PMCID: PMC2892820 DOI: 10.1530/rep-10-0140] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The androgen receptor (AR) is a member of the nuclear receptor superfamily, and is important for both male and female reproductive health. The receptor is a target for a number of post-translational modifications including phosphorylation, which has been intensively studied in vitro. However, little is known about the phosphorylation status of the receptor in target tissues in vivo. The common marmoset is a useful model for studying human reproductive functions, and comparison of the AR primary sequence from this primate shows high conservation of serines known to be phosphorylated in the human receptor and corresponding flanking amino acids. We have used a panel of phosphospecific antibodies to study AR phosphorylation in the marmoset ovary throughout the follicular phase and after treatment with GNRH antagonist or testosterone propionate. In normal follicular phase ovaries, total AR (both phosphorylated and non-phosphorylated forms) immunopositive staining was observed in several cell types including granulosa cells of developing follicles, theca cells and endothelial cells lining blood vessels. Receptor phosphorylation at serines 81, 308, and 650 was detected primarily in the granulosa cells of developing follicles, surface epithelium, and vessel endothelial cells. Testosterone treatment lead to a modest increase in AR staining in all stages of follicle studied, while GNRH antagonist had no effect. Neither treatment significantly altered the pattern of phosphorylation compared to the control group. These results demonstrate that phosphorylation of the AR occurs, at a subset of serine residues, in a reproductive target tissue in vivo, which appears refractory to hormonal manipulations.
Collapse
Affiliation(s)
- Iain J McEwan
- MRC Human Reproductive Sciences Unit, Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, Scotland, UK.
| | | | | | | | | | | |
Collapse
|
33
|
Eilso Nielsen M, Rasmussen IA, Fukuda M, Westergaard LG, Yding Andersen C. Concentrations of anti-Mullerian hormone in fluid from small human antral follicles show a negative correlation with CYP19 mRNA expression in the corresponding granulosa cells. Mol Hum Reprod 2010; 16:637-43. [DOI: 10.1093/molehr/gaq001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
34
|
Abstract
OBJECTIVE A monkey model of the menopausal transition (perimenopause) would facilitate efforts to understand better the effect of hormonal fluctuations during this life phase on the initiation of chronic diseases associated with the postmenopausal years. Antimüllerian hormone (AMH) is a promising marker of ovarian reserve (primordial follicle number) in women. Here, we describe the relationship between AMH and ovarian reserve in cynomolgus monkeys (Macaca fascicularis) estimated to be 12 to 15 years of age (approximately 36-45 y in women). METHODS AND RESULTS The results of daily vaginal swabbing (to detect menses) and thrice weekly blood sampling for 12 weeks indicate that AMH is relatively stable across the menstrual cycle (intraclass correlation, approximately 0.80), with a slight although significant (P < 0.02) reduction (approximately 1.4-fold) on days 2 to 5 postovulation. Substantial interindividual variation in AMH concentrations were observed between monkeys, with values ranging from 4.46 +/- 0.17 to 18.80 +/- 0.71 ng/mL (mean +/- SE). Antimüllerian hormone concentrations were reduced by approximately 63% after the removal of one ovary (7.6 +/- 0.77 vs 2.75 +/- 0.37 ng/mL; P < 0.001; n = 19) and were below the level of detection after the removal of both ovaries (5.8 +/- 0.42 to <0.05 ng/mL; P < 0.001; n = 84), suggesting that the ovary is likely to be either the major or the sole source of AMH in the monkey. Finally, we examined the association between AMH and primordial, primary, and secondary follicles in 29 monkeys and found significant associations with all follicle types (r = 0.78, r = 0.66, and r = 0.80, respectively; P < 0.01). CONCLUSIONS The relationship between AMH and ovarian reserve in the monkey is similar to that in women, suggesting that monkeys may be a useful model for studying hormonal fluctuations across the menopausal transition.
Collapse
|
35
|
Dumesic DA, Lesnick TG, Stassart JP, Ball GD, Wong A, Abbott DH. Intrafollicular antimüllerian hormone levels predict follicle responsiveness to follicle-stimulating hormone (FSH) in normoandrogenic ovulatory women undergoing gonadotropin releasing-hormone analog/recombinant human FSH therapy for in vitro fertilization and embryo transfer. Fertil Steril 2009; 92:217-21. [PMID: 18675414 DOI: 10.1016/j.fertnstert.2008.04.047] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 04/21/2008] [Accepted: 04/21/2008] [Indexed: 12/01/2022]
Abstract
OBJECTIVE To investigate the relationship between antimüllerian hormone (AMH) and steroidogenesis in follicles of normoandrogenic ovulatory women undergoing follicle-stimulating hormone (FSH) therapy for in vitro fertilization/embryo transfer (IVF-ET). DESIGN Prospective cohort. SETTING Institutional/private practice. PATIENT(S) 26 normoandrogenic ovulatory women. All women received gonadotropin-releasing hormone (GnRH) analog and ovarian stimulation for IVF-ET. INTERVENTION(S) Follicle fluid was aspirated at oocyte retrieval from the first follicle of each ovary. MAIN OUTCOME MEASURE(S) Follicle fluid was assayed for AMH, estradiol (E(2)), progesterone, androstenedione, testosterone, dihydrotestosterone, insulin, and FSH. RESULT(S) Intrafollicular AMH levels positively and negatively correlated with E(2) and FSH concentrations in follicles, respectively, causing a positive relationship between follicle fluid AMH levels and E(2)/FSH ratios as a measure of follicle sensitivity to FSH. A positive relationship also existed in follicles between AMH levels and E(2)/androgen ratios as a marker of aromatase activity. CONCLUSION(S) The AMH levels in follicles of IVF patients positively correlate with follicle sensitivity to FSH.
Collapse
Affiliation(s)
- Daniel A Dumesic
- Reproductive Medicine and Infertility Associates, 2101 Woodwinds Drive, Woodbury, MN 55125, USA.
| | | | | | | | | | | |
Collapse
|
36
|
Abir R, Ao A, Zhang XY, Garor R, Nitke S, Fisch B. Vascular endothelial growth factor A and its two receptors in human preantral follicles from fetuses, girls, and women. Fertil Steril 2009; 93:2337-47. [PMID: 19409555 DOI: 10.1016/j.fertnstert.2009.01.111] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2008] [Revised: 01/18/2009] [Accepted: 01/19/2009] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the expression of vascular endothelial growth factor A (VEGF-A) and that its two receptors (VEGFR1, VEGFR2) in human preantral follicles. DESIGN Immunohistochemical, in situ hybridization, and reverse transcriptase polymerase chain reaction (RT-PCR) study of the expression of the VEGF-A system in human ovaries. SETTING Major tertiary-care academic center. PATIENT(S) Twenty-two patients who underwent pregnancy terminations at 21-35 gestational weeks and 29 girls/women aged 5-39 years who underwent ovarian laparoscopies. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Laboratory analysis of human ovarian specimens. RESULT(S) Immunhistochemistry and in situ hybridization revealed the expression of the proteins and mRNA transcripts for VEGFR1 and VEGFR2 in oocytes, granulosa cells, and stroma cells from fetuses and girls/women. The protein for VEGF-A was detected immunohistochemically in oocytes, granulosa cells, and stroma cells from fetuses and girls. VEGF-A and VEGFR1 proteins were expressed more strongly than VEGFR2. VEGF-A(121), VEGF-A(165), and VEGF-A(189) isoforms were identified by RT-PCR in the ovarian samples from fetuses and women. CONCLUSION(S) The presence of the VEGF-A receptors, particularly in the granulosa cells, suggests that VEGF-A might be involved in proliferation initiation of primordial follicles or play an as yet unknown role in human preantral follicles.
Collapse
Affiliation(s)
- Ronit Abir
- Helen Schneider Hospital for Women, Rabin Medical Center, Beilinson Hospital, Petach Tikva 49100, Israel.
| | | | | | | | | | | |
Collapse
|
37
|
Gaytán F, Gaytán M, Castellano JM, Romero M, Roa J, Aparicio B, Garrido N, Sánchez-Criado JE, Millar RP, Pellicer A, Fraser HM, Tena-Sempere M. KiSS-1 in the mammalian ovary: distribution of kisspeptin in human and marmoset and alterations in KiSS-1 mRNA levels in a rat model of ovulatory dysfunction. Am J Physiol Endocrinol Metab 2009; 296:E520-31. [PMID: 19141682 DOI: 10.1152/ajpendo.90895.2008] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Kisspeptins, the products of the KiSS-1 gene acting via G protein-coupled receptor 54 (GPR54), have recently emerged as pivotal signals in the hypothalamic network triggering the preovulatory surge of gonadotropins and, hence, ovulation. Additional actions of kisspeptins at other levels of the hypothalamic-pituitary-ovarian axis have been suggested but remain to date scarcely studied. We report herein the pattern of expression of KiSS-1 and GPR54 in the human and nonhuman primate ovary and evaluate changes in ovarian KiSS-1 expression in a rat model of ovulatory dysfunction. KiSS-1 and GPR54 mRNAs were detected in human ovarian tissue and cultured granulosa-lutein cells. In good agreement, kisspeptin immunoreactivity was observed in cyclic human and marmoset ovaries, with prominent signals in the theca layer of growing follicles, corpora lutea, interstitial gland, and ovarian surface epithelium. GPR54 immunoreactivity was also found in human theca and luteal cells. Administration of indomethacin to cyclic female rats disturbed ovulation and resulted in a dramatic drop in ovarian KiSS-1, but not GPR54, cyclooxygenase-2 (COX-2), or progesterone receptor, mRNA levels at the time of ovulation; an effect mimicked by the selective COX-2 inhibitor NS398 and rescued by coadministration of PGE(2). Likewise, the stimulatory effect of human choriogonadotropin on ovarian KiSS-1 expression was partially blunted by indomethacin. In contrast, KiSS-1 mRNA levels remained unaltered in another model of ovulatory failure, i.e., the RU486-treated rat. In summary, we document for the first time the expression of KiSS-1/kisspeptin and GPR54 in the human and nonhuman primate ovary. In addition, we provide evidence for the ability of inhibitors of COX-2, known to disturb follicular rupture and ovulation, to selectively alter the expression of KiSS-1 gene in rat ovary. Altogether, our results are suggestive of a conserved role of local KiSS-1 in the direct control of ovarian functions in mammals.
Collapse
Affiliation(s)
- F Gaytán
- Department of Cell Biology, University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Duncan WC, van den Driesche S, Fraser HM. Inhibition of vascular endothelial growth factor in the primate ovary up-regulates hypoxia-inducible factor-1alpha in the follicle and corpus luteum. Endocrinology 2008; 149:3313-20. [PMID: 18388198 DOI: 10.1210/en.2007-1649] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Vascular endothelial growth factor (VEGF)-dependent angiogenesis is crucial for follicular growth, and corpus luteum formation and function, in the primate ovary. In the ovary VEGF can be hormonally regulated, but in other systems, the main regulator of VEGF expression is hypoxia. We hypothesized that hypoxia was involved in the regulation of angiogenesis in the cycling ovary. We therefore used immunohistochemistry to localize hypoxia-inducible factor (HIF)-1alpha in the marmoset ovary across the ovarian cycle. We also investigated the effect of VEGF inhibition, using VEGF Trap (aflibercept), on HIF-1alpha localization during the follicular and luteal phases of the cycle. Finally, we studied the effect of chorionic gonadotropin stimulation of the corpus luteum during early pregnancy. Nuclear HIF-1alpha staining was largely absent from normally growing preantral and antral follicles. However, there was marked up-regulation of nuclear HIF-1alpha in the granulosa cells at ovulation that persisted into the early corpus luteum. Mature corpora lutea and those collected during early pregnancy had minimal nuclear HIF-1alpha staining. The inhibition of VEGF in the mid-luteal stage resulted in a time-dependent up-regulation of luteal nuclear HIF-1alpha staining (P < 0.05). There was never any nuclear HIF-1alpha in the theca cells of the follicle, but VEGF Trap treatment during the follicular (P < 0.001) or luteal (P < 0.001) phase increased the proportion of antral follicles with nuclear HIF-1alpha staining in the granulosa cells. These results indicate that HIF-1alpha is up-regulated after vascular inhibition, using VEGF Trap, in the follicle and corpus luteum. However, it is also acutely up-regulated during ovulation. This suggests a role for HIF-1alpha in both hypoxic and hormonal regulation of ovarian VEGF expression in vivo.
Collapse
Affiliation(s)
- W Colin Duncan
- Obstetrics and Gynaecology, Simpson Centre for Reproductive Health, Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, United Kingdom.
| | | | | |
Collapse
|
39
|
Thomas FH, Wilson H, Silvestri A, Fraser HM. Thrombospondin-1 expression is increased during follicular atresia in the primate ovary. Endocrinology 2008; 149:185-92. [PMID: 17884943 DOI: 10.1210/en.2007-0835] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Thrombospondin (TSP)-1 is an antiangiogenic extracellular matrix glycoprotein that modulates several aspects of cellular function. The aim of this study was to determine the pattern of TSP-1 mRNA and protein expression as well as expression of its receptor CD36 in the marmoset ovary and to investigate the effects of inhibition of gonadotropins or VEGF activity on TSP-1 and CD36 expression in vivo. GnRH antagonist or VEGF Trap, a soluble decoy receptor, was administered on d 0 of the follicular phase of the cycle, and ovaries were collected at the end of the follicular phase (d 10). TSP-1 mRNA and protein were present in granulosa cells of preantral and antral follicles, with the highest staining at the late secondary and tertiary stages. Moreover, expression of TSP-1 mRNA and protein was significantly increased in tertiary follicles undergoing atresia. CD36 protein was detected in granulosa cells of preantral and antral follicles as well as in endothelial cells of large vessels. Inhibition of gonadotropin secretion or VEGF activity had no effect on TSP-1 expression; however, expression of CD36 protein was inhibited by the VEGF Trap. In conclusion, TSP-1 may be involved in the cessation of angiogenesis in follicles undergoing atresia; alternatively, TSP-1 may act on granulosa and/or endothelial cells to promote follicular atresia in the ovary. Angiogenesis is likely to involve a balance between pro- and antiangiogenic factors. Our results suggest that loss of VEGF activity does not regulate TSP-1 expression directly but may influence TSP-1 activity via down-regulation of the CD36 receptor.
Collapse
Affiliation(s)
- Fiona H Thomas
- Medical Research Council Human Reproductive Sciences Unit, University of Edinburgh Centre for Reproductive Biology, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| | | | | | | |
Collapse
|