1
|
Bilici ME, Şıklar Z, Çetinkaya S, Özsu E, Aycan Z, Berberoğlu M. Adrenal hypoandrogenism in adolescents with premature ovarian insufficiency. J Pediatr Endocrinol Metab 2025; 38:262-270. [PMID: 39786952 DOI: 10.1515/jpem-2024-0415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/14/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVES Premature ovarian insufficiency (POI) affects 1 in 10,000 children, with its molecular causes largely unknown. Adult studies suggest that low androgen levels induce ovarian insufficiency, but data on about this in children is limited. This study aims to assess the prevalence of low androgen levels in childhood POI and its relationship with adrenal insufficiency. METHODS Idiopathic POI adolescents were categorized into two groups based on DHEAS and total testosterone (TT) measured by chemiluminescence. Low androgen group (LAG) was defined using cut-offs according to Tanner pubarche staging. Demographic, clinical, and laboratory data were compared. Morning cortisol <7 μg/dL and/or ACTH >96 or <5 pg/mL were planned to undergo ACTH stimulation testing, with a peak cortisol response <18 μg/dL considered insufficient. RESULTS Forty-three adolescents, mean age 15.5 ± 1.3 years with a 46, XX karyotype, normal FMR1 mutation, FSH levels >40 mIU/mL, and low AMH levels were included. In 14 cases (37.8 %), DHEAS and TT were low. In the LAG, pubarche was absent in seven patients, and initial height SDS was significantly lower. Morning cortisol ranged from 7.9 to 23.5 μg/dL, with an ACTH of 29.4 ± 9.7 pg/mL. No differences in adrenal steroids or correlations between DHEAS and ACTH were observed. CONCLUSIONS Diminished androgen levels are prevalent in children with idiopathic POI. The potential for this condition to increase the risk of adrenal insufficiency and its impact on secondary ovarian insufficiency remains unclear. This study, the first of its kind in children, underscores the potential role of genetic factors in zona reticularis and ovarian development.
Collapse
Affiliation(s)
- Meliha Esra Bilici
- Bülent Ecevit University Faculty of Medicine, Department of Pediatric Endocrinology and Diabetes, 67630 Zonguldak, Türkiye
| | - Zeynep Şıklar
- Department of Pediatric Endocrinology, Ankara University School of Medicine, Ankara, Türkiye
| | - Semra Çetinkaya
- Dr Sami Ulus Training and Research Hospital for Maternity and Children's Health and Diseases, Ankara, Türkiye
| | - Elif Özsu
- Department of Pediatric Endocrinology, Ankara University School of Medicine, Ankara, Türkiye
| | - Zehra Aycan
- Department of Pediatric Endocrinology, Ankara University School of Medicine, Ankara, Türkiye
| | - Merih Berberoğlu
- Department of Pediatric Endocrinology, Ankara University School of Medicine, Ankara, Türkiye
| |
Collapse
|
2
|
Jinno M. Ovarian stimulation by promoting basal follicular growth. Reprod Biol Endocrinol 2025; 23:35. [PMID: 40050948 PMCID: PMC11884117 DOI: 10.1186/s12958-025-01356-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/04/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Most methods of ovarian stimulation rely on gonadotropin modulation. However, abnormal anti-Müllerian hormone concentrations are frequent in infertility, suggesting that defects in the gonadotropin-independent period of folliculogenesis preceding cyclic recruitment (i.e., basal follicular growth) may often occur. We need to better understand basal follicular growth and determine how to improve it. METHODS Section I summarizes a literature search concerning preantral and early antral folliculogenesis, cyclic recruitment, and selection. Section II presents current knowledge about interventions involving early antral folliculogenesis and cyclic recruitment. RESULTS While folliculogenesis following cyclic recruitment is gonadotropin-dependent, basal follicular growth is not. Basal follicular growth is regulated by follicle-stimulating hormone and local communication between the oocyte and its granulosa and thecal cells involving gap junctions and many autocrine/paracrine factors. This local communication sustains growth synergistically with follicle-stimulating hormone, but also suppresses this hormone to induce granulosa cell differentiation. As a follicle develops, its responsiveness to gonadotropin progressively increases. Section II describes 4 interventions affecting early antral folliculogenesis, including granulocyte colony-stimulating factor priming, bromocriptine rebound, carbohydrate metabolism intervention, and danazol priming, which have improved embryo development and live birth rate in patients with previous failures. CONCLUSION Basal follicular growth modulation can increase live birth rates.
Collapse
Affiliation(s)
- Masao Jinno
- Women's Clinic Jinno, 3-11-7 Kokuryou-Chou, Choufu City, Tokyo, 182-0022, Japan.
| |
Collapse
|
3
|
Bosdou JK, Venetis CA, Anagnostis P, Savvaidou D, Chatzimeletiou K, Zepiridis L, Goulis DG, Grimbizis G, Kolibianakis EM. Association of Basal Serum Androgen Concentration with Follicles Number on the Day of Triggering Final Oocyte Maturation in Low Responders According to the Bologna Criteria: A Prospective Cohort Study. Int J Mol Sci 2025; 26:1656. [PMID: 40004120 PMCID: PMC11855118 DOI: 10.3390/ijms26041656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Studies in animals have shown that androgens promote early follicular development and granulosa cell proliferation by augmenting follicle-stimulating hormone (FSH) receptor expression in granulosa cells. Inconsistency exists regarding the association between basal serum androgen levels and follicular development in low responders undergoing in vitro fertilization (IVF), although the number of studies is limited. The aim of the current study was to assess the association between basal serum androgen concentrations and the number of follicles ≥ 11 mm on the day of triggering final oocyte maturation in low responders undergoing IVF. This prospective study was performed from June 2020 to September 2024 in 96 low responders, categorized according to the Bologna criteria. Total testosterone, dehydroepiandrosterone sulfate (DHEAS), 17-OH progesterone (17-OH-P), Δ4-androstenedione (Δ4-A), and sex hormone-binding globulin (SHBG) were measured on the day of initiation of ovarian stimulation. No association was found between basal serum testosterone (coef.: -0.002, p = 0.98), DHEAS (coef.: -0.096, p = 0.35), 17-OH-P (coef.: +0.086, p = 0.40), Δ4-A (coef.: -0.028, p = 0.79), and SHBG (coef.: +0.160, p = 0.12) concentrations and the number of follicles ≥ 11 mm on the day of triggering final oocyte maturation. The results of the current study challenge the usefulness of basal serum androgen measurements prior to ovarian stimulation in low responders as predictors of ovarian response.
Collapse
Affiliation(s)
- Julia K. Bosdou
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (J.K.B.); (C.A.V.); (D.S.); (K.C.); (L.Z.); (G.G.); (E.M.K.)
| | - Christos A. Venetis
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (J.K.B.); (C.A.V.); (D.S.); (K.C.); (L.Z.); (G.G.); (E.M.K.)
| | - Panagiotis Anagnostis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece;
| | - Despoina Savvaidou
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (J.K.B.); (C.A.V.); (D.S.); (K.C.); (L.Z.); (G.G.); (E.M.K.)
| | - Katerina Chatzimeletiou
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (J.K.B.); (C.A.V.); (D.S.); (K.C.); (L.Z.); (G.G.); (E.M.K.)
| | - Leonidas Zepiridis
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (J.K.B.); (C.A.V.); (D.S.); (K.C.); (L.Z.); (G.G.); (E.M.K.)
| | - Dimitrios G. Goulis
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece;
| | - Grigoris Grimbizis
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (J.K.B.); (C.A.V.); (D.S.); (K.C.); (L.Z.); (G.G.); (E.M.K.)
- Unit of Reproductive Endocrinology, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece;
| | - Efstratios M. Kolibianakis
- Unit for Human Reproduction, 1st Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 56429 Thessaloniki, Greece; (J.K.B.); (C.A.V.); (D.S.); (K.C.); (L.Z.); (G.G.); (E.M.K.)
| |
Collapse
|
4
|
Bülow N, Macklon N. The role of letrozole in in vitro fertilization treatment: new remedy or old mirage? Fertil Steril 2025; 123:41-49. [PMID: 39486501 DOI: 10.1016/j.fertnstert.2024.10.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
Aromatase inhibitors, particularly letrozole (LZ), are now established as first-line ovulation induction agents, offering an effective ovarian stimulation strategy to enhance outcomes of intrauterine insemination. In recent years, they have also emerged as potentially valuable adjuvants to gonadotropin ovarian stimulation for in vitro fertilization, particularly in fertility preservation in women with estrogen-responsive cancers. Their primary mechanism of action is to reduce the circulating estrogen levels by inhibiting androgen aromatization. Recent studies have provided evidence that this property may confer therapeutic advantages in other patients undergoing in vitro fertilization. In this study, evidence supporting the role of adjuvant LZ in poor responders, as a moderator of ovarian hyperstimulation syndrome symptoms, and an agent for improving the luteal phase after ovarian stimulation is reviewed. The use of LZ for endometrial preparation in the frozen-thawed embryo transfer cycle is also considered.
Collapse
Affiliation(s)
- Nathalie Bülow
- The Fertility Clinic, Department of Gynecology, Fertility and Obstetrics, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | | |
Collapse
|
5
|
Tidwell A, Zhu J, Battiola T, Welt CK. Phenotypes Associated With Polycystic Ovary Syndrome Risk Variants. J Endocr Soc 2024; 9:bvae219. [PMID: 39687684 PMCID: PMC11646653 DOI: 10.1210/jendso/bvae219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Indexed: 12/18/2024] Open
Abstract
Context Polycystic ovary syndrome (PCOS) affects 10% of women of reproductive age. The genetic architecture of the disease is emerging, but there is little data exploring the effect of genetic risk on clinical presentation. Objective We hypothesized that genetic risk loci would influence measurable phenotypic traits. Methods This retrospective cohort study, conducted at an academic medical center, included women of European ancestry with PCOS (n = 404), as diagnosed by the National Institutes of Health criteria, and controls with regular menses and no hyperandrogenism (n = 408). We identified association between genetic risk variants and measured phenotypic traits using linear regression. Results In a combined analysis of cases and controls, 2 variants in loci containing the genes PRSS23 (P < .001) and FSHB (P < .001) were associated with gonadotropin levels. Two variants in loci containing NEIL2/GATA4 (P = .002) and CYP3 (P < .001) were associated with androgen levels. Three variants in loci containing SHBG (P = .001), ZBTB16 (P < .001), and CYP3 (P < .001) were associated with ovarian morphology. One variant in the locus containing FTO (P = .001) was associated with hip circumference and was influenced by body mass index. Conclusion These results demonstrate that PCOS genetic risk variants may influence hormone levels and ovarian morphology and increase the risk of obesity. Increased genetic risk for PCOS appears to drive traits that underly the classical clinical presentation of PCOS.
Collapse
Affiliation(s)
- Anna Tidwell
- Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Jia Zhu
- Division of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tess Battiola
- Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Corrine K Welt
- Division of Endocrinology, Metabolism and Diabetes, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
6
|
Gowkielewicz M, Lipka A, Zdanowski W, Waśniewski T, Majewska M, Carlberg C. Anti-Müllerian hormone: biology and role in endocrinology and cancers. Front Endocrinol (Lausanne) 2024; 15:1468364. [PMID: 39351532 PMCID: PMC11439669 DOI: 10.3389/fendo.2024.1468364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024] Open
Abstract
Anti-Müllerian hormone (AMH) is a peptide belonging to the transforming growth factor beta superfamily and acts exclusively through its receptor type 2 (AMHR2). From the 8th week of pregnancy, AMH is produced by Sertoli cells, and from the 23rd week of gestation, it is produced by granulosa cells of the ovary. AMH plays a critical role in regulating gonadotropin secretion, ovarian tissue responsiveness to pituitary hormones, and the pathogenesis of polycystic ovarian syndrome. It inhibits the transition from primordial to primary follicles and is considered the best marker of ovarian reserve. Therefore, measuring AMH concentration of the hormone is valuable in managing assisted reproductive technologies. AMH was initially discovered through its role in the degeneration of Müllerian ducts in male fetuses. However, due to its ability to inhibit the cell cycle and induce apoptosis, it has also garnered interest in oncology. For example, antibodies targeting AMHR2 are being investigated for their potential in diagnosing and treating various cancers. Additionally, AMH is present in motor neurons and functions as a protective and growth factor. Consequently, it is involved in learning and memory processes and may support the treatment of Alzheimer's disease. This review aims to provide a comprehensive overview of the biology of AMH and its role in both endocrinology and oncology.
Collapse
Affiliation(s)
- Marek Gowkielewicz
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Aleksandra Lipka
- Institute of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | - Wojciech Zdanowski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Tomasz Waśniewski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marta Majewska
- Department of Human Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Carsten Carlberg
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
- School of Medicine, Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
7
|
Rosenfield RL. The Search for the Causes of Common Hyperandrogenism, 1965 to Circa 2015. Endocr Rev 2024; 45:553-592. [PMID: 38457123 DOI: 10.1210/endrev/bnae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/23/2023] [Accepted: 03/06/2024] [Indexed: 03/09/2024]
Abstract
From 1965 to 2015, immense strides were made into understanding the mechanisms underlying the common androgen excess disorders, premature adrenarche and polycystic ovary syndrome (PCOS). The author reviews the critical discoveries of this era from his perspective investigating these disorders, commencing with his early discoveries of the unique pattern of plasma androgens in premature adrenarche and the elevation of an index of the plasma free testosterone concentration in most hirsute women. The molecular genetic basis, though not the developmental biologic basis, for adrenarche is now known and 11-oxytestosterones shown to be major bioactive adrenal androgens. The evolution of the lines of research into the pathogenesis of PCOS is historically traced: research milestones are cited in the areas of neuroendocrinology, insulin resistance, hyperinsulinism, type 2 diabetes mellitus, folliculogenesis, androgen secretion, obesity, phenotyping, prenatal androgenization, epigenetics, and complex genetics. Large-scale genome-wide association studies led to the 2014 discovery of an unsuspected steroidogenic regulator DENND1A (differentially expressed in normal and neoplastic development). The splice variant DENND1A.V2 is constitutively overexpressed in PCOS theca cells in long-term culture and accounts for their PCOS-like phenotype. The genetics are complex, however: DENND1A intronic variant copy number is related to phenotype severity, and recent data indicate that rare variants in a DENND1A regulatory network and other genes are related to PCOS. Obesity exacerbates PCOS manifestations via insulin resistance and proinflammatory cytokine excess; excess adipose tissue also forms testosterone. Polycystic ovaries in 40 percent of apparently normal women lie on the PCOS functional spectrum. Much remains to be learned.
Collapse
Affiliation(s)
- Robert L Rosenfield
- Department of Pediatrics and Medicine, The University of Chicago, Chicago, IL 94109, USA
- Department of Pediatrics, The University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
8
|
Cakir C, Kuspinar G, Aslan K, Bozyigit C, Kasapoglu I, Dirican M, Uncu G, Avci B. Dehydroepiandrosterone modulates the PTEN/PI3K/AKT signaling pathway to alleviate 4-vinylcyclohexene diepoxide-induced premature ovarian insufficiency in rats. Exp Anim 2024; 73:319-335. [PMID: 38494723 PMCID: PMC11254495 DOI: 10.1538/expanim.23-0179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/09/2024] [Indexed: 03/19/2024] Open
Abstract
Dehydroepiandrosterone (DHEA) is frequently integrated as an adjuvant in over a quarter of controlled ovarian hyperstimulation (COH) protocols, despite the ongoing debate regarding its impact. This study aimed to evaluate the efficacy and mechanism of action of DHEA on ovarian follicular development and ovarian response in rats with varying ovarian reserves. The study involved 75 rats categorized into 15 distinct groups. The ovarian tissues of rats in both the normal ovarian reserve group and the premature ovarian insufficiency (POI) group, induced by 4-vinylcyclohexene diepoxide (VCD) injection, were subjected to histomorphological and biochemical analyses following the administration of DHEA, either alone or in combination with COH. Follicle counting was performed on histological sections obtained from various tissues. Serum concentrations of anti-Müllerian hormone (AMH) and the quantification of specific proteins in ovarian tissue, including phosphatase and tensin homolog of chromosome 10 (PTEN), phosphoinositide 3-kinase (PI3K), phosphorylated protein kinase B (pAKT), cyclooxygenase 2 (COX-2), caspase-3, as well as assessments of total antioxidant status and total oxidant status, were conducted employing the ELISA method. The impact of DHEA exhibited variability based on ovarian reserve. In the POI model, DHEA augmented follicular development and ovarian response to the COH protocol by upregulating the PTEN/PI3K/AKT signaling pathway, mitigating apoptosis, inflammation, and oxidative stress, contrary to its effects in the normal ovarian reserve group. In conclusion, it has been determined that DHEA may exert beneficial effects on ovarian stimulation response by enhancing the initiation of primordial follicles and supporting antral follicle populations.
Collapse
Affiliation(s)
- Cihan Cakir
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Goktan Kuspinar
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Kiper Aslan
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Cengiz Bozyigit
- Department of Medical Biochemistry, Bursa City Hospital, Doğanköy District, Nilüfer Bursa, 16110, Türkiye
| | - Isil Kasapoglu
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Melahat Dirican
- Department of Medical Biochemistry, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Gurkan Uncu
- Department of Obstetrics and Gynecology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| | - Berrin Avci
- Department of Histology and Embryology, Bursa Uludag University School of Medicine, Görükle Campus, Nilüfer, Bursa, 16059, Türkiye
| |
Collapse
|
9
|
Naik S, Lepine S, Nagels HE, Siristatidis CS, Kroon B, McDowell S. Androgens (dehydroepiandrosterone or testosterone) for women undergoing assisted reproduction. Cochrane Database Syst Rev 2024; 6:CD009749. [PMID: 38837771 PMCID: PMC11152211 DOI: 10.1002/14651858.cd009749.pub3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
BACKGROUND Practitioners in the field of assisted reproductive technology (ART) continually seek alternative or adjunct treatments to improve ART outcomes. This Cochrane review investigates the adjunct use of synthetic versions of two naturally produced hormones, dehydroepiandrosterone (DHEA) and testosterone (T), in assisted reproduction. Steroid hormones are proposed to increase conception rates by positively affecting follicular response to gonadotrophin stimulation. This may lead to a greater oocyte yield and, subsequently, an increased chance of pregnancy. OBJECTIVES To assess the effectiveness and safety of DHEA and T as pre- or co-treatments in infertile women undergoing assisted reproduction. SEARCH METHODS We searched the following electronic databases up to 8 January 2024: the Gynaecology and Fertility Group (CGF) Specialised Register, CENTRAL, MEDLINE, Embase, PsycINFO, and trial registries for ongoing trials. We also searched citation indexes, Web of Science, PubMed, and OpenGrey. We searched the reference lists of relevant studies and contacted experts in the field for any additional trials. There were no language restrictions. SELECTION CRITERIA Randomised controlled trials (RCTs) comparing DHEA or T as an adjunct treatment to any other active intervention, placebo, or no treatment in women undergoing assisted reproduction. DATA COLLECTION AND ANALYSIS Two review authors independently selected studies, extracted relevant data, and assessed risk of bias. We pooled data from studies using fixed-effect models. We calculated odds ratios (ORs) for each dichotomous outcome. Analyses were stratified by type of treatment. We assessed the certainty of evidence for the main findings using GRADE methods. MAIN RESULTS We included 28 RCTs. There were 1533 women in the intervention groups and 1469 in the control groups. Apart from three trials, trial participants were women identified as 'poor responders' to standard in vitro fertilisation (IVF) protocols. The included trials compared either T or DHEA treatment with placebo or no treatment. Pre-treatment with DHEA versus placebo/no treatment: DHEA likely results in little to no difference in live birth/ongoing pregnancy rates (OR 1.30, 95% confidence interval (CI) 0.95 to 1.76; I² = 16%, 9 RCTs, N = 1433, moderate certainty evidence). This suggests that in women with a 12% chance of live birth/ongoing pregnancy with placebo or no treatment, the live birth/ongoing pregnancy rate in women using DHEA will be between 12% and 20%. DHEA likely does not decrease miscarriage rates (OR 0.85, 95% CI 0.53 to 1.37; I² = 0%, 10 RCTs, N =1601, moderate certainty evidence). DHEA likely results in little to no difference in clinical pregnancy rates (OR 1.18, 95% CI 0.93 to 1.49; I² = 0%, 13 RCTs, N = 1886, moderate certainty evidence). This suggests that in women with a 17% chance of clinical pregnancy with placebo or no treatment, the clinical pregnancy rate in women using DHEA will be between 16% and 24%. We are very uncertain about the effect of DHEA on multiple pregnancy (OR 3.05, 95% CI 0.47 to 19.66; 7 RCTs, N = 463, very low certainty evidence). Pre-treatment with T versus placebo/no treatment: T likely improves live birth rates (OR 2.53, 95% CI 1.61 to 3.99; I² = 0%, 8 RCTs, N = 716, moderate certainty evidence). This suggests that in women with a 10% chance of live birth with placebo or no treatment, the live birth rate in women using T will be between 15% and 30%. T likely does not decrease miscarriage rates (OR 1.63, 95% CI 0.76 to 3.51; I² = 0%, 9 RCTs, N = 755, moderate certainty evidence). T likely increases clinical pregnancy rates (OR 2.17, 95% CI 1.54 to 3.06; I² = 0%, 13 RCTs, N = 1152, moderate certainty evidence). This suggests that in women with a 12% chance of clinical pregnancy with placebo or no treatment, the clinical pregnancy rate in women using T will be between 17% and 29%. We are very uncertain about the effect of T on multiple pregnancy (OR 2.56, 95% CI 0.59 to 11.20; 5 RCTs, N = 449, very low certainty evidence). We are uncertain about the effect of T versus estradiol or T versus estradiol + oral contraceptive pills. The certainty of the evidence was moderate to very low, the main limitations being lack of blinding in the included trials, inadequate reporting of study methods, and low event and sample sizes in the trials. Data on adverse events were sparse; any reported events were minor. AUTHORS' CONCLUSIONS Pre-treatment with T likely improves, and pre-treatment with DHEA likely results in little to no difference, in live birth and clinical pregnancy rates in women undergoing IVF who have been identified as poor responders. DHEA and T probably do not decrease miscarriage rates in women under IVF treatment. The effects of DHEA and T on multiple pregnancy are uncertain. Research is needed to identify the optimal duration of treatment with T. Future studies should include data collection on adverse events and multiple pregnancy.
Collapse
Affiliation(s)
- Sandeep Naik
- Obstetrics and Gynaecology, Capital Coast District Heath Board, Wellington, New Zealand
| | - Sam Lepine
- Department of Obstetrics and Gynaecology, Capital and Coast District Health Board, Wellington, New Zealand
| | - Helen E Nagels
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Charalampos S Siristatidis
- Assisted Reproduction Unit, 2nd Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ben Kroon
- Queensland Fertility Group Research Foundation, Brisbane, Australia
- The University of Queensland, Brisbane, Australia
| | - Simon McDowell
- Obstetrics and Gynaecology, Capital Coast District Heath Board, Wellington, New Zealand
| |
Collapse
|
10
|
Zhao T, He M, Zhu Z, Zhang T, Zheng W, Qin S, Gao M, Wang W, Chen Z, Han J, Liu L, Zhou B, Wang H, Zhang H, Xia G, Wang J, Wang F, Wang C. P62 promotes FSH-induced antral follicle formation by directing degradation of ubiquitinated WT1. Cell Mol Life Sci 2024; 81:221. [PMID: 38763964 PMCID: PMC11102895 DOI: 10.1007/s00018-024-05251-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/21/2024]
Abstract
In females, the pathophysiological mechanism of poor ovarian response (POR) is not fully understood. Considering the expression level of p62 was significantly reduced in the granulosa cells (GCs) of POR patients, this study focused on identifying the role of the selective autophagy receptor p62 in conducting the effect of follicle-stimulating hormone (FSH) on antral follicles (AFs) formation in female mice. The results showed that p62 in GCs was FSH responsive and that its level increased to a peak and then decreased time-dependently either in ovaries or in GCs after gonadotropin induction in vivo. GC-specific deletion of p62 resulted in subfertility, a significantly reduced number of AFs and irregular estrous cycles, which were same as pathophysiological symptom of POR. By conducting mass spectrum analysis, we found the ubiquitination of proteins was decreased, and autophagic flux was blocked in GCs. Specifically, the level of nonubiquitinated Wilms tumor 1 homolog (WT1), a transcription factor and negative controller of GC differentiation, increased steadily. Co-IP results showed that p62 deletion increased the level of ubiquitin-specific peptidase 5 (USP5), which blocked the ubiquitination of WT1. Furthermore, a joint analysis of RNA-seq and the spatial transcriptome sequencing data showed the expression of steroid metabolic genes and FSH receptors pivotal for GCs differentiation decreased unanimously. Accordingly, the accumulation of WT1 in GCs deficient of p62 decreased steroid hormone levels and reduced FSH responsiveness, while the availability of p62 in GCs simultaneously ensured the degradation of WT1 through the ubiquitin‒proteasome system and autophagolysosomal system. Therefore, p62 in GCs participates in GC differentiation and AF formation in FSH induction by dynamically controlling the degradation of WT1. The findings of the study contributes to further study the pathology of POR.
Collapse
Affiliation(s)
- Ting Zhao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Meina He
- College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou Province, 550025, China
| | - Zijian Zhu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Tuo Zhang
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou Province, 550025, China
| | - Wenying Zheng
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Shaogang Qin
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Meng Gao
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Wenji Wang
- School of Life Science, Taizhou University, Taizhou, 318000, China
| | - Ziqi Chen
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Jun Han
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Longping Liu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Bo Zhou
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, Fujian Province, 361005, China
| | - Hua Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Guoliang Xia
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
- Key Laboratory of Ministry of Education for Conservation and Utilization of Special Biological Resources in the Western China, College of Life Science, Ningxia University, Yinchuan, 750021, China
| | - Jianbin Wang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fengchao Wang
- Transgenic Animal Center, National Institute of Biological Sciences, Beijing, 102206, China
| | - Chao Wang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
- China Agricultural University, No.2 Yuan Ming Yuan West Road, Haidian District, Beijing, 100193, China.
| |
Collapse
|
11
|
Zhang S, Wei Y, Gao X, Song Y, Huang Y, Jiang Q. Unveiling the Ovarian Cell Characteristics and Molecular Mechanism of Prolificacy in Goats via Single-Nucleus Transcriptomics Data Analysis. Curr Issues Mol Biol 2024; 46:2301-2319. [PMID: 38534763 DOI: 10.3390/cimb46030147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/29/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
Increases in litter size, which are influenced by ovulation, are responsible for between 74% and 96% of the economic value of genetic progress, which influences selection. For the selection and breeding of highly prolific goats, genetic mechanisms underlying variations in litter size should be elucidated. Here, we used single-nucleus RNA sequencing to analyze 44,605 single nuclei from the ovaries of polytocous and monotocous goats during the follicular phase. Utilizing known reference marker genes, we identified 10 ovarian cell types characterized by distinct gene expression profiles, transcription factor networks, and reciprocal interaction signatures. An in-depth analysis of the granulosa cells revealed three subtypes exhibiting distinct gene expression patterns and dynamic regulatory mechanisms. Further investigation of cell-type-specific prolificacy-associated transcriptional changes elucidated that "downregulation of apoptosis", "increased anabolism", and "upstream responsiveness to hormonal stimulation" are associated with prolificacy. This study provides a comprehensive understanding of the cell-type-specific mechanisms and regulatory networks in the goat ovary, providing insights into the molecular mechanisms underlying goat prolificacy. These findings establish a vital foundation for furthering understanding of the molecular mechanisms governing folliculogenesis and for improving the litter size in goats via molecular design breeding.
Collapse
Affiliation(s)
- Sanbao Zhang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| | - Yirong Wei
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| | - Xiaotong Gao
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| | - Ying Song
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| | - Yanna Huang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| | - Qinyang Jiang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning 530004, China
| |
Collapse
|
12
|
Hugues JN. Subtle perturbations of ovarian steroidogenesis in patients classified as Poseidon Group 3. Which consequences for therapeutic strategy? Front Endocrinol (Lausanne) 2024; 15:1231585. [PMID: 38384970 PMCID: PMC10879926 DOI: 10.3389/fendo.2024.1231585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 01/09/2024] [Indexed: 02/23/2024] Open
Abstract
The optimal strategy for stimulation of young women with a low ovarian reserve is still a challenging issue because the physio-pathogeny of this disorder is often unknown. As androgen production by the ovary plays a crucial role in folliculogenesis, it was tempting to speculate that subtle perturbations in ovarian steroidogenesis might participate to the low responsiveness to gonadotrophins. Indeed, in vitro analysis of human luteinized granulosa cells has recently provided evidence for some enzymatic deficits in steroidogenesis and altered response to gonadotrophins. Therefore, improving androgen environment of women classified in Poseidon Group 3 should be considered. In this clinical situation, the potential benefit of androgen supplementation or stimulation of theca cells by LH-activity products are respectively discussed.
Collapse
|
13
|
Shah D, Jirge PR. Anti-Mullerian Hormone and Fertility Treatment Decisions in Polycystic Ovary Syndrome: A Literature Review. J Hum Reprod Sci 2024; 17:16-24. [PMID: 38665612 PMCID: PMC11041323 DOI: 10.4103/jhrs.jhrs_153_23] [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: 11/12/2023] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 04/28/2024] Open
Abstract
Anti-Mullerian hormone is a robust marker of ovarian reserve and ovarian response in in vitro fertilisation (IVF). However, its role extends beyond improving the safety of IVF by aiding in choosing appropriate protocols and dosing. This review looks at the value of pre-treatment anti-Mullerian hormone (AMH) value in choosing the appropriate modality of treatment and its predictive ability for the outcomes of such treatment. It briefly addresses the factors that may modulate AMH levels and make clinical decision-making challenging.
Collapse
Affiliation(s)
- Duru Shah
- Gynaecworld, The Centre for Women’s Health and Fertility, Mumbai, Maharashtra, India
| | - Padma Rekha Jirge
- Sushrut Assisted Conception Clinic, Shreyas Hospital, Kolhapur, Maharashtra, India
| |
Collapse
|
14
|
Vann K, Weidner AE, Walczyk AC, Astapova O. Paxillin knockout in mouse granulosa cells increases fecundity†. Biol Reprod 2023; 109:669-683. [PMID: 37552051 PMCID: PMC10651069 DOI: 10.1093/biolre/ioad093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/29/2023] [Accepted: 08/02/2023] [Indexed: 08/09/2023] Open
Abstract
Paxillin is an intracellular adaptor protein involved in focal adhesions, cell response to stress, steroid signaling, and apoptosis in reproductive tissues. To investigate the role of paxillin in granulosa cells, we created a granulosa-specific paxillin knockout mouse model using Cre recombinase driven by the Anti-Müllerian hormone receptor 2 gene promoter. Female granulosa-specific paxillin knockout mice demonstrated increased fertility in later reproductive age, resulting in higher number of offspring when bred continuously up to 26 weeks of age. This was not due to increased numbers of estrous cycles, ovulated oocytes per cycle, or pups per litter, but this was due to shorter time to pregnancy and increased number of litters in the granulosa-specific paxillin knockout mice. The number of ovarian follicles was not significantly affected by the knockout at 30 weeks of age. Granulosa-specific paxillin knockout mice had slightly altered estrous cycles but no difference in circulating reproductive hormone levels. Knockout of paxillin using clustered regularly interspaced short palindromic repeat-associated protein 9 (CRISPR-Cas9) in human granulosa-derived immortalized KGN cells did not affect cell proliferation or migration. However, in cultured primary mouse granulosa cells, paxillin knockout reduced cell death under basal culture conditions. We conclude that paxillin knockout in granulosa cells increases female fecundity in older reproductive age mice, possibly by reducing granulosa cell death. This study implicates paxillin and its signaling network as potential granulosa cell targets in the management of age-related subfertility.
Collapse
Affiliation(s)
- Kenji Vann
- Division of Endocrinology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Adelaide E Weidner
- Division of Endocrinology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Ariana C Walczyk
- Division of Endocrinology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Olga Astapova
- Division of Endocrinology, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
15
|
Orvieto R. Stop GnRH-agonist/GnRH-antagonist protocol: a different insight on ovarian stimulation for IVF. Reprod Biol Endocrinol 2023; 21:13. [PMID: 36710334 PMCID: PMC9885692 DOI: 10.1186/s12958-023-01069-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Ovarian stimulation (OS) is one of the key factors in the success of in vitro fertilization-embryo transfer (IVF-ET), by enabling the recruitment of numerous healthy fertilizable oocytes and, thereby, multiple embryos. The Stop GnRH-agonist/GnRH-antagonist (GnRH-ag/GnRH-ant), which offers all the advantages of using long suppressive GnRH-ag, with GnRH-ant, is in my opinion a valuable addition to the armamentarium of OS protocols. It allows cycle programming, better follicular synchronization and offers successful outcome in a variety of challenging cases such as poor responders, Poseidon group 4 poor prognosis patients, those with elevated peak progesterone (P) serum levels, poor embryo quality or repeated IVF failures.
Collapse
Affiliation(s)
- Raoul Orvieto
- Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Ramat Gan, Israel.
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel.
- The Tarnesby-Tarnowski Chair for Family Planning and Fertility Regulation, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel.
| |
Collapse
|
16
|
Yang W, Chen X, Liu Z, Zhao Y, Chen Y, Geng Z. Integrated transcriptome and proteome revealed that the declined expression of cell cycle-related genes associated with follicular atresia in geese. BMC Genomics 2023; 24:24. [PMID: 36647001 PMCID: PMC9843891 DOI: 10.1186/s12864-022-09088-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 12/16/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Geese exhibit relatively low reproductive performance, and follicular atresia is an important factor that restricts the egg production of geese. Systematic analysis of the regulation of follicle atresia in geese through transcriptome and proteome levels could provide meaningful information on clarifying the mechanism of follicle atresia in poultry. RESULT The granulosa cell layer was loose, disintegrated and showed apoptosis in atretic follicles and remained intact in normal follicles. The hormone levels of FSH and LH were significantly decreased in the atresia follicles compared to the normal follicles (P < 0.05). A total of 954 differentially expressed genes (DEGs, 315 increased and 639 decreased) and 161 differentially expressed proteins (DEPs, 61 increased and 100 decreased) were obtained in atresia follicles compared to normal follicles, of which, 15 genes were differentially expressed in both transcriptome and proteome. The DEGs were mainly enriched in sodium transmembrane transport, plasma membrane, and transmembrane transporter activity based on the GO enrichment analysis and in the cell cycle pathway based on the KEGG enrichment analysis. The DEPs were mainly enriched in localization, lysosome, and phospholipid-binding based on the GO enrichment analysis. Candidate genes Smad2/3, Smad4, Annexin A1 (ANXA1), Stromelysin-1 (MMP3), Serine/threonine-protein kinase (CHK1), DNA replication licensing factor (MCM3), Cyclin-A2 (CCNA2), mitotic spindle assembly checkpoint protein (MAD2), Cyclin-dependent kinase 1 (CDK1), fibroblast growth factor 12 (FGF12), and G1/S-specific cyclin-D1 (CCND1) were possibly responsible for the regulation of atresia. CONCLUSION The cell cycle is an important pathway for the regulation of follicular atresia. Sodium outflow and high expression of MMP3 and MMP9 could be responsible for structural destruction and apoptosis of follicular cells.
Collapse
Affiliation(s)
- Wanli Yang
- grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036 China
| | - Xingyong Chen
- grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036 China ,grid.411389.60000 0004 1760 4804Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, NO. 130 Changjiang West Rd, Hefei, 230036 China
| | - Zhengquan Liu
- grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036 China
| | - Yutong Zhao
- grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036 China
| | - Yufei Chen
- grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036 China
| | - Zhaoyu Geng
- grid.411389.60000 0004 1760 4804College of Animal Science and Technology, Anhui Agricultural University, No. 130 Changjiang West Road, Hefei, 230036 China ,grid.411389.60000 0004 1760 4804Anhui Province Key Laboratory of Local Livestock and Poultry Genetic Resource Conservation and Bio-breeding, Anhui Agricultural University, NO. 130 Changjiang West Rd, Hefei, 230036 China
| |
Collapse
|
17
|
Higher live birth rate following transdermal testosterone pretreatment in poor responders: a systematic review and meta-analysis. Reprod Biomed Online 2023; 46:81-91. [PMID: 36369150 DOI: 10.1016/j.rbmo.2022.09.022] [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: 07/03/2022] [Revised: 09/08/2022] [Accepted: 09/26/2022] [Indexed: 11/05/2022]
Abstract
A systematic review and meta-analysis was performed aiming to identify good-quality randomized controlled trials (RCT) evaluating testosterone pretreatment in poor responders. Eight RCTs were analysed, evaluating 797 women. Transdermal testosterone gel was used in all studies, with a dose ranging from 10 to 12.5 mg/day for 10-56 days. The main outcome measure was achievement of pregnancy, expressed as clinical pregnancy or live birth. Testosterone pretreatment was associated with a significantly higher live birth (risk ratio [RR] 2.07, 95% confidence interval [CI] 1.09-3.92) and clinical pregnancy rate (RR 2.25, 95% CI 1.54-3.30), as well as a significant increase in the number of cumulus-oocyte complexes retrieved. Significantly fewer days to complete ovarian stimulation, a lower total dose of gonadotrophins, a lower cancellation rate due to poor ovarian response and a thicker endometrium on the day of triggering of final oocyte maturation were observed. No significant differences were observed in oestradiol concentration, the numbers of follicles ≥17 mm, metaphase II oocytes, two-pronuclear oocytes and embryos transferred, and the proportion of patients with embryo transfer. The current study suggests that the probability of pregnancy is increased in poor responders pretreated with transdermal testosterone who are undergoing ovarian stimulation for IVF.
Collapse
|
18
|
Devillers MM, François CM, Chester M, Corre R, Cluzet V, Giton F, Cohen-Tannoudji J, Guigon CJ. Androgen receptor signaling regulates follicular growth and steroidogenesis in interaction with gonadotropins in the ovary during mini-puberty in mice. Front Endocrinol (Lausanne) 2023; 14:1130681. [PMID: 37152943 PMCID: PMC10154677 DOI: 10.3389/fendo.2023.1130681] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
In females, androgens contribute to ovarian diseases such as polycystic ovarian syndrome (PCOS), but their action is also crucial for ovarian physiology, i.e., follicular growth and estradiol (E2) synthesis during reproductive life, in interaction with the gonadotropins LH and FSH. However, it is unclear whether androgens already play a role in the ovary at mini-puberty, a phase of postnatal development with active follicular growth and high E2 levels. Therefore, we analyzed the potential actions of androgens on the ovary and their possible interaction with gonadotropins during this period in mice. We used molecular-based studies and pharmacological approaches in vivo and on cultured ovaries. We found that mini-pubertal ovaries produce significant amounts of testosterone and display androgen receptor (AR) expression in growing follicles, both under the control of LH. By blocking AR signaling either in vivo or in ovarian cultures, we found that this pathway may participate in the regulation of prepubertal E2 synthesis and follicular growth, possibly by regulating the expression of a number of key intra-ovarian regulators, including FSH receptor (Fshr), the aromatase enzyme converting androgens into estrogens (Cyp19a1) and the cell cycle inhibitor p27KIP1 (Cdkn1b). We further showed that AR may stimulate FSH-mediated regulation of Cyp19a1 through its action on Fshr mRNA abundance. Overall, this work supports the idea that AR signaling is already activated in mini-pubertal ovaries to regulate E2 synthesis and follicular growth, at the interplay with LH and FSH signaling. Its early action may, thus, contribute to the implementation of early ovarian function with possible impacts on reproductive function.
Collapse
Affiliation(s)
- Marie M. Devillers
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Charlotte M. François
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Mélanie Chester
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Raphaël Corre
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Victoria Cluzet
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Frank Giton
- AP-HP, Pôle biologie-Pathologie Henri Mondor, Inserm IMRB U955, Créteil, France
| | - Joëlle Cohen-Tannoudji
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
| | - Céline J. Guigon
- Université Paris-Cité, CNRS, Inserm, Biologie Fonctionnelle et Adaptative, Paris, France
- *Correspondence: Céline J. Guigon,
| |
Collapse
|
19
|
Kasuga-Yamashita F, Baba T, Nagao S, Fujibe Y, Morishita M, Kuno Y, Mariya T, Honnma H, Endo T, Kiya T, Saito T. Letrozole increases preantral follicle growth and decreases estradiol production without impairing follicle survival. J Ovarian Res 2022; 15:136. [PMID: 36564850 PMCID: PMC9789635 DOI: 10.1186/s13048-022-01073-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/08/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Letrozole has been reported to be effective in treating anovulation, preventing ovarian hyperstimulation syndrome (OHSS), and retrieving oocytes in breast cancer patients. However, the role and mechanism of letrozole in follicular development remain unclear. RESULTS We treated mouse preantral follicles with various treatments; we found no significant difference in follicle survival rates in the letrozole (LET) group compared with the control group, but the average diameter of follicles in the LET group tended to be larger (CTRL vs. LET 30, p = 0.064; CTRL vs. LET 100, p = 0.025). The estradiol concentrations in culture media of the LET group were significantly lower than those observed in the control group (CTRL vs. LET 30, p = 0.038; CTRL vs. LET 100, p = 0.025). We further found a marked increase in follicle-stimulating hormone receptor (FSHR) gene expression in response to letrozole treatment (CTRL vs. LET 30, p = 0.075; CTRL vs. LET 100, p = 0.034). This result suggested that increased FSHR expression promotes follicle development. Letrozole inhibited aromatase activity, but the effect was limited. Letrozole did not significantly reduce vascular endothelial growth factor (VEGF) gene expression. CONCLUSIONS Letrozole may promote follicle development by increasing the expression of FSHR. Letrozole may be useful for fertility preservation of patients with estrogen-dependent cancers such as breast cancer and various other cancers. Whether letrozole has a direct effect in reducing OHSS requires further investigation.
Collapse
Affiliation(s)
- Fukiko Kasuga-Yamashita
- grid.263171.00000 0001 0691 0855Department of Obstetrics and Gynecology, Sapporo Medical University, South 1 West 16, 060-8543 Sapporo, Hokkaido Japan
| | - Tsuyoshi Baba
- grid.263171.00000 0001 0691 0855Department of Obstetrics and Gynecology, Sapporo Medical University, South 1 West 16, 060-8543 Sapporo, Hokkaido Japan
| | - Sachiko Nagao
- grid.263171.00000 0001 0691 0855Department of Obstetrics and Gynecology, Sapporo Medical University, South 1 West 16, 060-8543 Sapporo, Hokkaido Japan
| | - Yuya Fujibe
- grid.263171.00000 0001 0691 0855Department of Obstetrics and Gynecology, Sapporo Medical University, South 1 West 16, 060-8543 Sapporo, Hokkaido Japan
| | - Miyuki Morishita
- grid.263171.00000 0001 0691 0855Department of Obstetrics and Gynecology, Sapporo Medical University, South 1 West 16, 060-8543 Sapporo, Hokkaido Japan
| | - Yoshika Kuno
- grid.263171.00000 0001 0691 0855Department of Obstetrics and Gynecology, Sapporo Medical University, South 1 West 16, 060-8543 Sapporo, Hokkaido Japan
| | - Tasuku Mariya
- grid.263171.00000 0001 0691 0855Department of Obstetrics and Gynecology, Sapporo Medical University, South 1 West 16, 060-8543 Sapporo, Hokkaido Japan
| | - Hiroyuki Honnma
- Sapporo ART Clinic, 1-2 North 7 West 4, 060-0807 Sapporo, Hokkaido Japan
| | - Toshiaki Endo
- grid.263171.00000 0001 0691 0855Department of Obstetrics and Gynecology, Sapporo Medical University, South 1 West 16, 060-8543 Sapporo, Hokkaido Japan
| | - Tamotsu Kiya
- Ena Asabu ART Clinic, 2-2-7 Asabu, 001-0045 Sapporo, Hokkaido Japan
| | - Tsuyoshi Saito
- grid.263171.00000 0001 0691 0855Department of Obstetrics and Gynecology, Sapporo Medical University, South 1 West 16, 060-8543 Sapporo, Hokkaido Japan
| |
Collapse
|
20
|
Esencan E, Beroukhim G, Seifer DB. Age-related changes in Folliculogenesis and potential modifiers to improve fertility outcomes - A narrative review. Reprod Biol Endocrinol 2022; 20:156. [PMID: 36397149 PMCID: PMC9670479 DOI: 10.1186/s12958-022-01033-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/06/2022] [Indexed: 11/19/2022] Open
Abstract
Reproductive aging is characterized by a decline in oocyte quantity and quality, which is directly associated with a decline in reproductive potential, as well as poorer reproductive success and obstetrical outcomes. As women delay childbearing, understanding the mechanisms of ovarian aging and follicular depletion have become increasingly more relevant. Age-related meiotic errors in oocytes are well established. In addition, it is also important to understand how intraovarian regulators change with aging and how certain treatments can mitigate the impact of aging. Individual studies have demonstrated that reproductive pathways involving antimullerian hormone (AMH), vascular endothelial growth factor (VEGF), neurotropins, insulin-like growth factor 1 (IGF1), and mitochondrial function are pivotal for healthy oocyte and cumulus cell development and are altered with increasing age. We provide a comprehensive review of these individual studies and explain how these factors change in oocytes, cumulus cells, and follicular fluid. We also summarize how modifiers of folliculogenesis, such as vitamin D, coenzyme Q, and dehydroepiandrosterone (DHEA) may be used to potentially overcome age-related changes and enhance fertility outcomes of aged follicles, as evidenced by human and rodent studies.
Collapse
Affiliation(s)
- Ecem Esencan
- Yale School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, New Haven, CT, USA.
| | - Gabriela Beroukhim
- Yale School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, New Haven, CT, USA
| | - David B Seifer
- Yale School of Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, New Haven, CT, USA
| |
Collapse
|
21
|
Ezoe K, Fukuda J, Takeshima K, Shinohara K, Kato K. Letrozole-induced endometrial preparation improved the pregnancy outcomes after frozen blastocyst transfer compared to the natural cycle: a retrospective cohort study. BMC Pregnancy Childbirth 2022; 22:824. [PMID: 36344952 PMCID: PMC9639274 DOI: 10.1186/s12884-022-05174-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
Abstract
Background Letrozole treatment is considered an effective option in endometrial preparation for frozen embryo transfers in patients with ovulation disorders or irregular menstruation; however, the effectiveness of letrozole-induced endometrial preparation remains unclear in ovulatory patients. Furthermore, there is no comparative study reporting on pregnancy complications and congenital anomalies after frozen embryo transfers comparing natural and letrozole-assisted cycles. This study examined whether letrozole-induced endometrial preparation affected pregnancy outcomes, perinatal outcomes, and congenital anomalies after single vitrified-warmed blastocyst transfers (SVBTs) in ovulatory patients, as compared with the natural cycle. Methods This historic cohort study included only patients with unexplained infertility. Overall, 14,611 patients who underwent SVBTs between July 2015 and June 2020, comprising both natural and letrozole-assisted cycles, were included. Multiple covariates that impact outcomes were used for propensity score matching; 1,911 patients in the letrozole group were matched to 12,700 patients in the natural group, and the clinical records of 1,910 patients in each group were retrospectively analysed. Cycle characteristics, pregnancy outcomes (clinical pregnancy, ongoing pregnancy, and live birth), and incidence of pregnancy complications and congenital anomalies were statistically compared between the two groups. Results Multivariate logistic regression analysis showed that letrozole administration during SVBT cycles significantly improved the live birth rate (P = 0.0355). Gestational age, birth length, birth weight, and infant sex, as well as the incidence of pregnancy complications and birth defects, were statistically comparable between the two groups. Furthermore, multivariate logistic regression analysis revealed that the perinatal outcomes were not affected by letrozole-induced endometrial preparation. Conclusions Letrozole-induced endometrial preparation improved the live birth rate compared with the natural cycle, without adverse effects on perinatal outcomes and congenital anomalies after SVBTs. Therefore, letrozole-induced endometrial preparation might be a safe and more effective strategy, especially for patients with insufficient luteal function. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-05174-0.
Collapse
Affiliation(s)
- Kenji Ezoe
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023 Japan
| | - Junichiro Fukuda
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023 Japan
| | - Kazumi Takeshima
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023 Japan
| | - Kazunori Shinohara
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023 Japan
| | - Keiichi Kato
- Kato Ladies Clinic, 7-20-3 Nishishinjuku, Shinjuku-Ku, Tokyo, 160-0023 Japan
| |
Collapse
|
22
|
Bhattacharya K, Saha I, Sen D, Bose C, Chaudhuri GR, Dutta S, Sengupta P, Bhattacharya S, Barman SS, Syamal AK. Role of anti-Mullerian hormone in polycystic ovary syndrome. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2022. [DOI: 10.1186/s43043-022-00123-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractPolycystic ovary syndrome (PCOS) is the most common gynecological endocrine disorders affecting up to 10% of all females in their reproductive age, and its cause of onset is still elusive. A spectrum of recent research reflected diverse associations between increased plasma level of anti-Mullerian hormone (AMH) and different clinical features of PCOS. Since AMH levels reflect the pool of growing follicles that potentially can ovulate, it can be stated that serum AMH levels can be used to assess the “functional ovarian reserve,” rather mentioning it as the “ovarian reserve.” AMH also appears to be a premier endocrine parameter for the assessment of atrophied ovarian follicular pool in response to age of individuals. AMH hinders the follicular development as well as the follicular recruitment and ultimately resulting in follicular arrest which is the key pathophysiologic condition for the onset of PCOS. Furthermore, FSH-induced aromatase activity remains inhibited by AMH that aids emergence of other associated clinical signs of PCOS, such as excess androgen, followed by insulin resistance among the PCOS individuals. Given the versatile association of AMH with PCOS and scarcity in literature explaining the underling mechanisms how AMH relates with PCOS, this review article will discuss the roles of AMH in the pathogenesis of PCOS which may introduce a new era in treatment approach of PCOS.
Collapse
|
23
|
AOP key event relationship report: Linking decreased androgen receptor activation with decreased granulosa cell proliferation of gonadotropin-independent follicles. Reprod Toxicol 2022; 112:136-147. [PMID: 35868514 DOI: 10.1016/j.reprotox.2022.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 02/08/2023]
Abstract
We recently proposed to formally recognize Key Event Relationships (KERs) as building blocks of Adverse Outcome Pathways (AOPs) that can be independently developed and peer-reviewed. Here, we follow this approach and provide an independent KER from AOP345, which describes androgen receptor (AR) antagonism leading to decreased female fertility. This KER connects AR antagonism to reduced granulosa cell proliferation of gonadotropin-independent follicles (KER2273). We have developed both the KER and the two adjacent Key Events (KEs). A systematic approach was used to ensure that all relevant supporting evidence for KER2273 was retrieved. Supporting evidence for the KER highlights the importance of AR action during the early stages of follicular development. Both biological plausibility and empirical evidence are presented, with the latter also assessed for quality. We believe that tackling isolated KERs instead of whole AOPs will accelerate the AOP development. Faster AOP development will lead to the development of simple test methods that will aid screening of chemicals, endocrine disruptor identification, risk assessment, and subsequent regulation.
Collapse
|
24
|
Sex Steroid Receptors in Polycystic Ovary Syndrome and Endometriosis: Insights from Laboratory Studies to Clinical Trials. Biomedicines 2022; 10:biomedicines10071705. [PMID: 35885010 PMCID: PMC9312843 DOI: 10.3390/biomedicines10071705] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 12/13/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) and endometriosis are reproductive disorders that may cause infertility. The pathology of both diseases has been suggested to be associated with sex steroid hormone receptors, including oestrogen receptors (ER), progesterone receptors (PRs) and androgen receptors (ARs). Therefore, with this review, we aim to provide an update on the available knowledge of these receptors and how their interactions contribute to the pathogenesis of PCOS and endometriosis. One of the main PCOS-related medical conditions is abnormal folliculogenesis, which is associated with the downregulation of ER and AR expression in the ovaries. In addition, metabolic disorders in PCOS are caused by dysregulation of sex steroid hormone receptor expression. Furthermore, endometriosis is related to the upregulation of ER and the downregulation of PR expression. These receptors may serve as therapeutic targets for the treatment of PCOS-related disorders and endometriosis, considering their pathophysiological roles. Receptor agonists may be applied to increase the expression of a specific receptor and treat endometriosis or metabolic disorders. In contrast, receptor antagonist functions to reduce receptor expression and can be used to treat endometriosis and induce ovulation. Understanding PCOS and the pathological roles of endometriosis sex steroid receptors is crucial for developing potential therapeutic strategies to treat infertility in both conditions. Therefore, research should be continued to fill the knowledge gap regarding the subject.
Collapse
|
25
|
Poulsen LC, Warzecha AK, Bülow NS, Bungum L, Macklon NS, Yding Andersen C, Skouby SO. Effects of letrozole cotreatment on endocrinology and follicle development in women undergoing ovarian stimulation in an antagonist protocol. Hum Reprod 2022; 37:1557-1571. [PMID: 35652260 DOI: 10.1093/humrep/deac119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 04/17/2022] [Indexed: 12/25/2022] Open
Abstract
STUDY QUESTION What are the downstream endocrine and paracrine consequences of letrozole (LZ) cotreatment during ovarian stimulation and is follicle growth and recruitment affected? SUMMARY ANSWER Letrozole cotreatment induces marked changes in both the follicular and luteal phase endocrinology causing potentiation of follicle diameter and an improved corpus luteum function without affecting the secondarily recruited follicle cohort. WHAT IS KNOWN ALREADY Letrozole is a third-generation aromatase inhibitor that is well-established as an effective ovulatory agent, while its possible benefits in standard in vitro fertilization protocols are less thoroughly investigated. STUDY DESIGN, SIZE, DURATION This study included a double-blinded, placebo-controlled, randomized study with LZ or placebo intervention during ovarian stimulation for IVF treatment, an observational preceding baseline natural cycle and a succeeding follow-up visit. Participants were enrolled between August 2016 and November 2018. Data from the randomized, stimulated cycle were part of a larger RCT, which was previously published. PARTICIPANTS/MATERIALS, SETTING, METHODS The study was conducted at a public fertility clinic at Herlev Hospital, Denmark, including 31 healthy, normo-responding women eligible for IVF treatment. They underwent a natural baseline cycle and were subsequently randomized to receive either LZ 5 mg (n = 16) or placebo (n = 15) daily during ovarian stimulation from cycle day (CD) 2-3 until induction of ovulation. Throughout both cycles, monitoring was performed every third day with transvaginal ultrasound for assessment of follicle count and diameter, and blood analyses for the determination of twelve endocrine and paracrine parameters. A follow-up assessment was performed at CD2-3 in the succeeding cycle. In the randomized part of the study, we determined differences in blood parameters, follicle recruitment, and follicle diameter. In the observational part of the study, we assessed follicle recruitment in between cycles and its correlation to endocrine parameters. MAIN RESULTS AND THE ROLE OF CHANCE Letrozole cotreatment significantly suppressed oestradiol (E2) concentrations in the follicular phase (area under the curve (AUC) -58% (95% CI [-70%; -43%], P < 0.001)) and luteal phase (AUC -39% [-63%; -1%], P = 0.046). This had a marked effect on the endocrine and paracrine output with increased follicular phase luteinizing hormone (AUC +37% [3%; 82%], P = 0.033), androstenedione (AUC +36% [6%; 74%], P = 0.016), testosterone (AUC +37% [7%; 73%], P = 0.013) and 17-OH-progesterone (AUC +114% [10%; 318%], P = 0.027). Furthermore, follicle-stimulating hormone (FSH) was increased at stimulation day 5 in the LZ group (P < 0.05). In the luteal phase, increased corpus luteum output was reflected by elevated progesterone (AUC +44% [1%; 104%], P = 0.043), inhibin A (AUC +52% [11%; 108%], P = 0.011), androstenedione (AUC +31% [9%; 58%], P = 0.006) and testosterone (AUC +29% [6%; 57%], P = 0.012) in the LZ group. The altered balance between oestrogens and androgens was reflected in a markedly reduced SHBG concentration in the LZ group throughout the luteal phase (AUC -35% [-52%; -11%], P = 0.009). Endocrine and paracrine parameters were similar between groups at the follow-up visit. Letrozole cotreatment significantly increased the mean number of follicles >16 mm at oocyte retrieval (7.2 vs 5.2, difference: 2.0, 95% CI [0.1; 3.8], P = 0.036), while the mean total number of follicles at oocyte retrieval was the same (23.7 vs 23.5, difference: 0.2 [-5.8; 6.1], P = 0.958), and the mean FSH consumption during the stimulated cycle was similar (1500 vs 1520 IU, difference -20 IU [-175; 136], P = 0.794). Between cycles, the mean antral follicle count at CD2-3 was unchanged (natural cycle 19.0, stimulated cycle 20.9, follow-up cycle 19.7, P = 0.692) and there was no effect of LZ cotreatment on the recruitment of the next follicle cohort (test for interaction, P = 0.821). LIMITATIONS, REASONS FOR CAUTION This study included a relatively small, selected group of healthy women with an expected normal ovarian function and reserve, and the effects of LZ may therefore be different in other patient groups. WIDER IMPLICATIONS OF THE FINDINGS We confirm some previous findings concerning increased follicle growth and increased endogenous FSH and androgen production, which support the rationale for further studies on the use of LZ cotreatment, for example, as a form of endogenous androgen priming sensitizing the follicle to FSH. Letrozole appears to improve the luteal phase with better stimulation of corpus luteum and progesterone secretion. STUDY FUNDING/COMPETING INTEREST(S) The authors declare no conflicts of interest relating to the present work. TRIAL REGISTRATION NUMBER NCT02939898.
Collapse
Affiliation(s)
- Liv C Poulsen
- Department of Gynaecology and Obstetrics, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Denmark
| | - Agnieszka K Warzecha
- Department of Gynaecology and Obstetrics, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Denmark
| | - Nathalie S Bülow
- Department of Gynaecology and Obstetrics, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Denmark.,The Fertility Department, Copenhagen University Hospital, Copenhagen, Denmark
| | - Leif Bungum
- Fertility Clinic, Department of Gynaecology and Obstetrics, Zealand University Hospital, Køge, Denmark
| | - Nicholas S Macklon
- Fertility Clinic, Department of Gynaecology and Obstetrics, Zealand University Hospital, Køge, Denmark.,London Women's Clinic, London, UK
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, The Juliane Marie Centre for Women, Children and Reproduction, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sven O Skouby
- Department of Gynaecology and Obstetrics, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Denmark
| |
Collapse
|
26
|
Hsu CC, Hsu I, Lee LH, Hsu R, Hsueh YS, Lin CY, Chang HH. Ovarian Follicular Growth through Intermittent Vaginal Gonadotropin Administration in Diminished Ovarian Reserve Women. Pharmaceutics 2022; 14:pharmaceutics14040869. [PMID: 35456706 PMCID: PMC9025251 DOI: 10.3390/pharmaceutics14040869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/10/2022] [Accepted: 04/13/2022] [Indexed: 02/05/2023] Open
Abstract
It is a challenge to obtain enough oocytes during in vitro fertilization (IVF) in women who have a poor ovarian response (POR) in achieving conception. We have adopted the characteristics of the first uterine pass effect, which we pioneered in employing the vaginal administration of gonadotropins in women receiving IVF treatments. In our previous study employing vaginal administration, faster absorption and slower elimination of gonadotropins were demonstrated, and, female subjects presented proper ovarian follicle growth and pregnancy rates. In this study, during 2016–2020, 300 to 675 IU of gonadotropins were administered vaginally every three days in 266 POR women for their controlled ovarian hyperstimulation (COH). The injections were performed with needles angled at 15–30° towards the middle-upper portions of the bilateral vaginal wall, with an injection depth of 1–2 mm. For the COH results, these women, on average, received 3.0 ± 0.9 vaginal injections and a total dose of 1318.4 ± 634.4 IU gonadotropins, resulting in 2.2 ± 1.9 mature oocytes and 1.0 ± 1.2 good embryos. Among these embryos, 0.9 ± 1.0 were transferred to reach a clinical pregnancy rate of 18.1% and a live birth rate of 16.7%. In conclusion, the intermittent vaginal administration of gonadotropins proved to be effective in POR women for their IVF treatments.
Collapse
Affiliation(s)
- Chao-Chin Hsu
- Taiwan United Birth-Promoting Experts Fertility Clinic, Tainan 710, Taiwan
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 104, Taiwan;
- Department of Obstetrics and Gynecology, National Cheng Kung University Hospital, Tainan 701, Taiwan
- Correspondence: (C.-C.H.); (H.H.C.); Tel.: +886-6-2353535 (ext. 5683) (H.H.C.)
| | - Isabel Hsu
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei 104, Taiwan;
| | | | - Rosie Hsu
- Department of Pediatrics, National Taiwan University Hospital, Taipei 104, Taiwan;
| | - Yuan-Shuo Hsueh
- Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, Tainan 711, Taiwan;
| | - Chih-Ying Lin
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
| | - Hui Hua Chang
- Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
- School of Pharmacy, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Department of Pharmacy, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Department of Pharmacy, National Cheng Kung University Hospital, Dou-Liou Branch, Yunlin 640, Taiwan
- Correspondence: (C.-C.H.); (H.H.C.); Tel.: +886-6-2353535 (ext. 5683) (H.H.C.)
| |
Collapse
|
27
|
Neves AR, Montoya-Botero P, Polyzos NP. Androgens and diminished ovarian reserve: the long road from basic science to clinical implementation. A comprehensive and systematic review with meta-analysis. Am J Obstet Gynecol 2022; 227:401-413.e18. [PMID: 35364061 DOI: 10.1016/j.ajog.2022.03.051] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 03/16/2022] [Accepted: 03/24/2022] [Indexed: 11/28/2022]
Abstract
OBJECTIVE This study aimed to present a narrative review regarding androgen production, androgens' role in folliculogenesis, and the available therapeutic approaches for androgen supplementation, and to perform a systematic review and meta-analysis regarding the impact of androgens (dehydroepiandrosterone/testosterone) compared with placebo or no treatment on ovarian response and pregnancy outcomes in patients with diminished ovarian reserve and/or poor ovarian responders. DATA SOURCES An electronic search of MEDLINE, Embase, Cochrane Library, Cochrane Central Register of Controlled Trials, Scopus, ClinicalTrials.gov, the ISRCTN registry, and the World Health Organization International Clinical Trials Registry, was conducted for studies published until September 2021. STUDY ELIGIBILITY CRITERIA Randomized controlled trials that compared ovarian response and/or pregnancy outcomes between the different in vitro fertilization protocols using androgens (ie, dehydroepiandrosterone and testosterone) and conventional in vitro fertilization stimulation in patients with diminished ovarian reserve and/or poor ovarian responders were included. METHODS The quality of each study was evaluated with the revised Cochrane risk-of-bias tool for randomized trials (RoB 2). The meta-analysis used random-effects models. All results were interpreted on the basis of intention-to-treat analysis (defined as the inclusion of all randomized patients in the denominator). Risk ratios and 95% confidence intervals were used and combined for meta-analysis. RESULTS No significant differences were found regarding the number of oocytes retrieved (mean difference, 0.76; 95% confidence interval, -0.35 to 1.88), mature oocytes retrieved (mean difference, 0.25; 95% confidence interval, -0.27 to 0.76), clinical pregnancy rate (risk ratio, 1.17; 95% confidence interval, 0.87-1.57), live-birth rate (risk ratio, 0.97; 95% confidence interval, 0.47-2.01), or miscarriage rate (risk ratio, 0.80; 95% confidence interval, 0.29-2.22) when dehydroepiandrosterone priming was compared with placebo or no treatment. Testosterone pretreatment yielded a higher number of oocytes retrieved (mean difference, 0.94; 95% confidence interval, 0.46-1.42), a higher clinical pregnancy rate (risk ratio, 2.07; 95% confidence interval, 1.33-3.20), and higher live-birth rate (risk ratio, 2.09; 95% confidence interval, 1.11-3.95). CONCLUSION Although dehydroepiandrosterone did not present a clear effect on outcomes of assisted reproductive techniques, we found a potentially beneficial effect of testosterone priming on ovarian response and pregnancy outcomes. However, results should be interpreted with caution, taking into account the low to moderate quality of the available evidence.
Collapse
Affiliation(s)
- Ana Raquel Neves
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain; Autonomous University of Barcelona, Cerdanyola del Vallès, Spain
| | - Pedro Montoya-Botero
- Conceptum - Unidad de Fertilidad del Country, Bogotá, Colombia; Department of Epidemiology and Biostatistics, Fundación Universitaria de Ciencias de la Salud - FUCS, Bogotá, Colombia
| | - Nikolaos P Polyzos
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain; Faculty of Medicine and Health Sciences, Ghent University (UZ Gent), Ghent, Belgium.
| |
Collapse
|
28
|
Liu S, Hong L, Mo M, Xiao S, Wang X, Fan X, Zhang S, Diao L, Zeng Y. Association of antimüllerian hormone with polycystic ovarian syndrome phenotypes and pregnancy outcomes of in vitro fertilization cycles with fresh embryo transfer. BMC Pregnancy Childbirth 2022; 22:171. [PMID: 35236324 PMCID: PMC8892693 DOI: 10.1186/s12884-022-04518-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 02/21/2022] [Indexed: 11/15/2022] Open
Abstract
Objective The current study was undertaken to investigate the relationship between antimüllerian hormone (AMH) and polycystic ovarian syndrome (PCOS) phenotypes and to determine whether AMH is associated with pregnancy outcomes in infertile women undergoing their first in vitro fertilization (IVF) treatment. Methods We performed a retrospective cohort study of 2973 infertile women, including 418 women with PCOS undergoing their first IVF treatment at a private fertility center from January 2014 to March 2018. Women were stratified into three groups using cutoffs defined by the 25th and 75th percentiles of the serum AMH level: 746 women had AMH ≤ 2.25 ng/mL; 1486 women had AMH between 2.25 to 5.71 ng/mL; and 741 women had AMH > 5.71 ng/mL. Endocrine characteristics, PCOS phenotypes, stimulation outcomes, pregnancy outcomes were compared among these groups. When there were any statistical differences (P < 0.05) among the three groups, Bonferroni test was performed as post-hoc tests to determine where the statistical differences existed. To assess the relationships between AMH and pregnancy outcomes in total patients and PCOS patients, logistic regression analysis, adjusted for potential confounding covariates, were performed. Results Women with high AMH had greater prevalence of hyperandrogenism (HA), polycystic ovarian morphology (PCOM) and amenorrhea than women with low or average AMH. The clinical pregnancy rate were significantly higher in the high-AMH group compared with low- and average-AMH groups (69.9% vs. 58.8% and 64.7% respectively; P < 0.001). The live birth rate was significantly lower in women with AMH ≤ 2.25 ng/mL compared with average- and high-AMH groups (47.6% vs. 55.2 and 59.5% respectively; P < 0.001). However, after controlling for maternal age, oocyte yield, as well as other confounders, AMH was no longer associated with a higher live birth rate (aOR 1.037, 95% CI 0.853–1.261, P = 0.717; aOR 1.099, 95% CI 0.858–1.408, P = 0.455, respectively) and clinical pregnancy rate (aOR 1.064, 95% CI 0.834–1.359, P = 0.617; aOR 1.181, 95% CI 0.875–1.595, P = 0.276, respectively). Moreover, pregnancy outcomes did not differ in PCOS women according to AMH quartiles. Conclusion Increased AMH levels associated with PCOS severity and greater ovarian stimulation. However, AMH was not associated with clinical pregnancy rate and live birth rate after controlling for other confounders in women undergoing IVF. Thus, AMH should not be used to alter clinical decisions and exclude patients based on a low or even undetectable AMH value. Supplementary Information The online version contains supplementary material available at 10.1186/s12884-022-04518-0.
Collapse
Affiliation(s)
- Su Liu
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, PR China
| | - Ling Hong
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, PR China
| | - Meilan Mo
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, PR China
| | - Shan Xiao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, PR China
| | - Xuejin Wang
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, PR China
| | - Xinfeng Fan
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, PR China
| | - Sainan Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University & Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, PR China
| | - Lianghui Diao
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, PR China
| | - Yong Zeng
- Shenzhen Key Laboratory of Reproductive Immunology for Peri-Implantation, Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, PR China.
| |
Collapse
|
29
|
Lee DS, Knittel T, Deschner T, Heistermann M, Higham JP. Testing the role of testosterone versus estrogens in mediating reproductive transitions in female rhesus macaques. Horm Behav 2022; 139:105123. [PMID: 35149292 DOI: 10.1016/j.yhbeh.2022.105123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 11/16/2022]
Abstract
In male vertebrates, testosterone is generally known to coordinate reproductive trade-offs, in part by promoting the transition to the next reproduction at the expense of current parental care. The role of testosterone in reproductive transitions has been little tested in female vertebrates, especially in mammals. The present study sought to fill this gap, by first undertaking an experimental study, in which we identified DHT, androstenediol, and in particular etiocholanolone, as fecal androgen metabolites which reflect serum testosterone concentration in female rhesus macaques (Macaca mulatta). Using concentrations of fecal etiocholanolone as proxy for circulating testosterone, we then conducted a field study on 46 free-ranging rhesus macaques of Cayo Santiago, Puerto Rico, to test if testosterone mediates the trade-off between reproductive transition (a higher chance of reproducing in the next year) and current reproduction (providing more care to current offspring). While the evidence for testosterone was weak, the testing of fecal immunoreactive estrogen metabolites suggested a potential role of estrogen in reproductive trade-offs. We found large individual differences in fecal etiocholanolone concentrations during the early postpartum period that were unexplained even after accounting for sociodemographic factors such as age and dominance rank. Further investigation is needed to understand this variation. Our study suggests that the actions of testosterone in females may not have evolved to fulfil the same role in primate reproductive transitions as it does in males, and we encourage more studies to consider the function of testosterone in reproductive behaviors and life history transitions in females of mammalian taxa.
Collapse
Affiliation(s)
- D Susie Lee
- Department of Anthropology, New York University, 25 Waverly Place, New York 10003, NY, USA; New York Consortium in Evolutionary Primatology, New York 10024, NY, USA.
| | - Tina Knittel
- Max Planck Institute for Evolutionary Anthropology, Interim Group Primatology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Tobias Deschner
- Max Planck Institute for Evolutionary Anthropology, Interim Group Primatology, Deutscher Platz 6, 04103 Leipzig, Germany
| | - Michael Heistermann
- Endocrinology Laboratory, German Primate Center, Leibniz Institute for Primate Research, Kellnerweg 4, 37077 Göttingen, Germany
| | - James P Higham
- Department of Anthropology, New York University, 25 Waverly Place, New York 10003, NY, USA; New York Consortium in Evolutionary Primatology, New York 10024, NY, USA
| |
Collapse
|
30
|
Pretreatment: Does it improve quantity or quality? Fertil Steril 2022; 117:657-663. [DOI: 10.1016/j.fertnstert.2022.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 11/24/2022]
|
31
|
Priya K, Setty M, Babu UV, Pai KSR. Implications of environmental toxicants on ovarian follicles: how it can adversely affect the female fertility? ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:67925-67939. [PMID: 34628616 PMCID: PMC8718383 DOI: 10.1007/s11356-021-16489-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/07/2021] [Indexed: 05/06/2023]
Abstract
The pool of primordial follicles formed in the ovaries during early development determines the span and quality of fertility in the reproductive life of a woman. As exposure to occupational and environmental toxicants (ETs) has become inevitable, consequences on female fertility need to be established. This review focuses on the ETs, especially well-studied prototypes of the classes endocrine disrupting chemicals (EDCs), heavy metals, agrochemicals, cigarette smoke, certain chemicals used in plastic, cosmetic and sanitary product industries etc that adversely affect the female fertility. Many in vitro, in vivo and epidemiological studies have indicated that these ETs have the potential to affect folliculogenesis and cause reduced fertility in women. Here, we emphasize on four main conditions: polycystic ovary syndrome, primary ovarian insufficiency, multioocytic follicles and meiotic defects including aneuploidies which can be precipitated by ETs. These are considered main causes for reduced female fertility by directly altering the follicular recruitment, development and oocytic meiosis. Although substantial experimental evidence is drawn with respect to the detrimental effects, it is clear that establishing the role of one ET as a risk factor in a single condition is difficult as multiple conditions have common risk factors. Therefore, it is important to consider this as a matter of public and wildlife health.
Collapse
Affiliation(s)
- Keerthi Priya
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Manjunath Setty
- Department of Pharmacognosy, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Uddagiri Venkanna Babu
- Phytochemistry Department, R & D Centre, The Himalaya Drug Company, Makali, Tumkur Road, Bangalore, Karnataka, 562162, India
| | - Karkala Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
32
|
Bülow NS, Holt MD, Skouby SO, Petersen KB, Englund ALM, Pinborg A, Macklon NS. Co-treatment with letrozole during ovarian stimulation for IVF/ICSI: a systematic review and meta-analysis. Reprod Biomed Online 2021; 44:717-736. [DOI: 10.1016/j.rbmo.2021.12.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022]
|
33
|
Bülow NS, Skouby SO, Warzecha AK, Udengaard H, Andersen CY, Holt MD, Grøndahl ML, Nyboe Andersen A, Sopa N, Mikkelsen ALE, Pinborg A, Macklon NS. Impact of letrozole co-treatment during ovarian stimulation with gonadotrophins for IVF: a multicentre, randomized, double-blinded placebo-controlled trial. Hum Reprod 2021; 37:309-321. [PMID: 34792133 DOI: 10.1093/humrep/deab249] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/20/2021] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION Does letrozole co-treatment during ovarian stimulation with gonadotrophins for IVF reduce the proportion of women with premature progesterone levels above 1.5 ng/ml at the time of triggering final oocyte maturation? SUMMARY ANSWER The proportion of women with premature progesterone above 1.5 ng/ml was not significantly affected by letrozole co-treatment. WHAT IS KNOWN ALREADY IVF creates multiple follicles with supraphysiological levels of sex steroids interrupting the endocrine milieu and affects the window of implantation. Letrozole is an effective aromatase inhibitor, normalizing serum oestradiol, thereby ameliorating some of the detrimental effects of IVF treatment. STUDY DESIGN, SIZE, DURATION A randomized, double-blinded placebo-controlled trial investigated letrozole intervention during stimulation for IVF with FSH. The trial was conducted at four fertility clinics at University Hospitals in Denmark from August 2016 to November 2018. PARTICIPANTS/MATERIALS, SETTING, METHODS A cohort of 129 women with expected normal ovarian reserve (anti-Müllerian hormone 8-32 nmol/l) completed an IVF cycle with fresh embryo transfer and received co-treatment with either 5 mg/day letrozole (n = 67) or placebo (n = 62), along with the FSH. Progesterone, oestradiol, FSH, LH and androgens were analysed in repeated serum samples collected from the start of the stimulation to the mid-luteal phase. In addition, the effect of letrozole on reproductive outcomes, total FSH consumption and adverse events were assessed. MAIN RESULTS AND THE ROLE OF CHANCE The proportion of women with premature progesterone >1.5 ng/ml was similar (6% vs 0% (OR 0.0, 95% CI [0.0; 1.6], P = 0.12) in the letrozole versus placebo groups, respectively), whereas the proportion of women with mid-luteal progesterone >30 ng/ml was significantly increased in the letrozole group: (59% vs 31% (OR 3.3, 95% CI [1.4; 7.1], P = 0.005)). Letrozole versus placebo decreased oestradiol levels on the ovulation trigger day by 68% (95% CI [60%; 75%], P < 0.0001). Other hormonal profiles, measured as AUC, showed the following results. The increase in LH in the letrozole group versus placebo group was 38% (95% CI [21%; 58%], P < 0.0001) and 34% (95% CI [11%; 61%], P = 0.006) in the follicular and luteal phases, respectively. In the letrozole group versus placebo group, testosterone increased by 79% (95% CI [55%; 105%], P < 0.0001) and 49% (95% CI [30%; 72%], P < 0.0001) in the follicular and luteal phases, respectively. In the letrozole group versus placebo group, the increase in androstenedione was by 85% (95% CI [59%; 114%], P < 0.0001) and 69% (95% CI [48%; 94%], P < 0.0001) in the follicular and luteal phases, respectively. The ongoing pregnancy rate was similar between the letrozole and placebo groups (31% vs 39% (risk-difference of 8%, 95% CI [-25%; 11%], P = 0.55)). No serious adverse reactions were recorded in either group. The total duration of exogenous FSH stimulation was 1 day shorter in the intervention group, significantly reducing total FSH consumption (mean difference -100 IU, 95% CI [-192; -21], P = 0.03). LIMITATIONS, REASONS FOR CAUTION Late follicular progesterone samples were collected on the day before and day of ovulation triggering for patient logistic considerations, and the recently emerged knowledge about diurnal variation of progesterone was not taken into account. The study was powered to detect hormonal variations but not differences in pregnancy outcomes. WIDER IMPLICATIONS OF THE FINDINGS Although the use of letrozole has no effect on the primary outcome, the number of women with a premature increase in progesterone on the day of ovulation triggering, the increased progesterone in the mid-luteal phase due to letrozole may contribute to optimizing the luteal phase endocrinology. The effect of letrozole on increasing androgens and reducing FSH consumption may be used in poor responders. However, the effect of letrozole on implantation and ongoing pregnancy rates should be evaluated in a meta-analysis or larger randomized controlled trial (RCT). STUDY FUNDING/COMPETING INTEREST(S) Funding was received from EU Interreg for ReproUnion and Ferring Pharmaceuticals, and Roche Diagnostics contributed with assays. N.S.M. and A.P. have received grants from Ferring, Merck Serono, Anecova and Gedeon Richter, and/or personal fees from IBSA, Vivoplex, ArtPred and SPD, outside the submitted work. The remaining authors have no competing interests. TRIAL REGISTRATION NUMBERS NCT02939898 and NCT02946684. TRIAL REGISTRATION DATE 15 August 2016. DATE OF FIRST PATIENT’S ENROLMENT 22 August 2016.
Collapse
Affiliation(s)
- Nathalie Søderhamn Bülow
- Department of Obstetrics and Gynaecology, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Copenhagen, Denmark.,The Fertility Department, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sven Olaf Skouby
- Department of Obstetrics and Gynaecology, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Copenhagen, Denmark
| | - Agnieszka Katarzyna Warzecha
- Department of Obstetrics and Gynaecology, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Copenhagen, Denmark
| | - Hanne Udengaard
- Department of Obstetrics and Gynaecology, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Copenhagen, Denmark
| | - Claus Yding Andersen
- Laboratory of Reproductive Biology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marianne Dreyer Holt
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Zealand University Hospital, Køge, Denmark
| | - Marie Louise Grøndahl
- Department of Obstetrics and Gynaecology, Endocrinological and Reproductive Unit, Copenhagen University Hospital, Herlev Hospital, Herlev, Copenhagen, Denmark
| | | | - Negjyp Sopa
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital, Hvidovre, Denmark
| | - Anne Lis Englund Mikkelsen
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Zealand University Hospital, Køge, Denmark
| | - Anja Pinborg
- The Fertility Department, Copenhagen University Hospital, Copenhagen, Denmark.,Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital, Hvidovre, Denmark
| | - Nicholas Stephen Macklon
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Zealand University Hospital, Køge, Denmark.,London Women's Clinic, London, UK
| |
Collapse
|
34
|
The Use of Androgen Priming in Women with Reduced Ovarian Reserve Undergoing Assisted Reproductive Technology. Semin Reprod Med 2021; 39:207-219. [PMID: 34500477 DOI: 10.1055/s-0041-1735646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Androgen priming with either dehydroepiandrosterone (DHEA) or testosterone has been suggested as an adjunct to improve in vitro fertilization (IVF) outcomes in women with diminished ovarian reserve (DOR). Numerous studies have investigated the effects of both DHEA and testosterone on IVF outcome. The results were inconsistent, and the quality of most studies is substandard. Meta-analyses have consistently reported that DHEA does appear to significantly improve IVF outcome in women with predicted or proven poor ovarian response (POR), but these have included some normal responders and/or nonrandomized studies. Our meta-analyses including randomized controlled trials (RCTs) incorporating only women with DOR or POR suggest that DHEA confers no benefit. While meta-analyses of RCTs on the use of testosterone in women with DOR or POR showed an improved IVF outcome, most studies included are of low quality with high risk of bias. When analysis of data from studies of only low-risk bias was performed, such a benefit with testosterone was not observed. Although recruitment may well be a challenge, a large, well-designed RCT is, however, still warranted to investigate whether or not androgen priming with either DHEA or testosterone should be recommended as an adjuvant treatment for women with DOR or POR undergoing IVF.
Collapse
|
35
|
Zhang WY, Gardner RM, Kapphahn KI, Ramachandran MK, Murugappan G, Aghajanova L, Lathi RB. The impact of estradiol on pregnancy outcomes in letrozole-stimulated frozen embryo transfer cycles. F S Rep 2021; 2:320-326. [PMID: 34553158 PMCID: PMC8441577 DOI: 10.1016/j.xfre.2021.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 10/25/2022] Open
Abstract
OBJECTIVE To assess the impact of low estradiol (E2) levels in letrozole-stimulated frozen embryo transfer (FET) cycles on pregnancy and neonatal outcomes. DESIGN Retrospective cohort. SETTING University-affiliated fertility center. PATIENTS All patients who underwent letrozole-stimulated FET cycles from January 2017 to April 2020 (n = 217). The "Low E2" group was defined as those with E2 serum levels on the day of trigger <10th percentile level (E2 <91.16 pg/mL, n = 22) and the "Normal E2" group was defined as those with E2 serum levels ≥10th percentile level (E2 ≥91.16 pg/mL, n = 195). INTERVENTIONS None. MAIN OUTCOME MEASURES Pregnancy outcomes including rates of clinical pregnancy, clinical miscarriage, and live birth. Neonatal outcomes including gestational age at delivery, birth weight, and Apgar score. RESULTS The mean ± SD estradiol level was 66.8 ± 14.8 pg/mL for the "Low E2" group compared with 366.3 ± 322.1 pg/mL for the "Normal E2" group. There were otherwise no substantial differences in cycle characteristics such as endometrial thickness on the day of ovulation trigger and progesterone levels in early pregnancy. The "Low E2" group had a significantly higher clinical miscarriage rate (36.4% vs. 8.8%, adjusted odds ratio 8.06) and lower live birth rate (31.8% vs. 57.9%, adjusted odds ratio 0.28). Neonatal outcomes such as gestational age at delivery, mean birth weight, Apgar scores, and incidence of newborn complications were not clinically different between the groups. CONCLUSION Low E2 levels were associated with a significantly higher miscarriage rate and lower live birth rate, suggesting that E2 levels in the follicular phase may have an effect on cycle outcomes. Given the rise in use of FET, further studies are needed to confirm our findings and understand the mechanisms.
Collapse
Affiliation(s)
- Wendy Y. Zhang
- Department of Medicine, Stanford University School of Medicine, Stanford, California
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Rebecca M. Gardner
- Quantitative Sciences Unit, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Kristopher I. Kapphahn
- Quantitative Sciences Unit, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Maya K. Ramachandran
- Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Gayathree Murugappan
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Lusine Aghajanova
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| | - Ruth B. Lathi
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
36
|
Solernou R, Peralta S, Casals G, Guimera M, Solsona M, Borras A, Manau D, Fàbregues F. The Follicular Output Rate (FORT) as a method to evaluate transdermal testosterone efficacy in poor responders. JBRA Assist Reprod 2021; 25:229-234. [PMID: 33507716 PMCID: PMC8083864 DOI: 10.5935/1518-0557.20200086] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objective: Follicular Output Rate (FORT) is an efficient quantitative and qualitative marker of ovarian responsiveness to gonadotropins. Transdermal testosterone (TT) has been used as adjuvant therapy to gonadotrophins in order to improve ovarian response in poor responders (PR). The aim of this study was to analyze whether TT can improve follicular sensitivity to gonadotropins using FORT. Methods: This retrospective study, held in a tertiary-care university hospital included 90 PR patients, according to the Bologna criteria. Patients in Group 1 (n = 46) received transdermal application of testosterone preceding gonadotrophin ovarian stimulation under pituitary suppression. In Group 2 (n = 44) ovarian stimulation was carried out with high-dose gonadotrophin in association with minidose GnRH agonist protocol. We analyzed ovarian stimulation parameters and IVF outcomes. We determined antral follicle count (AFC) (3-8 mm) before ovarian stimulation, pre-ovulatory follicle count (PFC) (16-22 mm) and the day of hCG administration. We calculated the FORT using the PFCx100/AFC ratio. Results: Baseline characteristics and ovarian reserve parameters were similar in both groups. FORT and oocytes retrieved were significantly higher in group 1 vs group 2. There were no significant differences in pregnancy rates. In group 1 there was a significant correlation between FORT and AFC. Conclusions: This study suggests that the potential beneficial mechanism of TT in poor responder patients may be based on increasing the antral follicle sensitivity to gonadotrophin. FORT is an excellent tool to demonstrate this.
Collapse
Affiliation(s)
- Roser Solernou
- Institute Clinic of Gynecology, Obstetrics and Gynecology. Hospital Clinic. Barcelona, Spain
| | - Sara Peralta
- Institute Clinic of Gynecology, Obstetrics and Gynecology. Hospital Clinic. Barcelona, Spain
| | - Gemma Casals
- Institute Clinic of Gynecology, Obstetrics and Gynecology. Hospital Clinic. Barcelona, Spain
| | - Marta Guimera
- Institute Clinic of Gynecology, Obstetrics and Gynecology. Hospital Clinic. Barcelona, Spain
| | - Marina Solsona
- Institute Clinic of Gynecology, Obstetrics and Gynecology. Hospital Clinic. Barcelona, Spain
| | - Aina Borras
- Institute Clinic of Gynecology, Obstetrics and Gynecology. Hospital Clinic. Barcelona, Spain
| | - Dolores Manau
- Institute Clinic of Gynecology, Obstetrics and Gynecology. Hospital Clinic. Barcelona, Spain
| | - Francesc Fàbregues
- Institute Clinic of Gynecology, Obstetrics and Gynecology. Hospital Clinic. Barcelona, Spain
| |
Collapse
|
37
|
Orisaka M, Miyazaki Y, Shirafuji A, Tamamura C, Tsuyoshi H, Tsang BK, Yoshida Y. The role of pituitary gonadotropins and intraovarian regulators in follicle development: A mini-review. Reprod Med Biol 2021; 20:169-175. [PMID: 33850449 PMCID: PMC8022101 DOI: 10.1002/rmb2.12371] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/15/2021] [Accepted: 01/25/2021] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND The process of follicle development is tightly regulated by pituitary gonadotropins (follicle-stimulating hormone [FSH] and luteinizing hormone [LH]) and intraovarian regulators (eg, steroids, growth factors, and cytokines). METHODS This review outlines recent findings on the mechanisms of human follicle development, based on the research on animal models such as mice, rats, cows, and sheep. MAIN FINDINGS Phosphatidylinositol 3-kinase/protein kinase B signaling pathway and anti-Müllerian hormone are involved in primordial follicle activation during the gonadotropin-independent phase. The intraovarian regulators, such as androgen, insulin-like growth factor system, activin, oocyte-derived factors (growth differentiation factor-9 and bone morphogenetic protein 15), and gap junction membrane channel protein (connexin), play a central role in the acquisition of FSH dependence in preantral follicles during the gonadotropin-responsive phase. Antral follicle development can be divided into FSH-dependent growth and LH-dependent maturation. The indispensable tetralogy for follicle selection and final maturation of antral follicles involves (a) acquisition of LH dependence, (b) greater capacity for E2 production, (c) activation of the IGF system, and (d) an antiapoptotic follicular microenvironment. CONCLUSION We reproductive endocrinologists should accumulate further knowledge from animal model studies to develop methods that promote early folliculogenesis and connect to subsequent gonadotropin therapy in infertile women.
Collapse
Affiliation(s)
- Makoto Orisaka
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| | - Yumiko Miyazaki
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| | - Aya Shirafuji
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| | - Chiyo Tamamura
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| | - Hideaki Tsuyoshi
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| | - Benjamin K. Tsang
- Reproductive Biology UnitDepartments of Obstetrics & Gynecology and Cellular & Molecular MedicineUniversity of OttawaOttawaONCanada
- Chronic Disease ProgramOttawa Hospital Research InstituteCritical Care WingThe Ottawa Hospital ‐ General CampusOttawaONCanada
| | - Yoshio Yoshida
- Department of Obstetrics and GynecologyUniversity of FukuiYoshida‐GunJapan
| |
Collapse
|
38
|
Roy S, Huang B, Sinha N, Wang J, Sen A. Androgens regulate ovarian gene expression by balancing Ezh2-Jmjd3 mediated H3K27me3 dynamics. PLoS Genet 2021; 17:e1009483. [PMID: 33784295 PMCID: PMC8034747 DOI: 10.1371/journal.pgen.1009483] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 04/09/2021] [Accepted: 03/12/2021] [Indexed: 02/07/2023] Open
Abstract
Conventionally viewed as male hormone, androgens play a critical role in female fertility. Although androgen receptors (AR) are transcription factors, to date very few direct transcriptional targets of ARs have been identified in the ovary. Using mouse models, this study provides three critical insights about androgen-induced gene regulation in the ovary and its impact on female fertility. First, RNA-sequencing reveals a number of genes and biological processes that were previously not known to be directly regulated by androgens in the ovary. Second, androgens can also influence gene expression by decreasing the tri-methyl mark on lysine 27 of histone3 (H3K27me3), a gene silencing epigenetic mark. ChIP-seq analyses highlight that androgen-induced modulation of H3K27me3 mark within gene bodies, promoters or distal enhancers have a much broader impact on ovarian function than the direct genomic effects of androgens. Third, androgen-induced decrease of H3K27me3 is mediated through (a) inhibiting the expression and activity of Enhancer of Zeste Homologue 2 (EZH2), a histone methyltransferase that promotes tri-methylation of K27 and (b) by inducing the expression of a histone demethylase called Jumonji domain containing protein-3 (JMJD3/KDM6B), responsible for removing the H3K27me3 mark. Androgens through the PI3K/Akt pathway, in a transcription-independent fashion, increase hypoxia-inducible factor 1 alpha (HIF1α) protein levels, which in turn induce JMJD3 expression. Furthermore, proof of concept studies involving in vivo knockdown of Ar in the ovary and ovarian (granulosa) cell-specific Ar knockout mouse model show that ARs regulate the expression of key ovarian genes through modulation of H3K27me3.
Collapse
Affiliation(s)
- Sambit Roy
- Reproductive and Developmental Sciences Program, Department of Animal Sciences, Michigan State University, East Lansing, MI, United States of America
| | - Binbin Huang
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, United States of America
| | - Niharika Sinha
- Reproductive and Developmental Sciences Program, Department of Animal Sciences, Michigan State University, East Lansing, MI, United States of America
| | - Jianrong Wang
- Department of Computational Mathematics, Science and Engineering, Michigan State University, East Lansing, MI, United States of America
| | - Aritro Sen
- Reproductive and Developmental Sciences Program, Department of Animal Sciences, Michigan State University, East Lansing, MI, United States of America
- * E-mail:
| |
Collapse
|
39
|
Orvieto R, Nahum R, Aizer A, Haas J, Kirshenbaum M. A Novel Stimulation Protocol for Poor-Responder Patients: Combining the Stop GnRH-ag Protocol with Letrozole Priming and Multiple-Dose GnRH-ant: A Proof of Concept. Gynecol Obstet Invest 2021; 86:149-154. [PMID: 33761501 DOI: 10.1159/000513669] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/10/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE The objective of this study was to examine whether the combined Stop GnRH-agonist (GnRH-ag), letrozole priming, and multiple-dose GnRH-antagonist (GnRH-ant) protocol may improve in vitro fertilization/intracytoplasmic sperm injection cycle in poor ovarian responders (PORs). DESIGN This was a historical cohort, proof of concept study under tertiary setting at University affiliated Medical Center. PATIENTS Five PORs fulfilling the POSEIDON Group 4 criteria were included. MAIN OUTCOME MEASURES Number of oocytes retrieved, number of top-quality embryos (TQEs), and controlled ovarian hyperstimulation (COH) variables were the main outcome measures. RESULTS The combined Stop GnRH-ag, letrozole priming, and multiple-dose GnRH-ant COH protocol revealed significantly higher number of follicles >13 mm on the day of hCG administration and higher number of oocytes retrieved, with non-significantly more TQEs and a reasonable clinical pregnancy rate. CONCLUSIONS The combined Stop GnRH-ag, letrozole priming, and multiple-dose GnRH-ant COH protocol is a valuable tool in the armamentarium for treating POSEIDON Group 4 patients. Further large prospective studies are needed to elucidate its role in POR and to identify the specific characteristics of women (before initiating ovarian stimulation) that will aid both fertility specialists' counseling and their patients in adjusting the appropriate COH protocol.
Collapse
Affiliation(s)
- Raoul Orvieto
- Department of Obstetrics and Gynecology, Infertility and IVF Unit, Chaim Sheba Medical Center, Tel Hashomer, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel, .,The Tarnesby-Tarnowski Chair for Family Planning and Fertility Regulation, at the Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel,
| | - Ravit Nahum
- Department of Obstetrics and Gynecology, Infertility and IVF Unit, Chaim Sheba Medical Center, Tel Hashomer, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adva Aizer
- Department of Obstetrics and Gynecology, Infertility and IVF Unit, Chaim Sheba Medical Center, Tel Hashomer, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jigal Haas
- Department of Obstetrics and Gynecology, Infertility and IVF Unit, Chaim Sheba Medical Center, Tel Hashomer, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Kirshenbaum
- Department of Obstetrics and Gynecology, Infertility and IVF Unit, Chaim Sheba Medical Center, Tel Hashomer, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
40
|
Abdelmeguid Y, Yakout N, Oshiba A, Zain M, Kotb M. Huge ovarian cyst in a neonate with classical 21-hydroxylase deficiency. Clin Pediatr Endocrinol 2021; 30:57-60. [PMID: 33446954 PMCID: PMC7783129 DOI: 10.1297/cpe.30.57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/09/2020] [Indexed: 11/05/2022] Open
Abstract
Congenital adrenal hyperplasia is the most common cause of ambiguous genitalia worldwide,
with an incidence of 1 in 15,000 live births. The most frequently-occurring subtype,
21-hydroxylase deficiency, results in diminished production of aldosterone and cortisol as
well as increased androgen secretion. Previous studies have reported a relationship
between ovarian cyst formation and adrenal androgen excess; nevertheless, neonatal large
ovarian cysts have rarely been reported in newborns with congenital adrenal hyperplasia.
Herein, we present the unique case of a neonate with classical 21-hydroxylase deficiency
who underwent surgery for a huge unilateral solitary ovarian follicular cyst on the
seventh postnatal day. Possible mechanisms by which androgen excess may cause ovarian cyst
formation are also discussed.
Collapse
Affiliation(s)
- Yasmine Abdelmeguid
- Department of Pediatric Endocrinology, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Nada Yakout
- Department of Pathology, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Ahmed Oshiba
- Department of Pediatric Surgery, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Mostafa Zain
- Department of Pediatric Surgery, Alexandria Faculty of Medicine, Alexandria, Egypt
| | - Mostafa Kotb
- Department of Pediatric Surgery, Alexandria Faculty of Medicine, Alexandria, Egypt
| |
Collapse
|
41
|
Neves AR, Montoya-Botero P, Polyzos NP. The Role of Androgen Supplementation in Women With Diminished Ovarian Reserve: Time to Randomize, Not Meta-Analyze. Front Endocrinol (Lausanne) 2021; 12:653857. [PMID: 34079524 PMCID: PMC8165260 DOI: 10.3389/fendo.2021.653857] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/23/2021] [Indexed: 11/24/2022] Open
Abstract
The management of patients with diminished ovarian reserve (DOR) remains one of the most challenging tasks in IVF clinical practice. Despite the promising results obtained from animal studies regarding the importance of androgens on folliculogenesis, the evidence obtained from clinical studies remains inconclusive. This is mainly due to the lack of an evidence-based methodology applied in the available trials and to the heterogeneity in the inclusion criteria and IVF treatment protocols. In this review, we analyze the available evidence obtained from animal studies and highlight the pitfalls from the clinical studies that prevent us from closing the chapter of this line of research.
Collapse
Affiliation(s)
- Ana Raquel Neves
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain
- Faculty of Medicine, Autonomous University of Barcelona, Cerdanyola del Vallès, Spain
| | - Pedro Montoya-Botero
- Department of Reproductive Medicine, Conceptum – Unidad de Fertilidad del Country, Bogotá, Colombia
| | - Nikolaos P. Polyzos
- Department of Obstetrics, Gynecology and Reproductive Medicine, Dexeus University Hospital, Barcelona, Spain
- Faculty of Medicine and Health Sciences, Ghent University (UZ Gent), Gent, Belgium
- *Correspondence: Nikolaos P. Polyzos,
| |
Collapse
|
42
|
Rosenfield RL, Cooke DW, Radovick S. Puberty in the Female and Its Disorders. SPERLING PEDIATRIC ENDOCRINOLOGY 2021:528-626. [DOI: 10.1016/b978-0-323-62520-3.00016-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
43
|
Bernstein LR, Treff NR. Editorial: Causes of Oocyte Aneuploidy and Infertility in Advanced Maternal Age and Emerging Therapeutic Approaches. Front Endocrinol (Lausanne) 2021; 12:652990. [PMID: 33708177 PMCID: PMC7940751 DOI: 10.3389/fendo.2021.652990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 01/22/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Lori R. Bernstein
- Pregmama, LLC, Gaithersburg, MD, United States
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Veterinary Integrative Biosciences, Texas A&M College of Veterinary Medicine, College Station, TX, United States
- *Correspondence: Lori R. Bernstein,
| | - Nathan R. Treff
- Genomic Prediction Inc., North Brunswick, NJ, United States
- Genomic Prediction Clinical Laboratory, North Brunswick, NJ, United States
| |
Collapse
|
44
|
Abstract
Letrozole, an aromatase inhibitor that blocks estrogen synthesis by inhibiting the final step of the estrogen biosynthetic pathway, has been used in the applications of a wide range of infertility settings. It has been more than 20 years since the initial clinical trial of letrozole for ovulation induction. In light of the accumulating clinical and basic evidence, the efficacy and safety of letrozole have been identified. This mini review focuses on our current knowledge of the applications and mechanisms of letrozole for female infertility and various questions are put forward about how letrozole could be more effectively used.
Collapse
|
45
|
Abstract
Polycystic ovary syndrome is a complex and heterogenous disorder involving multiple organ systems and different molecular pathways. It is tightly associated with obesity and especially abdominal obesity. As body weight reduction is the main modifiable risk factor for polycystic ovary syndrome, therapeutic approaches in overweight or obese women with polycystic ovary syndrome have been developed. Liraglutide is a glucagon-like peptide-1 receptor agonist that promotes sustained weight loss, as well as abdominal fat reduction, in individuals with obesity, prediabetes, and type 2 diabetes mellitus. The majority of current clinical studies have demonstrated that liraglutide therapy achieved significant reductions in body weight, body mass index, and abdominal circumference in overweight and obese women with polycystic ovary syndrome. Liraglutide therapy promoted significant improvements in free testosterone and sex hormone-binding globulin levels in some studies. Important metabolic and hormonal improvements were also reported after the combination of liraglutide with metformin. Increased menstrual frequency, as well as potential positive effects in reproduction, were described. However, the small number of participants, short duration, and low daily liraglutide dose are some of the main limitations of these studies. Larger and longer, multi-centred, double-blind, placebo-controlled trials of liraglutide monotherapy or combination therapy, with prolonged post-interventional monitoring, are crucially anticipated. Metabolic, hormonal, and reproductive primary outcomes should be uniformly addressed, to tailor future targeted treatment approaches, according to the patient phenotype and needs. This will improve long-term therapeutic outcomes in this population.
Collapse
|
46
|
Bonardi B, Massarotti C, Bruzzone M, Goldrat O, Mangili G, Anserini P, Spinaci S, Arecco L, Del Mastro L, Ceppi M, Demeestere I, Lambertini M. Efficacy and Safety of Controlled Ovarian Stimulation With or Without Letrozole Co-administration for Fertility Preservation: A Systematic Review and Meta-Analysis. Front Oncol 2020; 10:574669. [PMID: 33117711 PMCID: PMC7575927 DOI: 10.3389/fonc.2020.574669] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 09/01/2020] [Indexed: 01/01/2023] Open
Abstract
Background: The co-administration of letrozole during controlled ovarian stimulation (COS) with gonadotropins is used to limit the potentially harmful effects of a supra-physiological rise in estrogen levels on hormone-sensitive cancers. However, the efficacy and safety of adding letrozole to COS remain debated. Methods: This is a systematic review and meta-analysis of published studies that compared the efficacy and safety of COS with co-administration of letrozole vs. COS without letrozole in all patient populations. A secondary analysis was done including only the studies in breast cancer patients. The primary efficacy endpoint was the number of retrieved mature Metaphase II (MII) oocytes. Secondary efficacy and safety endpoints were total number of oocytes, maturation rate, fertilization rate, number of cryopreserved embryos, peak estradiol levels, progesterone levels, and total gonadotropin dose. Data for each endpoint were reported and analyzed thorough mean ratio (MR) with 95% confidence interval (CI). Results: A total of 11 records were selected including 2,121 patients (990 patients underwent COS with letrozole and 1,131 COS without letrozole). The addition of letrozole to COS did not have any negative effect on the number of mature oocytes collected (MR = 1.00, 95% CI = 0.87–1.16; P = 0.967) and the other efficacy endpoints. COS with letrozole was associated with significantly decreased peak estradiol levels (MR = 0.28, 95% CI = 0.24–0.32; P < 0.001). Similar results were observed in the secondary analysis including only breast cancer patients. Conclusions: These findings are reassuring on the efficacy and safety of COS with gonadotropins and letrozole and are particularly important for fertility preservation in women with hormone-sensitive cancers.
Collapse
Affiliation(s)
- Benedetta Bonardi
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Claudia Massarotti
- Physiopathology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marco Bruzzone
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Giorgia Mangili
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Anserini
- Physiopathology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Stefano Spinaci
- Division of Breast Surgery, Ospedale Villa Scassi, Genova, Italy
| | - Luca Arecco
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.,Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.,Breast Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Marcello Ceppi
- Clinical Epidemiology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Isabelle Demeestere
- Research Laboratory on Human Reproduction, Université Libre de Bruxelles (ULB), Brussels, Belgium.,Fertility Clinic, CUB-Hôpital Erasme, Brussels, Belgium
| | - Matteo Lambertini
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, University of Genova, Genova, Italy.,Department of Medical Oncology, U.O.C. Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
47
|
The Benefits of Testosterone Therapy in Poor Ovarian Responders Undergoing In Vitro Fertilisation (IVF). EUROPEAN MEDICAL JOURNAL 2020. [DOI: 10.33590/emj/20-00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Introduction: Poor ovarian responders are the most challenging patients in reproductive medicine and no successful treatment has been proposed. Androgens are thought to play an important role during early folliculogenesis and diminished levels are associated with decreased ovarian sensitivity to follicle-stimulating hormone. This study aimed to determine whether pretreatment with testosterone improves the results in poor responders undergoing in vitro fertilisation (IVF). Materials and methods: This observational pilot study enrolled 33 poor responders undergoing IVF. Eleven patients were pretreated with 250 mg intramuscular testosterone and compared to a control group of 22 patients. The participants were tested for free testosterone, dehydroepiandrosterone sulfate, sex hormone binding globulin, and anti-mullerian hormone (AMH). Results: The two groups had similar baseline characteristics. Significant improvement was reached in the hormones free testosterone, dehydroepiandrosterone sulfate, and sex hormone binding globulin in the testosterone-pretreatment group. No difference was detected in antral follicle count (5.06 versus 4.24); AMH (0.51 versus 0.53), mature oocytes (2.2 versus 2.32), and the number of embryos (1.2 versus 1.33) between the study and control groups, respectively. There was a slow improvement in fertilisation rate but without any significance (62.97% versus 57.61%). However, the cancellation rate of the ovarian stimulation was much greater in the control group (18.18%) in comparison with the study group (0.0%). Pregnancy rate (PR) in the testosterone group was higher than controls (PR per cycle: 27.3% versus 4.6; p=0.09). Conclusion: Based on the limited number of patients studied, pretreatment with testosterone seems to improve PR and cancellation rate in poor responders but failed to affect antral follicle count, AMH, and the number of mature oocytes and embryos. Given these results, further research would provide more certainty.
Collapse
|
48
|
Tremblay PG, Sirard MA. Gene analysis of major signaling pathways regulated by gonadotropins in human ovarian granulosa tumor cells (KGN)†. Biol Reprod 2020; 103:583-598. [PMID: 32427331 DOI: 10.1093/biolre/ioaa079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 03/17/2020] [Accepted: 05/18/2020] [Indexed: 11/13/2022] Open
Abstract
The female reproductive function largely depends on timing and coordination between follicle-stimulating hormone (FSH) and luteinizing hormone. Even though it was suggested that these hormones act on granulosa cells via shared signaling pathways, mainly protein kinases A, B, and C (PKA, PKB, and PKC), there is still very little information available on how these signaling pathways are regulated by each hormone to provide such differences in gene expression throughout folliculogenesis. To obtain a global picture of the principal upstream factors involved in PKA, PKB, and PKC signaling in granulosa cells, human granulosa-like tumor cells (KGN) were treated with FSH or specific activators (forskolin, SC79, and phorbol 12-myristate 13-acetate) for each pathway to analyze gene expression with RNA-seq technology. Normalization and cutoffs (FC 1.5, P ≤ 0.05) revealed 3864 differentially expressed genes between treatments. Analysis of major upstream regulators showed that PKA is a master kinase of early cell differentiation as its activation resulted in the gene expression profile that accompanies granulosa cell differentiation. Our data also revealed that the activation of PKC in granulosa cells is also a strong differentiation signal that could control "advanced" differentiation in granulosa cells and the inflammatory cascade that occurs in the dominant follicle. According to our results, PKB activation provides support for PKA-stimulated gene expression and is also involved in granulosa cell survival throughout follicular development. Taken together, our results provide new information on PKA, PKB, and PKC signaling pathways and their roles in stimulating a follicle at the crossroad between maturation/ovulation and atresia.
Collapse
Affiliation(s)
- Patricia G Tremblay
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculté des sciences de l'agriculture et de l'alimentation, Département des Sciences animales, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Marc-André Sirard
- Centre de recherche en Reproduction, Développement et Santé Intergénérationnelle, Faculté des sciences de l'agriculture et de l'alimentation, Département des Sciences animales, Université Laval, Québec, QC, G1V 0A6, Canada
| |
Collapse
|
49
|
A review of the physiology behind letrozole applications in infertility: are current protocols optimal? J Assist Reprod Genet 2020; 37:2093-2104. [PMID: 32712844 PMCID: PMC7492298 DOI: 10.1007/s10815-020-01892-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/13/2020] [Indexed: 11/23/2022] Open
Abstract
Letrozole is a targeted aromatase inhibitor which has primarily been used in post-menopausal women with breast cancer. Recently, it has been utilized in infertile pre-menopausal women because of its ability to enhance FSH production for ovulation induction. However, the ovarian follicle’s response to FSH is only a part of the endocrine events occurring in a developing follicle. The health of the small antral follicles is driven primarily by androgens, which contribute to granulosa cell mitosis, sensitivity to FSH, and resistance to atresia. In contrast, elevated androgens in the late antral to pre-ovulatory follicle have a negative impact on follicle health and lead to atresia and cystic follicle formation. This ovarian physiologic data suggests that current applications of letrozole to infertility may be squandering some of the primary benefits available in using letrozole to promote follicle development. Four applications of letrozole to infertility that have appeared in the medical literature are reviewed. Androgen-related benefits are reviewed and various questions put forward about how letrozole could be more effectively used to help patients in these settings.
Collapse
|
50
|
Zwiefelhofer EM, Mapletoft RJ, Adams GP. An attempt to potentiate the ovarian superstimulatory response in cattle by co-treatment with an aromatase inhibitor. Theriogenology 2020; 157:1-6. [PMID: 32768722 DOI: 10.1016/j.theriogenology.2020.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/09/2020] [Accepted: 07/16/2020] [Indexed: 11/16/2022]
Abstract
Letrozole is used for the treatment of subfertility in women undergoing ovarian superstimulation, but the mechanism of action has not been investigated critically. The objective was to test the hypothesis that treatment with letrozole will potentiate the superstimulatory response following gonadotropin treatment by increasing the number of follicles present at ovarian follicular wave emergence in cattle. In Experiment 1, ovarian follicular wave emergence was synchronized among beef heifers (n = 8) by transvaginal ultrasound-guided follicle ablation. On Day 0 (wave emergence), a letrozole-releasing device (LRD) was placed intravaginally for 5 days, followed again by transvaginal follicle ablation on Day 5. The number of follicles ≥3 mm was recorded by transrectal ultrasonography on Days 0 and 6.5 (i.e., pre- vs. post-LRD treatment). In Experiment 2, non-lactating dairy cows were assigned randomly to one of two groups (n = 15/gp) after follicle ablation-induced synchronization of wave emergence (Day 0), and given either an LRD or sham device for 5 days. Superstimulatory treatment was initiated on Day 0, consisting of 8 doses of 50 mg of porcine FSH im at 12 h intervals, and luteolytic doses of prostaglandin on Days 3 and 3.5. The LRD/sham devices were removed on Day 3.5, GnRH was given im on Day 5, estrus response was determined on Days 5 and 6, and the ovarian response was recorded by ultrasonography on Days 0, 3.5, 5, 6.5, and 12. In Experiment 1, no difference was detected in the number of antral follicles at wave emergence pre- vs. post-LRD treatment (23.2 ± 3.2 vs. 23.5 ± 3.8 follicles; P = 0.67; mean ± SEM). In Experiment 2, the interval from prostaglandin treatment to estrus was longer (50.3 ± 1.1 vs. 40.7 ± 2.0 h; P < 0.001) and less variable (residuals: 3.1 ± 0.5 vs. 6.7 ± 0.9 h; P < 0.01) in the LRD vs. sham group. The proportion of ovulations (number of CL on Day 12 over the number of follicles ≥3 mm on Day 0) did not differ (0.65 ± 0.02 vs. 0.70 ± 0.02; P = 0.15) nor did the number of CL on Day 12 (15.9 ± 2.5 vs. 19.0 ± 2.0; P = 0.32) between the LRD and sham groups. In summary, treatment with letrozole did not increase the number of antral follicles at wave emergence or the superstimulatory response, but increased precision in the interval to estrus and may be useful for artificial insemination at a fixed time in superstimulatory protocols.
Collapse
Affiliation(s)
- Eric M Zwiefelhofer
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Reuben J Mapletoft
- Department of Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Gregg P Adams
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|