Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Assist Reprod Genet ; 38(2): 513-516, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33409752

ABSTRACT

PURPOSE: To describe a case of a young woman who presented for fertility preservation and underwent ovarian stimulation with an etonogestrel implant in place. METHODS: A 24-year old, gravida 0, with an etonogestrel implant and newly diagnosed lower extremity sarcoma and DVT desiring oocyte cryopreservation prior to adjuvant chemotherapy and radiation. To avoid delay in her oncologic care and allow for continued use of contraception post-retrieval, the patient underwent controlled ovarian hyperstimulation (COH) without removal of the etonogestrel implant. RESULTS: Baseline labs included follicle-stimulating hormone 9 mIU/mL, luteinizing hormone 4.9 mIU/mL, estradiol 42 pg/mL, anti-Müllerian hormone 5.1 ng/mL, and antral follicle count greater than 40. The patient was placed on an antagonist protocol and stimulated with 125 IU Gonal-F and 75 IU Menopur. She received a total of 12 days of gonadotropin stimulation. On the day of trigger, her estradiol was 1472 pg/mL, lead follicle 21.5 mm with a total of 25 follicles measured > 12 mm. She was triggered with 5000 U hCG. She had a total of 23 oocytes retrieved, 17 of which were metaphase II and vitrified. CONCLUSIONS: COH and successful oocyte cryopreservation can be achieved in patients with an etonogestrel implant in situ without apparent detrimental effects to oocyte yield or maturity. Due to the etonogestrel implant's inhibitory effects on LH, it is recommended to use an hCG trigger for final oocyte maturation.


Subject(s)
Desogestrel/administration & dosage , Fertility Preservation , Infertility, Female/drug therapy , Neoplasms/complications , Adult , Anti-Mullerian Hormone/administration & dosage , Cryopreservation , Female , Follicle Stimulating Hormone/administration & dosage , Gonadotropin-Releasing Hormone/administration & dosage , Humans , Infertility, Female/etiology , Infertility, Female/metabolism , Infertility, Female/pathology , Luteinizing Hormone/administration & dosage , Neoplasms/pathology , Oocyte Retrieval/methods , Oocytes/drug effects , Oocytes/growth & development , Oogenesis/drug effects , Oogenesis/genetics , Ovarian Hyperstimulation Syndrome , Ovulation Induction/methods , Prostheses and Implants/adverse effects , Vitrification
2.
J Assist Reprod Genet ; 36(2): 229-239, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30430314

ABSTRACT

PURPOSE: To determine the effects of PGL1001, a somatostatin receptor isoform-2 (SSTR-2) antagonist, on ovarian follicle development, oocyte fertilization, and subsequent embryo developmental potential in the rhesus macaque. METHODS: Cycling female rhesus macaques (N = 8) received vehicle through one menstrual (control) cycle, followed by daily injections of PGL1001, a SSTR-2 antagonist, for three menstrual (treatment) cycles. Main endpoints include overall animal health and ovarian hormones (e.g., estradiol [E2], progesterone [P4], and anti-Müllerian hormone [AMH]), ovarian circumference, numbers of oocytes and their maturation status following controlled ovarian stimulation (COS), as well as oocyte fertilization and subsequent blastocyst rates that were assessed in control and PGL1001 treatment cycles. Circulating PGL1001 levels were assessed at baseline as well as 6, 60, and 90 days during treatment. RESULTS: PGL1001 treatment did not impact overall animal health, menstrual cycle length, or circulating levels of ovarian hormones (E2, P4, and AMH) in comparison to vehicle treatment during natural cycles. PGL1001 treatment increased (p Ë‚ 0.05) ovarian circumference and the day 8 to day 1 ratio of AMH levels (p Ë‚ 0.05) during a COS protocol, as well as oocyte fertilization rates compared to the vehicle treatment interval. Blastocyst development rates were not significantly different between vehicle and PGL1001 treatment groups. CONCLUSION: Prolonged treatment with PGL1001 appears to be safe and does not affect rhesus macaque general health, menstrual cycle length, or ovarian hormone production. Interestingly, PGL1001 treatment increased the fertilization rate of rhesus macaque oocytes collected following ovarian stimulation.


Subject(s)
Embryonic Development/drug effects , Oocytes/drug effects , Ovarian Follicle/growth & development , Receptors, Somatostatin/antagonists & inhibitors , Animals , Anti-Mullerian Hormone/administration & dosage , Blastocyst/drug effects , Female , Fertilization/drug effects , Humans , Macaca mulatta , Ovarian Follicle/drug effects , Ovulation Induction/methods , Progesterone/administration & dosage , Somatostatin/metabolism
3.
Fertil Steril ; 110(6): 1162-1172, 2018 11.
Article in English | MEDLINE | ID: mdl-30396561

ABSTRACT

OBJECTIVE: To study whether follicular growth and oocyte maturation can be improved by antimüllerian hormone (AMH) modulation at specific stages of follicular development. DESIGN: Primary and secondary follicles were cultured in a matrix-free system and were assigned to the control group and the group with AMH supplementation during the preantral stage and neutralizing AMH antibody addition during the antral stage. SETTING: National primate research center. ANIMAL(S): Adult, female rhesus macaques (Macaca mulatta). INTERVENTION(S): None. MAIN OUTCOME MEASURE(S): Follicle survival, growth, steroid and paracrine factor production, and oocyte competence were evaluated. Follicles were assessed for expression of genes that are critical for gonadotropin signaling, cumulus cell glycolysis, and oocyte quality. RESULT(S): Primary follicles formed "organoids" and developed to the antral stage in group culture. AMH exposure during the preantral stage increased organoid diameters. Oocytes from the AMH-treated organoids had greater diameters and matured to the metaphase II (MII) stage. Secondary follicles developed to the antral stage during individual culture. The AMH exposure during the preantral stage and AMH antibody treatment during the antral stage increased follicle diameters, vascular endothelial growth factor and follistatin production, differentiation factor 9 expression, and oocyte diameters. The MII oocytes from the AMH-modulated group developed to the morula stage after IVF, with one to the blastocyst stage. CONCLUSION(S): AMH supplementation at the preantral stage and depletion at the antral stage enhanced primate follicular development and oocyte competence in vitro. The improved embryonic development supports in vitro follicle maturation as a potential approach for fertility preservation.


Subject(s)
Anti-Mullerian Hormone/administration & dosage , In Vitro Oocyte Maturation Techniques/methods , Oocytes/drug effects , Oocytes/growth & development , Ovarian Follicle/drug effects , Ovarian Follicle/growth & development , Animals , Cell Culture Techniques/methods , Female , Macaca mulatta
4.
J Assist Reprod Genet ; 35(10): 1831-1841, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30043336

ABSTRACT

OBJECTIVE: To determine whether recombinant AMH (rAMH) could prevent post-transplant follicular depletion by acting on the stemness markers Oct-4, Sox2, and NANOG. MATERIALS AND METHODS: This was an experimental study where 12 ovariectomized nude mice were xenotransplanted with vitrified/warmed ovarian cortex obtained from a pre-pubertal girl and Alzet pumps delivering rAMH, or placebo (control), were inserted intra-abdominally. Previously vitrified/warmed ovarian cortex fragments were transplanted after 7 days and then harvested after 14 days from pump placement. We performed real-time RT-PCR analyses, ELISA for AMH, FSH, and estradiol, histologic measurement of ovarian follicles, and immunohistochemistry for Ki67 and TUNEL. The main outcome measures were serum levels and tissue expression of the parameters under investigation and follicle count. RESULTS: Serum AMH, FSH, and estradiol reflected post-ovariectomy profiles and were mildly influenced by rAMH administration. Ovarian cortex expression of AMH, AMH-R2, VEGF, GDF9, Oct-4, and Sox2 was lower in rAMH mice than in controls, while NANOG was upregulated. There was a non-significant decrease in primordial follicles after vitrification-warming, and xenotransplantation further decreased this number. There were lower cell replication and depressed apoptosis in the rAMH group. CONCLUSIONS: Administration of recombinant AMH in the peri-transplant period did not protect the initial follicular depletion but decreased apoptosis and cellular activation and regulated stem cell markers' tissue expression. These results aid our understanding of the inhibitory effects of AMH on follicular development and show the benefit of administering exogenous AMH at the time of pre-pubertal ovarian cortex transplant to protect the follicles from pre-activation and premature depletion.


Subject(s)
Anti-Mullerian Hormone/genetics , Heterografts/metabolism , Ovarian Follicle/transplantation , Ovary/transplantation , Animals , Anti-Mullerian Hormone/administration & dosage , Anti-Mullerian Hormone/blood , Apoptosis/genetics , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Gene Expression Regulation, Developmental , Heterografts/drug effects , Heterografts/growth & development , Humans , Mice , Nanog Homeobox Protein/genetics , Octamer Transcription Factor-3/genetics , Ovarian Follicle/growth & development , Ovarian Follicle/metabolism , Ovariectomy , Ovary/drug effects , Ovary/growth & development , Ovary/metabolism , SOXB1 Transcription Factors/genetics , Transplantation, Heterologous , Vitrification
5.
Breast Cancer Res Treat ; 171(3): 649-656, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29943121

ABSTRACT

PURPOSE: This study evaluated predictive factors for recovery of ovarian function after ovarian protection by GnRH agonist during chemotherapy in young breast cancer patients. METHODS: This prospective cohort study analyzed 105 young breast cancer patients who were studied longitudinally after receiving GnRH agonist during cyclophosphamide-based chemotherapy for ovarian protection. Associations between pretreatment hormones, clinical factors, and recovery of ovarian function (resumption of menstruation or anti-Müllerian hormone (AMH) ≥ 1 ng/ml) were evaluated at 12 months and long-term follow-up after completion of chemotherapy. RESULTS: Mean age was 32 years (range 23-42 years). In multivariate analyses, tamoxifen use (P = 0.035) and pretreatment follicle-stimulating hormone (FSH) (P = 0.032) were predictive of resumption of menstruation, and age (P = 0.019), tamoxifen use (P = 0.022), pretreatment FSH (P < 0.001), and AMH (P = 0.040) were predictors for AMH ≥ 1 ng/ml at 12 months. In addition, pretreatment AMH was a predictor for AMH ≥ 1 ng/ml after long-term follow-up. Receiver operating characteristic curve analyses gave area under the curve of 0.805 for resumption of menstruation and 0.903 for serum AMH concentration ≥ 1 ng/ml at 12 months, when age, tamoxifen use, pretreatment FSH, and AMH were combined. CONCLUSION: Pretreatment AMH (3.26 ng/ml), age (33.9 years), pretreatment FSH (5.5 IU/l), and tamoxifen use are useful predictors for AMH ≥ 1 ng/ml at 12 months after GnRH agonist. This finding will support patient and clinician decision-making regarding fertility preservation.


Subject(s)
Breast Neoplasms/drug therapy , Gonadotropin-Releasing Hormone/administration & dosage , Ovary/drug effects , Recovery of Function/drug effects , Adult , Anti-Mullerian Hormone/administration & dosage , Breast Neoplasms/blood , Breast Neoplasms/physiopathology , Drug Therapy , Drug-Related Side Effects and Adverse Reactions/physiopathology , Female , Follicle Stimulating Hormone/administration & dosage , Gonadotropin-Releasing Hormone/agonists , Humans , Menstruation/drug effects , Neoplasm Staging , Ovarian Function Tests/methods , Ovary/physiopathology , Tamoxifen/administration & dosage
6.
Nat Med ; 24(6): 834-846, 2018 06.
Article in English | MEDLINE | ID: mdl-29760445

ABSTRACT

Polycystic ovary syndrome (PCOS) is the main cause of female infertility worldwide and corresponds with a high degree of comorbidities and economic burden. How PCOS is passed on from one generation to the next is not clear, but it may be a developmental condition. Most women with PCOS exhibit higher levels of circulating luteinizing hormone, suggestive of heightened gonadotropin-releasing hormone (GnRH) release, and anti-Müllerian hormone (AMH) as compared to healthy women. Excess AMH in utero may affect the development of the female fetus. However, as AMH levels drop during pregnancy in women with normal fertility, it was unclear whether their levels were also elevated in pregnant women with PCOS. Here we measured AMH in a cohort of pregnant women with PCOS and control pregnant women and found that AMH is significantly more elevated in the former group versus the latter. To determine whether the elevation of AMH during pregnancy in women with PCOS is a bystander effect or a driver of the condition in the offspring, we modeled our clinical findings by treating pregnant mice with AMH and followed the neuroendocrine phenotype of their female progeny postnatally. This treatment resulted in maternal neuroendocrine-driven testosterone excess and diminished placental metabolism of testosterone to estradiol, resulting in a masculinization of the exposed female fetus and a PCOS-like reproductive and neuroendocrine phenotype in adulthood. We found that the affected females had persistently hyperactivated GnRH neurons and that GnRH antagonist treatment in the adult female offspring restored their neuroendocrine phenotype to a normal state. These findings highlight a critical role for excess prenatal AMH exposure and subsequent aberrant GnRH receptor signaling in the neuroendocrine dysfunctions of PCOS, while offering a new potential therapeutic avenue to treat the condition during adulthood.


Subject(s)
Anti-Mullerian Hormone/blood , Fetus/metabolism , Polycystic Ovary Syndrome/blood , Polycystic Ovary Syndrome/pathology , Adult , Animals , Anti-Mullerian Hormone/administration & dosage , Brain/pathology , Case-Control Studies , Estrous Cycle , Female , Fertility , Gonadotropin-Releasing Hormone/metabolism , Green Fluorescent Proteins/metabolism , Humans , Luteinizing Hormone/metabolism , Mice, Inbred C57BL , Neurons/metabolism , Neurosecretory Systems/metabolism , Ovary/pathology , Phenotype , Placenta/pathology , Polycystic Ovary Syndrome/physiopathology , Pregnancy , Pregnancy Trimester, Second/blood
7.
Reprod Sci ; 25(8): 1218-1223, 2018 08.
Article in English | MEDLINE | ID: mdl-29141508

ABSTRACT

OBJECTIVE: To test whether recombinant anti-Müllerian hormone (AMH) can inhibit ovarian cortex function by modulating the expression of other hormone receptors. MATERIALS AND METHODS: Pilot experimental study with ovarian cortex obtained from 5 patients. Immediately after explant, the ovarian cortex specimens were divided into 5 equal fragments. One fragment was flash-frozen (uncultured) and 4 were incubated for 48 hours at 37°C in a pH-adjusted gamete buffer medium with increasing AMH concentrations of 0, 5, 25, and 50 ng/mL. After incubation, all specimens were rinsed and flash-frozen for polymerase chain reaction (PCR) executed in triplicates. We utilized real-time reverse transcription-polymerase chain reaction (RT-PCR) to determine messenger RNA (mRNA) levels of AMH and its receptor Anti-Müllerian Hormone-Receptor 2 (AMH-R2), follicle stimulating hormone receptor (FSH-R), luteinizing hormone receptor (LH-R), inhibin B, and insulin-like growth factor 1 receptor 1 (IGF1-R1) in ovarian cortex tissue. In addition, we performed Ki-67 immunostaining to evaluate cell proliferation in the treatment groups. RESULTS: Absence of recombinant human AMH (rAMH) caused upregulation of all markers. Exposure to increasing rAMH concentrations caused tissue AMH expression downregulation ( P = .024), while AMH-R2 ( P = .005), FSH-R ( P = .009), LH-R ( P = .003), and inhibin B ( P = .001) mRNA expression followed a bell-shaped response with an increased expression at low dose, followed by a decreased expression at higher doses. Expression of IGF1-R1 was independent ( P = .039) of rAMH exposure. The Ki-67 immunostaining showed an increased cell proliferation in the media control compared to the uncultured and the tissue cultured with rAMH. CONCLUSIONS: Culture with increasing rAMH concentrations caused downregulation of its own, as well as other hormone receptors, and a decreased ovarian cortex cell proliferation. These results help understanding the inhibitory effects of AMH on follicular development.


Subject(s)
Anti-Mullerian Hormone/metabolism , Ovary/metabolism , Receptors, Peptide/metabolism , Receptors, Pituitary Hormone/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Adult , Anti-Mullerian Hormone/administration & dosage , Female , Gene Expression Regulation , Humans , Inhibins/metabolism , Ovary/drug effects , Pilot Projects , Premenopause , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism , Receptors, FSH/metabolism , Receptors, LH/metabolism , Receptors, Somatomedin/metabolism , Recombinant Proteins
9.
Ugeskr Laeger ; 179(32)2017 Aug 07.
Article in Danish | MEDLINE | ID: mdl-28869028

ABSTRACT

The need for fertility treatment in Denmark is increasing, one of the main causes being postponed parenthood. Ovarian reserve is determined by the anti-Müllerian hormone (AMH) level and by antral follicle count. Both tests can predict the ovarian response to gonadotropin treatment prior to in vitro fertilization (IVF) and are used to individualize ovarian stimulation to achieve an appropriate number of follicles and to avoid ovarian hyperstimulation. A new assay detects low values of AMH and may predict who will benefit from IVF, and who should be referred directly to oocyte donation.


Subject(s)
Anti-Mullerian Hormone/blood , Infertility, Female , Anti-Mullerian Hormone/administration & dosage , Cell Count , Denmark , Enzyme-Linked Immunosorbent Assay , Female , Fertilization in Vitro/methods , Follicle Stimulating Hormone, Human/administration & dosage , Humans , Infertility, Female/diagnosis , Infertility, Female/therapy , Ovarian Follicle , Ovarian Reserve , Pregnancy
10.
Prog. obstet. ginecol. (Ed. impr.) ; 60(4): 341-346, jul.-ago. 2017. tab, graf
Article in Spanish | IBECS | ID: ibc-165799

ABSTRACT

Objetivo: la hormona antimülleriana es un marcador clínico de la reserva ovárica pero no disponemos de sus valores de referencia en la población española. Se han determinado sus valores de normalidad en relación a la edad en una amplia muestra de población española. Sujetos y métodos: se estudiaron 10.443 mujeres (edad 20-45 años). Todas las determinaciones séricas de hormona antimülleriana se realizaron mediante un test de ELISA (hormona antimülleriana Gen II ELISA assay; Beckman Coulter, Brea, CA, USA). Resultados: la edad media fue 36,6 ± 4,3 años. Los niveles de la hormona antimülleriana se correlacionaron inversamente con la edad (r = −0,35; p < 0,001). La edad ovárica aumentaba 1 año por cada descenso medio de 0,2 ng/ml de hormona antimülleriana. Se obtuvieron diferencias significativas en los valores de hormona antimülleriana entre Cataluña, Baleares y Andalucía. Conclusiones: este estudio ofrece estimaciones de los valores de referencia de hormona antimülleriana en función de la edad y contribuye a determinar con mayor precisión la reserva ovárica de las mujeres españolas (AU)


Objetives: Antimüllerian hormone is considered clinically useful in the evaluation of ovarian reserve, and few data exists regarding its distribution in the Spanish population. We determine normality values of antimüllerian hormone related to age in a large Spanish community cohort. Subjects and methods: We study 10,443 women (aged 20-45). Antimüllerian hormone values were analysed using an ELISA assay (antimüllerian hormone Gen II ELISA assay; Beckman Coulter, Brea, CA, USA). Results: The mean age of women was 36.6 ± 4.3 years. Antimüllerian hormone values were inversely correlated with age (r = −0.35; p < 0.001). From the regression equation, the estimated yearly decrease in antimüllerian hormone was 0.2 ng/ml. Significant differences were obtained in mean values of antimüllerian hormone among Cataluña, Baleares, and Andalucía. Conclusions: This study reports the distribution of antimüllerian hormone values in different age groups in Spain, and contribute to determine the ovarian reserve in Spanish women (AU)


Subject(s)
Humans , Female , Young Adult , Adult , Middle Aged , Anti-Mullerian Hormone/therapeutic use , Ovarian Function Tests/methods , Ovarian Reserve/physiology , Fertility/physiology , In Vitro Oocyte Maturation Techniques/methods , Anti-Mullerian Hormone/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Cross-Sectional Studies/methods , Analysis of Variance , Anti-Mullerian Hormone/administration & dosage , In Vitro Oocyte Maturation Techniques
11.
J Assist Reprod Genet ; 34(9): 1131-1136, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28660501

ABSTRACT

PURPOSE: Anti-Müllerian hormone (AMH) and forkhead box L2 (FOXL2) are two pivotal genes expressed in human granulosa cells (hGCs) where both genes share similar inhibitory functions on activation and follicular growth in order to preserve the ovarian follicle reserve. Furthermore, AMH and FOXL2 contribute to inhibit steroidogenesis, decreasing or preventing the activation of gonadotrophin-dependent aromatase CYP19A1 cytochrome P450 family 19 subfamily A member 1 (CYP19A1). The purpose of this study is to evaluate the role of AMH in regulating the expression of FOXL2. METHODS: Primary cultures of hGCs were treated with increasing concentrations of recombinant human AMH (rhAMH; range 10-100 ng/ml) for 3 h. Negative controls were performed using corresponding amounts of AMH vehicle. Total RNA or proteins were purified and quantified by spectrophotometry. FOXL2 and CYP19A1 gene expression, normalized by reference gene ribosomal protein S7 (RpS7), was evaluated by RT-qPCR. Each reaction was repeated in triplicate. Statistical analysis was performed. Extracted proteins were analyzed by immunoblot using anti-FOXL2 and anti-ß-actin as primary antibodies. RESULTS: rhAMH treatments tested did not modulate the basal expression of aromatase CYP19A1 gene. rhAMH (50 ng/ml) was able to increase FOXL2 gene expression and its intracellular content. CONCLUSIONS: This study demonstrated the existence of an AMH-FOXL2 relationship in hGCs. AMH is capable of increasing both gene and protein expression of FOXL2. Because FOXL2 induces AMH transcription, these ovarian factors could be finely regulated by a positive feedback loop mechanism to preserve the ovarian follicle reserve.


Subject(s)
Anti-Mullerian Hormone/genetics , Aromatase/genetics , Forkhead Box Protein L2/genetics , Steroids/biosynthesis , Anti-Mullerian Hormone/administration & dosage , Female , Gene Expression Regulation, Developmental/drug effects , Granulosa Cells/metabolism , Humans , Primary Cell Culture , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics
12.
Biomed Res Int ; 2016: 4286528, 2016.
Article in English | MEDLINE | ID: mdl-28097133

ABSTRACT

Background. Patients with poor ovarian response to ovarian hyperstimulation represent an interesting group for studying the impact of embryo cleavage irregularities on clinical outcome since all embryos, regardless of their quality, are usually transferred to the uterus. The aim of our study was to follow the morphokinetics of fertilized oocytes from natural cycles in poor responders. Methods. Zygotes from 53 cycles were cultured in vitro for 3 days. The morphokinetics of their development and transfer outcomes were retrospectively analyzed for the normally and irregularly cleaved embryos. Results. Of all embryos, 30.2% had single and 20.8% multiple cleavage irregularities with the following prevalence: developmental arrest 30.2%, direct cleavage to more than two cells 24.5%, chaotic cleavage 13.2%, and reverse cleavage 11.3%. These embryos had longer pronuclear phases, first cytokinesis, second embryo cell cycles, and less synchronized divisions. The transfer of normally developing embryos resulted in an implantation rate of 30.8% and a delivery rate of 23.1%, but irregularly cleaved embryos did not implant. Conclusions. The use of time-lapse microscopy in poor responder patients identified embryos with cleavage abnormalities that are related with no or extremely low implantation potential. Gained information about embryo quality is important for counselling patients about their expectations.


Subject(s)
Embryo Implantation/drug effects , Fertilization in Vitro , Oocytes/growth & development , Ovulation Induction/methods , Adult , Anti-Mullerian Hormone/administration & dosage , Blastocyst/drug effects , Blastocyst/pathology , Embryo Culture Techniques/methods , Embryo Transfer/methods , Female , Humans , Oocytes/drug effects , Oocytes/pathology , Pregnancy , Pregnancy Rate , Sperm Injections, Intracytoplasmic
13.
J Assist Reprod Genet ; 33(1): 95-100, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26631403

ABSTRACT

PURPOSE: Anti Müllerian Hormone (AMH) has a negative and inhibitory role in many functions of human granulosa-lutein cells (hGCs) including notoriously the reduction of the aromatase CYP19A1 expression induced by follicle-stimulating hormone (FSH). No data have been provided on the possible role of AMH in modulating the response to luteinizing hormone (LH) (alone or combined with FSH) as well as its effect on other enzymes involved in steroidogenesis including aromatase P450scc. The aim of this study was to investigate the role of AMH as regulator of the basal and stimulated steroids production by hGCs. METHODS: Primary culture of hGCs were incubated with hormones AMH, LH, and FSH, alone or in combination. The CYP19A1 and P450scc messenger RNA (mRNA) expression, normalized by housekeeping ribosomal protein S7 (RpS7) gene, was evaluated by reverse transcriptase quantitative PCR (RT-qPCR). Each reaction was repeated in triplicate. Negative controls using corresponding amount of vehicle control for each hormone treatment were performed. RESULT: AMH did not modulate the basal mRNA expression of both aromatase genes at any of the concentrations tested. Meanwhile, the strong mRNA induction of CYP19A1 and P450scc generated by a 24-h gonadotropin treatment (alone and combined) was suppressed by 20 ng/ml AMH added to culture medium. CONCLUSIONS: These findings contribute in clarifying the relationship between hormones regulating the early phase of steroidogenesis confirming that AMH is playing a suppressive role on CYP19A1 expression stimulated by gonadotropin in hGCs. Furthermore, a similar inhibitory effect for AMH was observed on P450scc gene expression when activated by gonadotropin treatment.


Subject(s)
Anti-Mullerian Hormone/metabolism , Aromatase/biosynthesis , Cholesterol Side-Chain Cleavage Enzyme/biosynthesis , Granulosa Cells/metabolism , Anti-Mullerian Hormone/administration & dosage , Culture Media/chemistry , Estradiol/biosynthesis , Female , Follicle Stimulating Hormone/administration & dosage , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation, Developmental/drug effects , Gonadotropins/administration & dosage , Granulosa Cells/drug effects , Humans , Luteinizing Hormone/administration & dosage , Luteinizing Hormone/metabolism , Primary Cell Culture , RNA, Messenger/biosynthesis
14.
Reprod Sci ; 23(1): 51-60, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26156852

ABSTRACT

Follicle loss occurs after ovary cryopreservation and transplantation. To preserve the follicle pool of cryopreserved or grafted ovaries, anti-Müllerian hormone (AMH), which inhibits ovarian follicle recruitment, was used in a mouse model. In experiment 1, ovaries were vitrified warmed with different doses of AMH (0, 5, 15, or 45 µg/mL) supplementation. In experiment 2, AMH (0, 50, 250, and 1250 µg/mL) was injected into mice before and/or after cryopreserved ovary autotransplantation, and the recipients remained for 7 or 28 days after grafting. Ovaries were evaluated by follicle morphology, density, and apoptosis ratio. Additionally, serum follicle-stimulating hormone was measured in experiment 2. Significantly decreased follicle apoptosis were detected in AMH-treated groups when compared to the control ovaries in experiment 1, meanwhile no positive effect of exogenous AMH was found in experiment 2. Thus, we suggest AMH supplementation during ovary vitrification warming has beneficial effect on reducing follicle apoptosis.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Apoptosis/drug effects , Ovary/drug effects , Animals , Anti-Mullerian Hormone/administration & dosage , Cryopreservation , Dose-Response Relationship, Drug , Female , Follicle Stimulating Hormone/blood , Mice , Ovarian Follicle/drug effects , Ovary/transplantation , Vitrification
15.
Fertil Steril ; 102(6): 1633-40.e5, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25256937

ABSTRACT

OBJECTIVE: To evaluate the dose-response relationship of a novel recombinant human FSH (rhFSH; FE 999049) with respect to ovarian response in patients undergoing IVF/intracytoplasmic sperm injection treatment; and prospectively study the influence of initial antimüllerian hormone (AMH) concentrations. DESIGN: Randomized, controlled, assessor-blinded, AMH-stratified (low: 5.0-14.9 pmol/L [0.7-<2.1 ng/mL]; high: 15.0-44.9 pmol/L [2.1-6.3 ng/mL]) trial. SETTING: Seven infertility centers in four countries. PATIENT(S): Two hundred sixty-five women aged ≤37 years. INTERVENTION(S): Controlled ovarian stimulation with either 5.2, 6.9, 8.6, 10.3, or 12.1 µg of rhFSH, or 11 µg (150 IU) of follitropin alfa in a GnRH antagonist cycle. MAIN OUTCOME MEASURE(S): Number of oocytes retrieved. RESULT(S): The number of oocytes retrieved increased in an rhFSH dose-dependent manner, from 5.2 ± 3.3 oocytes with 5.2 µg/d to 12.2 ± 5.9 with 12.1 µg/d. The slopes of the rhFSH dose-response curves differed significantly between the two AMH strata, demonstrating that a 10% increase in dose resulted in 0.5 (95% confidence interval 0.2-0.7) and 1.0 (95% confidence interval 0.7-1.3) more oocytes in the low and high AMH stratum, respectively. Fertilization rate and blastocyst/oocyte ratio decreased significantly with increasing rhFSH doses in both AMH strata. No linear relationship was observed between rhFSH dose and number of blastocysts overall or by AMH strata. Five cases of ovarian hyperstimulation syndrome were reported for the three highest rhFSH doses and in the high AMH stratum. CONCLUSION(S): Increasing rhFSH doses results in a linear increase in number of oocytes retrieved in an AMH-dependent manner. The availability of blastocysts is less influenced by the rhFSH dose and AMH level. CLINICAL TRIAL REGISTRATION NUMBER: NCT01426386.


Subject(s)
Anti-Mullerian Hormone/administration & dosage , Follicle Stimulating Hormone, Human/administration & dosage , Oocyte Retrieval , Adult , Female , Fertilization in Vitro/methods , Humans , Ovulation Induction/methods , Recombinant Proteins/administration & dosage , Sperm Injections, Intracytoplasmic/methods , Treatment Outcome
16.
PLoS One ; 9(6): e99393, 2014.
Article in English | MEDLINE | ID: mdl-24932501

ABSTRACT

Anti-mullerian hormone (AMH) is thought to reflect the growth of follicles and the ovarian function. However, the role of AMH in culture medium during in vitro maturation (IVM) on oocyte quality and subsequent development potential is unclear. The objective of this study is to investigate the effect of recombinant human AMH (rh-AMH) supplemented into IVM medium on oocyte quality. Cumulus-oocyte complexes (COCs) were obtained from ICR mice and cultured in vitro with the different concentrations (0-1,000 ng/ml) of rh-AMH. Following 16-18 h of culture, quantitative PCR and ELISA were performed to analyze GDF9 and BMP15 mRNA expression and protein production from the oocytes. Subsequently, in vitro fertilization (IVF) and early embryonic development were employed to further evaluate the quality of in vitro matured oocytes. The results showed that AMH was only expressed in cumulus cells but not in the oocytes. However, AMH most specific receptor, AMHR-II, was expressed in both oocytes and cumulus cells. The levels of GDF9 and BMP15 expression and blastocyst formation rate were significantly increased (p<0.05) when the IVM medium was supplemented with 100 ng/ml of rh-AMH. With AdH1-SiRNA/AMH for knocking down of AMH expression during IVM significantly reduced (p<0.05) the levels of GDF9 and BMP15 expression and blastocysts formation rate. These results suggest that AHM improves oocytes quality by up-regulating GDF9 and BMP15 expressions during IVM.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Oocytes/drug effects , Animals , Anti-Mullerian Hormone/administration & dosage , Anti-Mullerian Hormone/biosynthesis , Blastocyst/drug effects , Bone Morphogenetic Protein 15/biosynthesis , Bone Morphogenetic Protein 15/genetics , Cells, Cultured , Culture Media/pharmacology , Cumulus Cells/drug effects , Cumulus Cells/metabolism , Dose-Response Relationship, Drug , Embryonic Development , Female , Fertilization in Vitro , Gene Knockdown Techniques , Growth Differentiation Factor 9/biosynthesis , Growth Differentiation Factor 9/genetics , Mice , Mice, Inbred ICR , Oocytes/cytology , Oocytes/metabolism , Polymerase Chain Reaction , RNA, Small Interfering/pharmacology , Receptors, Peptide/biosynthesis , Receptors, Peptide/genetics , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/genetics , Recombinant Proteins/pharmacology
17.
J Cancer Surviv ; 8(4): 548-54, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24810980

ABSTRACT

PURPOSE: Anti-Müllerian hormone (AMH) is an indicator of oocyte reserve in healthy females. The role of AMH testing in oncology remains investigational, although its sensitivity and stability over the menstrual cycle make it an attractive screening test for fertility assessment among female cancer survivors. We measured AMH level in survivors of childhood cancer and evaluated its association with treatment and patient factors. METHODS: Participants were adult female survivors of childhood malignancy treated with chemotherapy. Serum AMH was measured at a random day of the menstrual cycle. Multivariate analysis was used to evaluate the association between AMH level, alkylating agent exposure using the cyclophosphamide equivalent dose (CED), and other covariates. RESULTS: Sixty-six females with a median attained age of 23.3 years were eligible for analysis. Median AMH was 25.5 pM (range 0.5-108.0), at a median time of 11.5 years (range 1.4-25.1) since cancer diagnosis. Twenty-three patients (34.8%) had low AMH, including a significant proportion that reported normal menstrual cycles. Compared to ALL survivors, sarcoma survivors had significantly lower AMH levels. Among alkylating agents evaluated, procarbazine had the greatest adverse effect on AMH. In multivariate analysis, higher CED (p = 0.001), older age at diagnosis (p < 0.001), and use of oral contraceptive pills (p = 0.04) remained significantly associated with lower AMH. CONCLUSIONS: Random AMH can reveal evidence of oocyte depletion among female survivors reporting normal cycles, although low AMH should be interpreted cautiously among those taking oral contraception. Age at exposure and CED can aid identification of those more likely to have low AMH, although CED may underestimate the effect of procarbazine on oocyte reserve. IMPLICATIONS FOR CANCER SURVIVORS: Measurement of AMH can reveal apparent depletion of ovarian reserve in female childhood cancer survivors reporting normal menstrual cycles. Sarcoma survivors and those exposed to procarbazine may benefit from targeted AMH evaluation in an outpatient setting, and thereby allow appropriate fertility counseling before the onset of premature ovarian failure. The cyclophosphamide equivalent dose may facilitate comparison of the potential effect of different regimens on fertility.


Subject(s)
Anti-Mullerian Hormone/therapeutic use , Adolescent , Adult , Anti-Mullerian Hormone/administration & dosage , Cross-Sectional Studies , Female , Humans , Neoplasms/mortality , Neoplasms/therapy , Ovarian Reserve , Survivors , Young Adult
18.
J Theor Biol ; 326: 21-35, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23485453

ABSTRACT

A system of 16 non-linear, delay differential equations with 66 parameters is developed to model hormonal regulation of the menstrual cycle of a woman from age 20 to 51. This mechanistic model predicts changes in follicle numbers and reproductive hormones that naturally occur over that time span. In particular, the model illustrates the decline in the pool of primordial follicles from age 20 to menopause as reported in the biological literature. Also, model simulations exhibit a decrease in antimüllerian hormone (AMH) and inhibin B and an increase in FSH with age corresponding to the experimental data. Model simulations using the administration of exogenous AMH show that the transfer of non-growing primordial follicles to the active state can be slowed enough to provide more follicles for development later in life and to cause a delay in the onset of menopause as measured by the number of primordial follicles remaining in the ovaries. Other effects of AMH agonists and antagonists are investigated in the setting of this model.


Subject(s)
Anti-Mullerian Hormone/administration & dosage , Menopause/drug effects , Menstrual Cycle/drug effects , Menstrual Cycle/physiology , Models, Biological , Reproduction/physiology , Adult , Computer Simulation , Female , Humans , Longevity/physiology , Middle Aged , Primary Ovarian Insufficiency/prevention & control , Time Factors , Young Adult
19.
Zhonghua Fu Chan Ke Za Zhi ; 44(3): 191-5, 2009 Mar.
Article in Chinese | MEDLINE | ID: mdl-19570444

ABSTRACT

OBJECTIVE: To investigate the effect of anti-Müllerian hormone (AMH) on hormone secretion and P450 aromatase mRNA expression from cultured human luteinized granulosa cells. METHODS: Human luteinized granulose cells were derived from 10 patients treated by in vitro fertilization-embryo transplantation (IVF-ET) in the Second Affiliated Hospital of Sun Yat-sen University from June to December 2006. Granulose cells were divided into group A, B, C, D, E depending on different concentration of AMH, testosterone group and blank group. 1x10(-7) mol/L testosterone and 1, 5, 10, 20, 50 microg/L AMH were added into the culture medium of group A, B, C, D and E. 1x10(-7) mol/L testosterone was added into the culture medium of testosterone group while no other ingredient was added into the medium of blank group. Estrogen levels in supernates were measured at 24, 48, 72 hours after cell incubation. RT-PCR was performed to detect the P450 aromatase mRNA expression in group B, C, D, E and testosterone group at 72 hours after cell incubation. RESULTS: (1) Estrogen levels in supernates of granulose cell culture at 24, 48, 72 hours were (8.529+/-0.381)x10(4), (10.977+/-0.436)x10(4), (13.309+/-0.506)x10(4) pmol/L in group A, (7.027+/-0.276)x10(4), (9.167+/-0.300)x10(4), (10.794+/-0.555)x10(4) pmol/L in group B, (6.039+/-0.226)x10(4), (7.585+/-0.548)x10(4), (8.797+/-0.518)x10(4) pmol/L in group C, (5.118+/-0.460)x10(4), (5.716+/-0.496)x10(4), (6.205+/-0.667)x10(4) pmol/L in group D, (4.932+/-0.148)x10(4), (5.323+/-0.184)x10(4), (5.629+/-0.212)x10(4) pmol/L in group E. When compared with blank group [(0.001+/-0.001)x10(4), (0.006+/-0.003)x10(4), (0.029+/-0.011)x10(4) pmol/L], the statistical differences were observed in group A, B, C, D, E (P<0.01); when compared with testosterone group [(8.418+/-0.569)x10(4), (10.841+/-0.689)x10(4), (13.301+/-0.637)x10(4) pmol/L], the statistical differences were observed in group B, C, D and E (P<0.01); statistical differences were also observed in group C, D and E when compared with group B, and also group D and E when compared with group C (P<0.01). No significant difference was observed between group D and E (P>0.05). In group A, B, C, D, E and testosterone group, the estrogen levels at 24 hours after cell culture were significantly lower than those at 48 and 72 hours (P<0.01); statistical difference was observed between estrogen levels at 48 and 72 hours (P<0.01). No significant difference was observed among 24, 48 and 72 hours in blank group (P>0.05). (2) Relative ratios of intensity of P450 aromatase/beta-actin at 72 hours of cell culture in group B, C, D and E were 0.6148+/-0.0046, 0.5156+/-0.0012, 0.4698+/-0.0027 and 0.4282+/-0.0017, respectively, which were statistically lower than that in testosterone group (0.8224+/-0.0021, P<0.01); statistical differences were also observed in group C, D and E when compared with group B, and also group D and E when compared with group C (P<0.01). No significant difference was observed between group D and E (P>0.05). CONCLUSION: It is suggested that AMH might affect estrogen synthesis by inhibiting P450 aromatose activity so that lead to hyperandrogenism microenvironment in local ovary.


Subject(s)
Anti-Mullerian Hormone/pharmacology , Aromatase/metabolism , Estradiol/metabolism , Granulosa Cells/drug effects , Granulosa Cells/metabolism , Adult , Anti-Mullerian Hormone/administration & dosage , Aromatase/genetics , Cells, Cultured , Enzyme Activation/drug effects , Female , Humans , Polycystic Ovary Syndrome/physiopathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Testosterone/administration & dosage , Testosterone/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...