1
|
Siristatidis CS, Papapanou M, Maheshwari A, Vaidakis D. In vitro maturation in subfertile women with polycystic ovarian syndrome undergoing assisted reproduction. Cochrane Database Syst Rev 2025; 2:CD006606. [PMID: 39912435 PMCID: PMC11800328 DOI: 10.1002/14651858.cd006606.pub5] [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: 02/07/2025]
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) occurs in 8% to 13% of all women of reproductive age and 50% of women presenting with infertility (i.e. inability to reach a pregnancy after 12 months or more of regular unprotected sexual intercourse). A proportion of these women ultimately need assisted reproductive technology. In vitro fertilisation (IVF)/intracytoplasmic sperm injection (ICSI) are assisted reproduction techniques used to raise the chances of a pregnancy. In women with PCOS, the supra-physiological doses of gonadotrophins used for controlled ovarian hyperstimulation (COH) often result in an exaggerated ovarian response characterised by the development of a large cohort of follicles of uneven quality, retrieval of immature oocytes, and increased risk of ovarian hyperstimulation syndrome (OHSS). A potentially effective intervention for women with PCOS-related infertility involves earlier retrieval of immature oocytes at the germinal-vesicle stage followed by in vitro maturation (IVM). This is the third update of this Cochrane review on the subject (after the last update on 27 June 2018). OBJECTIVES To assess the benefits and harms of IVM followed by IVF or ICSI versus conventional IVF or ICSI among women with PCOS. SEARCH METHODS On 27 February 2023, we searched the Cochrane Gynaecology and Fertility Group Specialised Register of Controlled Trials, CENTRAL, MEDLINE, Embase, and the Open Grey database. We further searched the National Institute for Health and Care Excellence (NICE) fertility assessment and treatment guidelines. We also searched reference lists of relevant papers and Google Scholar for any additional trials. SELECTION CRITERIA We included randomised controlled trials (RCTs) comparing IVM before IVF or ICSI with conventional IVF or ICSI for infertile women with PCOS, irrespective of language and country of origin. DATA COLLECTION AND ANALYSIS Two review authors independently selected studies, assessed the risk of bias, extracted data from studies, and, where needed, attempted to contact the authors for missing data. Our primary outcomes were live birth per woman randomised and miscarriage. We performed statistical analysis using Review Manager. We assessed the certainty of the evidence using GRADE and the risk of bias using the Cochrane RoB 2 tool. MAIN RESULTS We found four published trials suitable for inclusion in this update. The studies involved 810 subfertile women undergoing assisted reproductive technology. Two of four were already included in the previous version of the review, were published as abstracts in international conferences, and were at high risk of bias. The two new studies were at low risk of bias in all domains and in terms of all outcomes. We implemented the random-effects model for the quantitative analyses and restricted the primary analysis to studies at low risk of bias in all domains. We are very uncertain about the effect of IVM or capacitation IVM (a new biphasic IVM system improving the developmental competence of oocytes) on live birth when compared to IVF when a GnRH antagonist protocol was applied (odds ratio (OR) 0.47, 95% confidence interval (CI) 0.17 to 1.32; I2 = 91%; 2 studies, 739 participants; very low-certainty evidence). This suggests that if the chance of live birth following standard IVF is assumed to be 45.7%, then the chance of IVM would be 12.5% to 52.6%. In contrast, IVM or capacitation IVM increases miscarriage per clinical pregnancy (where clinical pregnancy was defined as evidence of a fetal heart beat on ultrasound at seven gestational weeks) in women with PCOS when compared to IVF (OR 1.66, 95% CI 1.02 to 2.70; I2 = 0%; 2 studies, 378 clinical pregnancies; high-certainty evidence). This suggests that if the chance of miscarriage following standard IVF is assumed to be 20.1%, then the chance using IVM would be 20.4% to 40.4%. Results remained similar when using the risk ratio (RR) as the measure of effect. We are uncertain about the effect of IVM or capacitation IVM on clinical pregnancy when compared to IVF when a GnRH antagonist protocol was applied (OR 0.49, 95% CI 0.14 to 1.70; I2 = 94%; 2 studies, 739 participants; very low-certainty evidence). The results were similar after pooling the RRs. IVM or capacitation IVM results in a large reduction in the incidence of moderate or severe OHSS as compared to IVF when a GnRH antagonist protocol was applied (OR 0.08, 95% CI 0.01 to 0.67; I2 = 0%; 2 studies, 739 participants; high-certainty evidence). This suggests that if the incidence of OHSS following IVF is assumed to be 3.5%, then the incidence with IVM would be 0% to 2.4%. Also, there is probably little to no difference in preterm birth between IVM or capacitation IVM and IVF after the application of a GnRH antagonist protocol (OR 0.69, 95% CI 0.31 to 1.52; I² = 45%; 2 studies, 739 participants; moderate-certainty evidence). As for congenital anomalies, one study reported no events, while another showed an uncertain effect of IVM (OR 0.33, 95% CI 0.01 to 8.24; 1 study, 351 participants; low-certainty evidence). Results remained similar when using the RR as the measure of effect. There were no data from any of the studies for cycle cancellation, oocyte fertilisation, or subgroup analyses. AUTHORS' CONCLUSIONS There is continuous scientific interest in IVM, and promising data have been published. Concerning live birth and clinical pregnancy, we are very uncertain about the effect of the technique when compared to IVF after using a GnRH antagonist protocol. In contrast, high-certainty evidence shows that IVM increases miscarriage per clinical pregnancy and reduces the incidence of moderate or severe OHSS in women with PCOS compared to IVF after a GnRH antagonist protocol. Regarding the rest of the outcomes, low- to moderate-certainty evidence showed little to no difference in preterm birth and risk of congenital anomalies between the two modalities. We eagerly anticipate further evidence from high-quality trials in the field (we found five ongoing trials).
Collapse
Affiliation(s)
- Charalampos S Siristatidis
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Michail Papapanou
- Assisted Reproduction Unit, Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Obstetrics, Gynecology, and Reproductive Medicine Working Group, Society of Junior Doctors, Athens, Greece
| | | | - Dennis Vaidakis
- Department of Primary Care and Population Health, Medical School, University of Nicosia, Nicosia, Cyprus
| |
Collapse
|
2
|
Karavani G, Shapira-Nass S, Schachter-Safrai N, Imbar T, Ben-Meir A. Polycystic ovary syndrome and morphokinetic embryonic development: a case-control study evaluating 791 embryos. F&S SCIENCE 2025:S2666-335X(25)00003-5. [PMID: 39837476 DOI: 10.1016/j.xfss.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE To investigate the association between polycystic ovary syndrome (PCOS) and the rate of embryo development, using time-lapse monitoring systems, compared with a control group of women with mechanical (tubal) factor infertility. DESIGN A retrospective case-control study conducted in a university-affiliated in vitro fertilization (IVF) unit. PATIENTS Women with PCOS undergoing IVF treatments and those with non-PCOS controls with tubal factor infertility only. Development morphokinetic milestones were compared and analysis of covariance for time to distinct cell number as well as logistic mixed models to determine predictors for embryos over the 75th percentile was performed. EXPOSURE Not applicable. MAIN OUTCOME MEASURES Embryo development morphokinetic parameters in women with and without PCOS undergoing IVF treatments. RESULTS The study included 791 embryos from 115 women, 364 embryos from 52 women with PCOS and 427 embryos from 63 women with non-PCOS controls with tubal factor infertility. The PCOS group was 4 years younger (30.07 ± 6.03 vs. 34.08 ± 4.84 years) and had higher number of oocytes retrieved (16.00 vs. 11.00), mature oocytes (11.00 vs. 7.00) and fertilized oocytes (8.00 vs. 5.00). The PCOS and control groups demonstrated comparable clinical pregnancy rates (55.8% vs. 32.1%), miscarriage rate (12.5% vs. 11.8%), and live birth rate (48.8% vs. 31.2%). Morphokinetic parameters were comparable between the groups. Although age was associated with later time to 5 and 8 discrete cells and start of blastulation (tSB), PCOS was only associated with later tSB, including tSB >75th percentile. CONCLUSION This study demonstrated comparable IVF outcomes in women with PCOS and non-PCOS controls. An analysis of time-lapse monitoring data from these patients showed no evidence that PCOS negatively affects embryonic development rate in women undergoing IVF cycles.
Collapse
Affiliation(s)
- Gilad Karavani
- In-Vitro Fertilization Unit, Department of Obstetrics and Gynecology, Hadassah Ein-Kerem Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| | | | | | - Tal Imbar
- In-Vitro Fertilization Unit, Department of Obstetrics and Gynecology, Hadassah Ein-Kerem Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Assaf Ben-Meir
- In-Vitro Fertilization Unit, Department of Obstetrics and Gynecology, Hadassah Ein-Kerem Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Fairtility Ltd., Tel Aviv, Israel
| |
Collapse
|
3
|
Palomba S, Molinaro M, Ingargiola A, Aversa A. Recent developments in drug treatment strategies for infertility in patients with polycystic ovary syndrome. Expert Opin Pharmacother 2024; 25:2191-2202. [PMID: 39428640 DOI: 10.1080/14656566.2024.2418985] [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: 08/17/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION Infertility related to polycystic ovary syndrome (PCOS) represents a significant challenge for women of reproductive age. Over the last few years, evidence-based medicine has driven new approaches for treating infertility in patients with PCOS, changing rapidly and deeply the clinical practice. AREAS COVERED The authors provide an in-depth examination of recent developments in drug treatment strategies that have impacted the clinical practice and changed the previous approach to infertility in patients with PCOS. EXPERT OPINION The authors identify four primary areas of interest that have impacted clinical practice in the last few years. Specifically, they discuss the current role of metformin administration in women with PCOS and infertility, the choice for using clomiphene citrate or letrozole as first-line treatment for ovulation induction, the use of new gonadotropin formulations for in vitro fertilization (IVF) program, and the elective embryo transfer in IVF cycles as golden standard treatment for patients with PCOS at high-risk for ovarian hyperstimulation syndrome.
Collapse
Affiliation(s)
- Stefano Palomba
- Department of Medical-Surgical Science and Translational Medicine, University Sapienza of Rome, Rome, Italy
- Unit of Obstetrics and Gynecology, Grande Ospedale Metropolitano of Reggio Calabria, Reggio Calabria, Italy
| | - Marianna Molinaro
- Department of Endocrinology, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Alice Ingargiola
- Department of Endocrinology, University Magna Graecia of Catanzaro, Catanzaro, Italy
| | - Antonio Aversa
- Department of Endocrinology, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
4
|
Kobayashi H, Shigetomi H, Matsubara S, Yoshimoto C, Imanaka S. Role of the mitophagy-apoptosis axis in the pathogenesis of polycystic ovarian syndrome. J Obstet Gynaecol Res 2024; 50:775-792. [PMID: 38417972 DOI: 10.1111/jog.15916] [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: 01/07/2024] [Accepted: 02/15/2024] [Indexed: 03/01/2024]
Abstract
AIM Polycystic ovary syndrome (PCOS) is a common endocrine disorder characterized by menstrual irregularities, androgen excess, and polycystic ovarian morphology, but its pathogenesis remains largely unknown. This review focuses on how androgen excess influences the molecular basis of energy metabolism, mitochondrial function, and mitophagy in granulosa cells and oocytes, summarizes our current understanding of the pathogenesis of PCOS, and discuss perspectives on future research directions. METHODS A search of PubMed and Google Scholar databases were used to identify relevant studies for this narrative literature review. RESULTS Female offspring born of pregnant animals exposed to androgens recapitulates the PCOS phenotype. Abnormal mitochondrial morphology, altered expression of genes related to glycolysis, mitochondrial biogenesis, fission/fusion dynamics, and mitophagy have been identified in PCOS patients and androgenic animal models. Androgen excess causes uncoupling of the electron transport chain and depletion of the cellular adenosine 5'-triphosphate pool, indicating further impairment of mitochondrial function. A shift toward mitochondrial fission restores mitochondrial quality control mechanisms. However, prolonged mitochondrial fission disrupts autophagy/mitophagy induction due to loss of compensatory reserve for mitochondrial biogenesis. Disruption of compensatory mechanisms that mediate the quality control switch from mitophagy to apoptosis may cause a disease phenotype. Furthermore, genetic predisposition, altered expression of genes related to glycolysis and oxidative phosphorylation, or a combination of these factors may also contribute to the development of PCOS. CONCLUSION In conclusion, fetuses exposed to a hyperandrogenemic intrauterine environment may cause the PCOS phenotype possibly through disruption of the compensatory regulation of the mitophagy-apoptosis axis.
Collapse
Affiliation(s)
- Hiroshi Kobayashi
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Hiroshi Shigetomi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Gynecology and Reproductive Medicine, Aska Ladies Clinic, Nara, Japan
| | - Sho Matsubara
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Medicine, Kei Oushin Clinic, Nishinomiya, Japan
| | - Chiharu Yoshimoto
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Prefecture General Medical Center, Nara, Japan
| | - Shogo Imanaka
- Department of Gynecology and Reproductive Medicine, Ms.Clinic MayOne, Kashihara, Japan
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| |
Collapse
|
5
|
Ban M, Sun Y, Chen X, Zhou X, Zhang Y, Cui L. Association between maternal polycystic ovarian syndrome undergoing assisted reproductive technology and pregnancy complications and neonatal outcomes: a systematic review and meta-analysis. J Ovarian Res 2024; 17:6. [PMID: 38184624 PMCID: PMC10770902 DOI: 10.1186/s13048-023-01331-x] [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: 07/27/2023] [Accepted: 12/17/2023] [Indexed: 01/08/2024] Open
Abstract
BACKGROUND Polycystic ovarian syndrome (PCOS) is recognized as the most prevalent endocrine disorder among women of reproductive age. While the utilization of assisted reproductive technology (ART) has resulted in favorable outcomes for infertility treatment in PCOS patients, the inherent pathophysiological features of the condition give rise to complications and consequences during pregnancy and delivery for both the mother and offspring. This study was to assess the correlation between maternal PCOS and various pregnancy complications and neonatal outcomes undergone ART. METHODS A systematic search was conducted on PubMed, EmBase, and the Cochrane Library to identify observational studies that investigated the association between PCOS and the risk of various pregnancy complications and neonatal outcomes, including gestational diabetes mellitus (GDM), hypertension in pregnancy (PIH), preeclampsia (PE), preterm birth, abortion, congenital malformations (CA), small for gestational age (SGA), large for gestational age (LGA), low birth weight (LBW), macrosomia, neonatal intensive care unit (NICU) admission and birth weight. Eligible studies were selected based on predetermined inclusion and exclusion criteria. The meta-analysis was conducted using Review Manager and Stata software, with odds ratios (ORs) or mean difference (MD), confidence intervals (CIs), and heterogeneity (I2) being calculated. The search was conducted up to March 2023. RESULTS A total of 33 studies with a combined sample size of 92,810 participants were identified. The findings indicate that PCOS is significantly associated with an increased risk of GDM (OR 1.51, 95% CI:1.17-1.94), PIH (OR 1.72, 95% CI:1.25-2.39), PE (OR 2.12, 95% CI:1.49-3.02), preterm birth (OR 1.29, 95% CI:1.21-1.39), and LBW (OR 1.29, 95% CI:1.14-1.47). In subgroup analyses, the risks of GDM (OR 1.80, 95% CI:1.23-2.62) and abortion (OR 1.41, 95% CI:1.08-1.84) were elevated in fresh embryo transferred (ET) subgroup, whereas elevated risk of PE (OR 1.82, 95% CI:1.17-2.83) and preterm birth (OR 1.31, 95% CI:1.21-1.42) was identified in frozen ET subgroup. Whatever with or without hyperandrogenism, patients with PCOS had a higher risk in preterm birth (OR 1.69, 95% CI: 1.31-2.18; OR 1.24, 95% CI:1.02-1.50) and abortion (OR 1.38, 95% CI:1.12-1.71; OR 1.23, 95% CI:1.06-1.43). CONCLUSION Our findings suggest that individuals with PCOS undergone ART are at a notably elevated risk for experiencing pregnancy complications and unfavorable neonatal outcomes. Nevertheless, to establish a definitive association between PCOS and pregnancy-related outcomes, it is necessary to conduct extensive prospective, blinded cohort studies and effectively control for confounding variables.
Collapse
Affiliation(s)
- Miaomiao Ban
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yifei Sun
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Xiaojing Chen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Xiaoqian Zhou
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Yiyuan Zhang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China
| | - Linlin Cui
- Center for Reproductive Medicine, the Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences, No.2021RU001), Jinan, 250012, Shandong, China.
- Key laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250012, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, 250012, Shandong, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, 250012, Shandong, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
6
|
Eralp B, Ibanoglu MC, Engin-Ustun Y. Evaluation of pregnancy and neonatal outcomes according to the phenotypic types of polycystic ovary syndrome: A prospective study. Int J Gynaecol Obstet 2023; 163:894-903. [PMID: 37278384 DOI: 10.1002/ijgo.14882] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/14/2023] [Accepted: 05/06/2023] [Indexed: 06/07/2023]
Abstract
OBJECTIVE We aimed to compare pregnancy and neonatal outcomes in the phenotypic subtypes of patients with polycystic ovary syndrome (PCOS). METHODS This prospective cohort included the patients with PCOS (n = 121) diagnosed according to the presence of androgen excess, ovulatory dysfunction, and/or polycystic ovary morphology and healthy controls (n = 125). We stratified PCOS as phenotype A (n = 45), phenotype B (n = 8), phenotype C (n = 32) and phenotype D (n = 35) and followed throughout pregnancy, comparing their outcomes. RESULTS The study population had a mean age of 28.7 ± 4.9 years and a mean BMI of 31.6 kg/m2 with no difference between the groups. Primary cesarean deliveries were significantly more common in PCOS patients (23.3%) than in the control group (17.6%, P = 0.021). The phenotype A group had significantly higher rates of gestational diabetes mellitus (GDM) (42.2%, P < 0.001) and fetal macrosomia (14.6%, P = 0.002) compared with the control group (4.8% and 0.8%m respectively). We detected a significantly lower rate of normal risk score on the double screening test in the PCOS group (59.0%) than in the control group (75.4%) and in the other groups (P = 0.001). CONCLUSION The rates of GDM, fetal macrosomia, and cesarean section were higher in the PCOS group, depending on the phenotype. We observed changes in risk calculation according to phenotypic types at aneuploidy screening.
Collapse
Affiliation(s)
- Berat Eralp
- Department of Gynecology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Mujde Can Ibanoglu
- Department of Gynecology, Ankara Etlik Zubeyde Hanım Women's Health Training and Research Hospital, Ankara, Turkey
| | - Yaprak Engin-Ustun
- Department of Gynecology, Ankara Etlik Zubeyde Hanım Women's Health Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
7
|
Danfeng D, Ke D, Dengxuan F, Xuelian L, Congjian X. Oocyte quality is impaired in a hyperandrogenic PCOS mouse model by increased Foxo1 expression. Reprod Biol 2023; 23:100812. [PMID: 37806115 DOI: 10.1016/j.repbio.2023.100812] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/10/2023]
Abstract
One of the most important characteristics of patients with polycystic ovary syndrome (PCOS) is excess androgen, which has adverse effects on pregnancy outcomes and increases the risk of offspring developing metabolic disorders. Foxo1 has been shown to play an important role in PCOS, but whether it has an affect on oocyte's quality in PCOS remains unclear. The current research investigated the effect of excess androgen exposure on mouse oocyte quality, as well as the possible molecular mechanism. Timelapse incubator was used to culture oocytes in vitro and evaluate the maturation process. The level of reactive oxygen species (ROS) and mitochondrial membrane potential were detected by laser confocal microscope. Immunofluorescence staining assays were performed to examine the expression of Foxo1 and γ-H2AX. Relative mRNA level of Foxo1 and Caspase3 were examined by RT-qPCR. Results showed Germinal vesicle breakdown and maturation rates of oocytes from hyperandrogenic PCOS mice were significantly decreased in vitro, while in vitro maturation showed a marked delay from the germinal vesicle breakdown to metaphase II stage in PCOS group. Expression levels of reactive oxygen species, Foxo1, Caspase3, and γ-H2AX were significantly increased, whereas mitochondrial membrane potential was significantly decreased in oocytes from PCOS mice. These results indicate that excess androgen exposure induced oxidative stress, which further induced high expression of Foxo1, resulting in more DNA damage and apoptosis in oocytes. The current findings provide new knowledge for exploring the mechanism of decreased oocyte quality in hyperandrogenic PCOS.
Collapse
Affiliation(s)
- Du Danfeng
- Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Deng Ke
- Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Fan Dengxuan
- Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China
| | - Li Xuelian
- Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China.
| | - Xu Congjian
- Obstetrics & Gynecology Hospital, Fudan University, Shanghai, China; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Shanghai, China.
| |
Collapse
|
8
|
Matorras R, Pijoan JI, Laínz L, Díaz-Nuñez M, Sainz H, Pérez-Fernandez S, Moreira D. Polycystic ovarian syndrome and miscarriage in IVF: systematic revision of the literature and meta-analysis. Arch Gynecol Obstet 2023; 308:363-377. [PMID: 36058943 DOI: 10.1007/s00404-022-06757-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/16/2022] [Indexed: 11/02/2022]
Abstract
PURPOSE To evaluate the risk of miscarriage in IVF cycles in women with PCOS. METHODS Systematic review and meta-analysis. Systematic search of MEDLINE, EMBASE and Google Scholar. The language search was restricted to English, Spanish and French, from 2000 to 2019, with crosschecking of references from relevant articles. Inclusion criteria were: (1) IVF cycles (2) a group of patients with PCOS was considered separately, (3) the miscarriage rate was reported, (4) there was a control group, (5) definition of PCOS according the Rotterdam criteria. Exclusion criteria were been excluded from the meta-analysis: (1) publication prior to the year 2000, (2) animal studies, (3) reviews, (4) abstracts or conference papers, (5) letters, (6) case reports, (7) studies comparing different IVF techniques, (8) studies comparing groups with and without metformin or other treatments, (9) studies on induced abortions. Risk of bias was assessed by the Newcastle-Ottawa score (NOS). All the included studies had a low risk of bias (NOS scores ranging 7-8). The review protocol was registered in PROSPERO (CRD42020186713). Seventeen studies were included in the meta-analysis. There was a total of 10,472 pregnancies (2650 in PCOS and 7822 in controls) of which 1885 were miscarriages (682 in PCOS and 1203 in controls). We considered the miscarriage rate (MR), preclinical MR, early MR, and late MR. RESULTS In IVF pregnancies the risk of miscarriage was significantly increased when considering miscarriages in total (RR = 1.59; CI = 1.45-1.75), preclinical miscarriages (RR = 1.59; CI = 1.35-1.88), and early miscarriages (RR = 1.44; CI = 1.16-1.79). The increased miscarriage rate persisted in Chinese and Western populations when considered separately. The risk of miscarriage was increased in the subgroup of fresh transfers (RR = 1.21; CI = 1.06-1.39) as well as in the subgroup including either fresh or frozen transfers (RR = 1.95; CI = 1.72-2.22). CONCLUSION PCOS is linked to an increased MR in IVF pregnancies both of miscarriages in total, and to an increase in preclinical and early miscarriages. PROSPERO NUMBER CRD42020186713.
Collapse
Affiliation(s)
- Roberto Matorras
- Human Reproduction Unit, Hospital de Cruces, Biocruces, Baracaldo, Vizcaya, Spain.
- IVIRMA. IVI Bilbao, Bizkaia, Spain.
| | - Jose Ignacio Pijoan
- Clinical Epidemiology Unit, Cruces University Hospital, Biocruces, Baracaldo, Vizcaya, Spain
| | - Lucía Laínz
- Human Reproduction Unit, Hospital de Cruces, Biocruces, Baracaldo, Vizcaya, Spain
| | - María Díaz-Nuñez
- Human Reproduction Unit, Hospital de Cruces, Biocruces, Baracaldo, Vizcaya, Spain
| | - Héctor Sainz
- Human Reproduction Unit, Hospital de Cruces, Biocruces, Baracaldo, Vizcaya, Spain
| | - Silvia Pérez-Fernandez
- Clinical Epidemiology Unit, Cruces University Hospital, Biocruces, Baracaldo, Vizcaya, Spain
| | - Dayana Moreira
- Human Reproduction Unit, Hospital de Cruces, Biocruces, Baracaldo, Vizcaya, Spain
| |
Collapse
|
9
|
Potdar N, Iyasere C. Early pregnancy complications including recurrent pregnancy loss and obesity. Best Pract Res Clin Obstet Gynaecol 2023; 90:102372. [PMID: 37451193 DOI: 10.1016/j.bpobgyn.2023.102372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/31/2023] [Accepted: 06/05/2023] [Indexed: 07/18/2023]
Abstract
This review on early pregnancy complications and obesity will focus on the known pregnancy complications such as miscarriage (whether spontaneous or after fertility treatment), polycystic ovaries and risk of miscarriage, recurrent pregnancy loss, ectopic pregnancy, hyperemesis gravidarum and birth defects. Evidence will be assessed and mechanistic pathways for the outcomes will be described. We know that obesity is now a pandemic and has an impact on early pregnancy complications. The evidence has been summarised to provide the reader with a comprehensive overview and advice for pregnant women with obesity in early pregnancy.
Collapse
Affiliation(s)
- Neelam Potdar
- Department of Reproductive Medicine, University Hospitals of Leicester NHS Trust, Leicester Royal Infirmary, LE1 5WW, UK; Department of Health Sciences, University of Leicester, UK.
| | - Cecilia Iyasere
- Department of Reproductive Medicine, University Hospitals of Leicester NHS Trust, Leicester Royal Infirmary, LE1 5WW, UK; Department of Health Sciences, University of Leicester, UK
| |
Collapse
|
10
|
Zhao YK, Gao YN, Wang LC, Wang J, Wang GJ, Wu HL. Correlation between abnormal energy metabolism of ovarian granulosa cells and in vitro fertilization-embryo transfer outcomes in patients with polycystic ovary syndrome and obesity. J Ovarian Res 2023; 16:145. [PMID: 37480140 PMCID: PMC10362761 DOI: 10.1186/s13048-023-01204-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/16/2023] [Indexed: 07/23/2023] Open
Abstract
CONTEXT Granulosa cells (GCs) that surround oocytes in mammalian reproduction play an active role in oocyte differentiation through proliferation and energy production. AIMS This study aimed to investigate the characteristics of the energy metabolism of ovarian GCs and the influence of GCs on the early embryonic development in polycystic ovary syndrome (PCOS). METHODS The clinical characteristics and in vitro fertilization-embryo transfer treatment outcomes of 39 patients with PCOS and 68 patients with simple tubal factor infertility who underwent controlled ovarian hyperstimulation were analyzed and summarized. The mitochondrial function and glucose metabolism level of the GCs were determined, as well as the content of oxidative stress markers in the follicular fluid (FF) of patients with and without PCOS. KEY RESULTS When compared to the non-PCOS group, patients with PCOS had a significantly increased number of retrieved oocytes but a significantly decreased number of high-quality embryos, available embryos, and high-quality blastocyst formation (P < 0.05). Furthermore, the mitochondrial membrane potential, adenosine triphosphate level, and mitochondrial DNA (mtDNA) copy number decreased in the GCs, whereas the levels of reactive oxygen species increased (P < 0.01). The levels of malondialdehyde and 8-oxo-deoxyguanosine (8-OHdG) in the follicular fluid (FF) of the patients with PCOS were higher than those of the control group (P < 0.05), and superoxide dismutase was increased by compensation (P < 0.05). In the PCOS group, the expressions of GLUT1, LDHA, and PFKP were lower than those in the non-PCOS group, and glucose levels were higher. CONCLUSIONS The low oocyte competence of PCOS may be associated with mitochondrial dysfunction and abnormal glycolysis. IMPLICATIONS This research offers explanations for the possible connections influencing human ovarian folliculogenesis.
Collapse
Affiliation(s)
- Ya-Kun Zhao
- Department of Obstetrics and Gynecology, Affiliated Hospital of Hebei University, NO. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei, China
| | - Ya-Na Gao
- Department of Obstetrics and Gynecology, Affiliated Hospital of Hebei University, NO. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei, China
| | - Ling-Chao Wang
- 66350 Medical Company of PLA, Baoding, 071000, Hebei, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Hebei University, NO. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei, China.
| | - Gai-Jing Wang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Hebei University, NO. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei, China
| | - Hong-Li Wu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Hebei University, NO. 212 Yuhua East Road, Lianchi District, Baoding, 071000, Hebei, China
| |
Collapse
|
11
|
Zhu K, Chen Y, Sang Y, Chen Q, Wang G, Zhu B, Lin T, Mao L, Zhu Y. Serum steroid metabolome on the day of oocyte retrieval in women with polycystic ovarian syndrome and its association with pregnancy outcome of in vitro fertilization. J Steroid Biochem Mol Biol 2023; 231:106311. [PMID: 37060931 DOI: 10.1016/j.jsbmb.2023.106311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/17/2023]
Abstract
Steroid hormone level is a crucial factor affecting the outcomes of in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI). The purpose of this study was to evaluate serum steroid metabolome on the day of oocyte retrieval in women with polycystic ovarian syndrome (PCOS) and explore whether specific steroids can be potential indicators to improve the prediction of pregnancy outcomes in PCOS patients undergoing IVF/ICSI. In this study, the serum levels of 21 steroids in 89 women with PCOS and 73 control women without PCOS on the day of oocyte retrieval of the first IVF/ICSI treatment cycle were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS). All patients subsequently received good-quality embryo transfer, and the correlation between their steroid profiles and pregnancy outcomes of the first embryo transfer (ET) was retrospectively analyzed. We found PCOS patients had aberrant levels of 11 out of 21 steroid hormones compared to control individuals, with androgen steroid hormones being considerably enhanced. Enzyme activity evaluation indicated that PCOS women might have abnormal activity of CYP17A1, CYP21A2, CYP11B2, CYP19A1, HSD3B, HSD11B, and HSD17B. Additionally, the level of 18-hydroxycorticosterone (p = 0.014), corticosterone (p = 0.035), and 17-hydroxypregnenolone (p = 0.005) were markedly higher in live birth group than in non- live birth group for PCOS women following frozen embryo transfer (FET). Multiple logistic regressions indicated that 18-hydrocorticosterone and 17-hydroxypregnenolone were independently associated with live birth outcomes of PCOS women following FET. Receiver operating characteristic (ROC) curve analysis revealed that 0.595ng/mL for 18-hydrocorticosterone level (AUC: 0.6936, p = 0.014).and 2.829ng/mL for 17-hydroxypregnenolone level (AUC: 0.7215, p = 0.005) were the best cutoff values to predict live birth outcomes of PCOS. In conclusion, the blood steroid metabolome was closely related to the IVF/ICSI outcomes of PCOS patients. 18-hydroxycorticosterone and 17-hydroxypregnenolone might be potential indicators to predict pregnancy outcomes of PCOS undergoing IVF/ICSI treatment. AVAILABILITY OF DATA AND MATERIALS: The data used in the current study are available from the database of Women's Hospital, School of Medicine, Zhejiang University on reasonable request.
Collapse
Affiliation(s)
- Kai Zhu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, Zhejiang, China; Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China
| | - Yunwen Chen
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, Zhejiang, China; Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China
| | - Yimiao Sang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, Zhejiang, China; Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China
| | - Qingqing Chen
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, Zhejiang, China; Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China
| | - Guiquan Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, Zhejiang, China; Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China
| | - Bo Zhu
- Department of Clinical Laboratory, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Tingting Lin
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, Zhejiang, China; Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China
| | - Luna Mao
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, Zhejiang, China; Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China
| | - Yimin Zhu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, Zhejiang, China; Key Laboratory of Reproductive Genetics (Ministry of Education) and Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, Zhejiang, China.
| |
Collapse
|
12
|
Nakanishi N, Osuka S, Kono T, Kobayashi H, Ikeda S, Bayasula B, Sonehara R, Murakami M, Yoshita S, Miyake N, Muraoka A, Kasahara Y, Murase T, Nakamura T, Goto M, Iwase A, Kajiyama H. Upregulated Ribosomal Pathway Impairs Follicle Development in a Polycystic Ovary Syndrome Mouse Model: Differential Gene Expression Analysis of Oocytes. Reprod Sci 2023; 30:1306-1315. [PMID: 36194357 DOI: 10.1007/s43032-022-01095-7] [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: 04/25/2022] [Accepted: 09/21/2022] [Indexed: 10/10/2022]
Abstract
Polycystic ovary syndrome (PCOS), a common endocrine disorder, is associated with impaired oocyte development, leading to infertility. However, the pathogenesis of PCOS has not been completely elucidated. This study aimed to determine the differentially expressed genes (DEGs) and epigenetic changes in the oocytes from a PCOS mouse model to identify the etiological factors. RNA-sequencing analysis revealed that 90 DEGs were upregulated and 27 DEGs were downregulated in mice with PCOS compared with control mice. DNA methylation analysis revealed 30 hypomethylated and 10 hypermethylated regions in the PCOS group. However, the DNA methylation status did not correlate with differential gene expression. The pathway enrichment analysis revealed that five DEGs (Rps21, Rpl36, Rpl36a, Rpl37a, and Rpl22l1) were enriched in ribosome-related pathways in the oocytes of mice with PCOS, and the immunohistochemical analysis revealed significantly upregulated expression levels of Rps21 and Rpl36. These results suggest that differential gene expression in the oocytes of mice in PCOS is related to impaired folliculogenesis. These findings improve our understanding of PCOS pathogenesis.
Collapse
Affiliation(s)
- Natsuki Nakanishi
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
- Department of Maternal and Perinatal Medicine, Nagoya University Hospital, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Satoko Osuka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomohiro Kono
- Department of Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Hisato Kobayashi
- Department of Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Shinya Ikeda
- Department of Bioscience, Tokyo University of Agriculture, 1-1-1 Sakuragaoka, Setagaya-ku, Tokyo, 156-8502, Japan
| | - Bayasula Bayasula
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Reina Sonehara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Mayuko Murakami
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Sayako Yoshita
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Natsuki Miyake
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Ayako Muraoka
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yukiyo Kasahara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomohiko Murase
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomoko Nakamura
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Maki Goto
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Akira Iwase
- Department of Obstetrics and Gynecology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, 371-8511, Japan
| | - Hiroaki Kajiyama
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
13
|
Xing J, Qiao G, Luo X, Liu S, Chen S, Ye G, Zhang C, Yi J. Ferredoxin 1 regulates granulosa cell apoptosis and autophagy in polycystic ovary syndrome. Clin Sci (Lond) 2023; 137:453-468. [PMID: 36752638 DOI: 10.1042/cs20220408] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 02/06/2023] [Accepted: 02/08/2023] [Indexed: 02/09/2023]
Abstract
Polycystic ovary syndrome (PCOS), a common reproductive endocrine disorder in women of reproductive age, causes anovulatory infertility. Increased apoptosis of granulosa cells has been identified as one of the key factors contributing to abnormal follicular development. Ferredoxin 1 (FDX1) encodes a small ferredoxin that is involved in the reduction in mitochondrial cytochromes and the synthesis of various steroid hormones and has the potential to influence the function of granulosa cells. In the present study, we aimed to determine the relationship between FDX1 and follicular granulosa cell function. To this end, we investigated the difference between FDX1 expression in the granulosa cells of 50 patients with PCOS and that of the controls. Furthermore, we sought to elucidate the role and mechanism of FDX1 in PCOS granulosa cells by establishing a mouse PCOS model with dehydroepiandrosterone and KGN (a steroidogenic human granulosa cell-like tumor cell line). The results indicated significant up-regulation of FDX1 in the granulosa cells after androgen stimulation. Knockdown of FDX1 promoted the proliferation of KGN and inhibited apoptosis. Moreover, FDX1 could regulate autophagy by influencing the autophagy proteins ATG3 and ATG7. Our results demonstrated that FDX1 plays a critical role in female folliculogenesis by mediating apoptosis, autophagy, and proliferation. Therefore, FDX1 may be a potential prognostic factor for female infertility.
Collapse
Affiliation(s)
- Jinshan Xing
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Gan Qiao
- Department of Pharmacology, School of Pharmacy, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Xin Luo
- Department of Pharmacology, School of Pharmacy, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Shuang Liu
- Department of Reproductive Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Shaokun Chen
- Department of Morphological Laboratory, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Geng Ye
- Department of Pharmacology, School of Pharmacy, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Chunxiang Zhang
- Nucleic Acid Medicine of Luzhou Key Laboratory, Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Jingyan Yi
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Nucleic Acid Medicine of Luzhou Key Laboratory, Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan, China
| |
Collapse
|
14
|
Wang DD, Cao JX, Jiang WJ, Hou JW, Yan MH, Sun ZG, Song JY. Comparison of pregnancy outcomes of letrozole-induced frozen-thawed embryo transfer cycles in PCOS women with two different abnormal ovulation patterns: A retrospective cohort study. Medicine (Baltimore) 2023; 102:e33049. [PMID: 36800580 PMCID: PMC9936047 DOI: 10.1097/md.0000000000033049] [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] [Indexed: 02/19/2023] Open
Abstract
No studies have been conducted on the impact of different types of ovulatory dysfunction on the outcomes of frozen-thawed embryo transfers (FETs) in a letrozole-stimulated cycle in women with polycystic ovarian syndrome (PCOS). This study aimed to compare whether pregnancy outcomes of the letrozole-induced protocol in FET cycles differed between oligo-ovulatory and anovulatory women with PCOS. In a retrospective cohort study, women with PCOS who had undergone letrozole-induced FET at a university-affiliated fertility clinic from February 2014 to October 2020 were identified. The primary end point was live birth rate (LBR) per embryo transfer. Propensity score matching and multivariate logistic regression analyses were performed to control for the relevant confounders. A total of 652 women with PCOS undergoing letrozole-induced FET were included in the final analysis. Three hundred sixty-three of these women had oligo-ovulatory periods, while 289 had anovulatory periods. Propensity score matching analysis showed that LBR did not differ between groups (36.8% in oligo-ovulatory group vs 32.8% in anovulatory group, P = .431). Nevertheless, after controlling for potential confounding factors, LBR was significantly lower in anovulatory than oligo-ovulatory women (adjusted odds ratio 1.57, 95% confidence interval 1.08-2.29, P = .018). Furthermore, the pregnancy loss rate among the oligo-ovulatory group remained lower than those among the anovulatory group (adjusted odds ratio 0.23, 95% confidence interval 0.12-0.44, P < .001). Despite adjustment for confounding factors, those with oligo-ovulatory PCOS had a higher LBR and lower pregnancy loss rate compared with those with anovulatory PCOS. This may indicate that when oligo-ovulation is detected, PCOS patients should be intervened in time to conceive as soon as possible. Prospective studies must be conducted in the future to verify our findings.
Collapse
Affiliation(s)
- Dan-Dan Wang
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Jing-Xian Cao
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Wen-Jing Jiang
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Jin-Wei Hou
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Meng-Han Yan
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Zhen-Gao Sun
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
- Reproductive and Genetic Center of Integrated Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
| | - Jing-Yan Song
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
- Reproductive and Genetic Center of Integrated Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, People’s Republic of China
- * Correspondence: Jing-Yan Song, The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, People’s Republic of China (e-mail: )
| |
Collapse
|
15
|
Medenica S, Spoltore ME, Ormazabal P, Marina LV, Sojat AS, Faggiano A, Gnessi L, Mazzilli R, Watanabe M. Female infertility in the era of obesity: The clash of two pandemics or inevitable consequence? Clin Endocrinol (Oxf) 2023; 98:141-152. [PMID: 35644933 PMCID: PMC10084349 DOI: 10.1111/cen.14785] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/07/2022] [Accepted: 05/25/2022] [Indexed: 01/04/2023]
Abstract
Obesity is an epidemic that has led to a rise in the incidence of many comorbidities: among others, reduced fertility is often under-evaluated in clinical practice. The mechanisms underlying the link between reduced fertility and obesity are numerous, with insulin resistance, hyperglycaemia and the frequent coexistence of polycystic ovary syndrome being the most acknowledged. However, several other factors concur, such as gut microbiome alterations, low-grade chronic inflammation and oxidative stress. Not only do women with obesity take longer to conceive, but in vitro fertilization (IVF) is also less likely to succeed. We herein provide an updated state-of-the-art regarding the molecular bases of what we could define as dysmetabolic infertility, focusing on the clinical aspects, as well as possible treatment.
Collapse
Affiliation(s)
- Sanja Medenica
- Department of Internal Medicine, Endocrinology Section, Clinical Center of Montenegro, Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Maria Elena Spoltore
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - Paulina Ormazabal
- Institute of Health Sciences, Universidad de O'Higgins, Rancagua, Chile
- Laboratory of Obesity and Metabolism in Geriatrics and Adults (OMEGA), Institute of Nutrition and Food Technology (INTA), Universidad de Chile, Macul, Santiago, Chile
| | - Ljiljana V Marina
- Department for Obesity, Metabolic and Reproductive Disorders, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Antoan Stefan Sojat
- Department for Obesity, Metabolic and Reproductive Disorders, Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Belgrade, Serbia
| | - Antongiulio Faggiano
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, AOU Sant'Andrea, Rome, Italy
| | - Lucio Gnessi
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| | - Rossella Mazzilli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, AOU Sant'Andrea, Rome, Italy
| | - Mikiko Watanabe
- Department of Experimental Medicine, Section of Medical Pathophysiology, Food Science and Endocrinology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
16
|
Baracat MCP, Baracat EC, Simões RS, Simões MJ, Maciel GAR, Azziz R, Soares JM. Hormonal and Metabolic Factors Influence the Action of Progesterone on the Endometrium of Women with Polycystic Ovary Syndrome. Diagnostics (Basel) 2023; 13:diagnostics13030382. [PMID: 36766487 PMCID: PMC9914468 DOI: 10.3390/diagnostics13030382] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/21/2023] Open
Abstract
Hormonal and metabolic factors may influence endometrial quality and interfere with the action of progesterone. Therefore, the aim of our study was to address this issue. Participants were recruited from an outpatient reproductive endocrinology clinic at an academic tertiary medical care centre. All subjects underwent endometrial biopsy (EB) in the follicular phase of the cycle prior to treatment. Thereafter, they were treated with micronized progesterone (400 mg/day × 10 days intravaginally) from days 14-28 of the next cycle. A second EB was performed between days 21-24 of the cycle (the second phase). The metabolic and hormonal serum levels were evaluated during the implantation window. EB samples were analysed using light microscopy for histomorphometric analysis. The endometrium of women with Polycystic Ovarian Syndrome (PCOS) in the second phase demonstrated a uniform surface epithelium with less leukocyte infiltration and an absence of apoptotic figures compared to the control group. (p < 0.021). The thickness of the surface epithelium in the second phase of the PCOS group correlated positively with free and bioavailable testosterone values. The number of stromal cells increases with increasing insulin levels. Our results suggest that histomorphometric abnormalities of the endometrium persist and are linked to androgen and insulin levels despite progesterone supplementation in PCOS.
Collapse
Affiliation(s)
- Maria Candida P. Baracat
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403 000, Brazil
| | - Edmund C. Baracat
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403 000, Brazil
| | - Ricardo S. Simões
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403 000, Brazil
| | - Manuel J. Simões
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403 000, Brazil
| | - Gustavo A. R. Maciel
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403 000, Brazil
| | - Ricardo Azziz
- Department of Obstetrics & Gynecology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Health Policy, Management, and Behavior, School of Public Health, University at Albany, SUNY, Albany, NY 12222, USA
| | - José Maria Soares
- Disciplina de Ginecologia, Departamento de Obstetrícia e Ginecologia, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, Sao Paulo 05403 000, Brazil
- Correspondence: ; Tel.: +55-(11)-982075781
| |
Collapse
|
17
|
Moini A, Rezaee T, Aleyasin A, Arabipoor A, Moayed ME. The effect of metabolic syndrome on controlled ovarian stimulation outcome in infertile women with polycystic ovary syndrome undergoing assisted reproductive technology cycles. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 67:111-118. [PMID: 36155124 PMCID: PMC9983795 DOI: 10.20945/2359-3997000000518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Objective To evaluate the effect of metabolic syndrome (MetS) diagnosis on oocyte quality and pregnancy outcomes in infertile women with polycystic ovary syndrome (PCOS) who undergoing antagonist-controlled ovarian stimulation (COS) and in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) cycles. Methods This prospective cohort study was conducted from November 2019 to November 2020 across two university-affiliated infertility centers in Iran. The PCOS diagnosis was defined according to the Rotterdam criteria. The patients prior to IVF/ICSI cycles were evaluated for MetS diagnosis. MetS was detected according to the National Cholesterol Education Program/Adult Treatment Panel III with the presence of at least three or more of the specific clinical criteria. The cycle outcomes were compared between MetS and non-MetS groups. Results Overall, 68 eligible infertile PCOS patients with MetS diagnosis and 126 without MetS participated. The MetS diagnosis was associated with the increased requirement of gonadotropins and the COS duration significantly (P = 0.001). Although the total numbers of retrieved and MII oocytes, obtained and topquality embryos as well as clinical pregnancy and live birth rates in the MetS group were lower than those of in the non-MetS group, the differences were not statistically significant (P > 0.05). In followup of the obstetrics complications, the rate of preeclampsia was significantly higher in patients with MetS (P = 0.02). Conclusion MetS diagnosis in PCOS patients was associated with non-significant poor COS and pregnancy outcome. Further studies with larger sample sizes are recommended to clarify the risk of MetS in patients undergoing ART cycles.
Collapse
Affiliation(s)
- Ashraf Moini
- Department of Obstetrics and Gynecology, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Breast Disease Research Center (BDRC), Tehran University of Medical Sciences, Tehran, Iran.,Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Tawoos Rezaee
- Department of Obstetrics and Gynecology, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran, .,Infertility Ward, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ashraf Aleyasin
- Department of Infertility, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Department of Obstetrics and Gynecology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Arezoo Arabipoor
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Marzieh Eslami Moayed
- Department of Obstetrics and Gynecology, Arash Women's Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Gunning MN, Christ JP, van Rijn BB, Koster MPH, Bonsel GJ, Laven JSE, Eijkemans MJC, Fauser BCJM. Predicting pregnancy chances leading to term live birth in oligo/anovulatory women diagnosed with PCOS. Reprod Biomed Online 2023; 46:156-163. [PMID: 36411204 DOI: 10.1016/j.rbmo.2022.09.024] [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: 08/09/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/08/2022]
Abstract
RESEARCH QUESTION Which patient features predict the time to pregnancy (TTP) leading to term live birth in infertile women diagnosed with polycystic ovary syndrome (PCOS)? DESIGN Prospective cohort follow-up study was completed, in which initial standardized phenotyping was conducted at two Dutch university medical centres from January 2004 to January 2014. Data were linked to the Netherlands Perinatal Registry to obtain pregnancy outcomes for each participant. All women underwent treatment according to a standardized protocol, starting with ovulation induction as first-line treatment. Predictors of pregnancies (leading to term live births) during the first year after PCOS diagnosis were evaluated. RESULTS A total of 1779 consecutive women diagnosed with PCOS between January 2004 and January 2014 were included. In the first year following screening, 659 (37%) women with PCOS attained a pregnancy leading to term birth (≥37 weeks of gestational age). A higher chance of pregnancy was associated with race, smoking, body mass index (BMI), insulin, total testosterone and sex hormone-binding globulin (SHBG) concentrations (c-statistic = 0.59). CONCLUSIONS Predictors of an increased chance of a live birth include White race, no current smoking, lower BMI, insulin and total testosterone concentrations, and higher SHBG concentrations. This study presents a nomogram to predict the chances of achieving a pregnancy (leading to a term live birth) within 1 year of treatment.
Collapse
Affiliation(s)
- Marlise N Gunning
- Department of Reproductive Medicine and Gynaecology, University Medical Center Utrecht Utrecht, the Netherlands
| | - Jacob P Christ
- Department of Reproductive Medicine and Gynaecology, University Medical Center Utrecht Utrecht, the Netherlands; Cleveland Clinic Lerner College of Medicine, Cleveland Ohio, USA; Department of Obstetrics & Gynecology, University of Washington Medical Center, SeattleWashington, USA.
| | - Bas B van Rijn
- Department of Obstetrics, University Medical Center Utrecht Utrecht, the Netherlands; Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maria P H Koster
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Gouke J Bonsel
- Department of Obstetrics, University Medical Center Utrecht Utrecht, the Netherlands
| | - Joop S E Laven
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marinus J C Eijkemans
- Department of Reproductive Medicine and Gynaecology, University Medical Center Utrecht Utrecht, the Netherlands; Julius Center for Health Sciences and Primary care, University Medical Center Utrecht Utrecht, the Netherlands
| | - Bart C J M Fauser
- Department of Reproductive Medicine and Gynaecology, University Medical Center Utrecht Utrecht, the Netherlands
| |
Collapse
|
19
|
Liu C, Wang M, Yao H, Cui M, Gong X, Wang L, Sui C, Zhang H. Inhibition of oocyte maturation by follicular extracellular vesicles of non-hyperandrogenic PCOS patients requiring IVF. J Clin Endocrinol Metab 2022; 108:1394-1404. [PMID: 36527699 DOI: 10.1210/clinem/dgac733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
CONTEXT Polycystic ovarian syndrome (PCOS) is one of the most common diseases that contribute to subfertility. Recent evidence showed that oocytes of women with PCOS matured in vitro away from the follicular fluid presented better potentials, whereas the reason remained unclear. OBJECTIVE To investigate whether follicular extracellular vesicles (EVs) of PCOS patients interfere with the quality of oocytes. METHODS Follicular EVs of women with PCOS (PCOS-EVs) and control women (CTRL-EVs) were isolated and determined using western blotting, nanoparticle tracking analysis, and transmission electron microscopy. The two types of EVs were co-cultured with murine germinal vesicle oocytes, respectively. Fluorescence labeled EVs were used to visualize internalization by oocytes. After co-culture, oocyte maturation rates were calculated. Mitochondria distribution and reactive oxygen species (ROS) level were detected in the different groups. Spindle morphology was evaluated using immunofluorescence. Moreover, the expression of catalase (CAT), glutathione synthetase (GSS), and superoxide dismutase (SOD) was determined in the oocytes. RESULTS Both PCOS-EVs and CTRL-EVs are bilayered vesicles, approximately 100-150 nm in size, and enriched in EV-associating protein markers. EVs were internalized by oocytes within one hour. Oocyte maturation rate decreased significantly in the PCOS-EV group compared with the CTRL-EV group; whereas the abnormal mitochondria distribution rate and abnormal spindle rate were significantly increased in the PCOS-EV group. Moreover, PCOS-EVs increased the ROS level and the expression of CAT, GSS, and SOD in the oocytes. CONCLUSIONS PCOS-EVs interfered with oocyte mitochondria and spindles and inhibited oocyte maturation. Moreover, oxidative stress induced by PCOS-EVs might be a potential cause.
Collapse
Affiliation(s)
- Chang Liu
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Meng Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Haixia Yao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
- Shangrao People's Hospital, Shangrao, China
| | - Mengge Cui
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Xueqi Gong
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Lan Wang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Cong Sui
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| | - Hanwang Zhang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology
| |
Collapse
|
20
|
Qiao G, Xing J, Luo X, Zhang C, Yi J. Integrated bioinformatics analysis and screening of hub genes in polycystic ovary syndrome. Endocrine 2022; 78:615-627. [PMID: 36068422 DOI: 10.1007/s12020-022-03181-x] [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: 06/16/2022] [Accepted: 08/23/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) is one of the most common endocrine and metabolic disorders, posing a serious threat to the health of women. Herein, we aimed to explore new biomarkers and potential therapeutic targets for PCOS by employing integrated bioinformatics tools. METHODS Three gene expression profile datasets (GSE138518, GSE155489, GSE106724) were obtained from the Gene Expression Omnibus database and the differentially expressed genes in PCOS and normal groups with an adjusted p-value < 0.05 and a |log fold change (FC) | > 1.2 were first identified using the DESeq package. The weighted correlation network analysis (WGCNA) R package was used to identify clusters of highly correlated genes or modules associated with PCOS. Protein-protein interaction (PPI) network analysis and visualization of genes in the key module were performed using the STRINGdb database and the NetworkX package (edge > 5), respectively. The genes overlapping among the key module genes and PCOS-associated genes were further analyzed. Ligand molecules with strong binding energy < -10 kJ/mol to GNB3 were screened in the drug library using MTiOpenScreen. AutoDock, ChimeraX, and BIOVIA Discovery Studio Visualizer were further used to elucidate the mechanism of ligand interaction with GNB3. Finally, the relationship between GNB3 and PCOS was verified using experimental models in vivo and in vitro. RESULTS Of the 11 modules identified by WGCNA, the black module had the highest correlation with PCOS (correlation = 0.96, P = 0.00016). The PPI network of 351 related genes revealed that VCL, GNB3, MYH11, LMNA, MLLT4, EZH2, PAK3, and CHRM1 have important roles in PCOS. The hub gene GNB3 was identified by taking the intersection of PCOS-related gene sets. MTiOpenScreen revealed that five compounds interacted with GNB3. Of these five, compound 1 had the strongest binding ability and can bind amino acids in the WD40 motif of GNB3, which in turn affects the function of the G protein-coupled receptor β subunit. GNB3 was also significantly downregulated in PCOS models. CONCLUSION We identified the hub gene GNB3 as the most important regulatory gene in PCOS. We suggest that compound 1 can target the WD40 motif of GNB3 to affect related functions and must be considered as a lead compound for drug development. This study will provide new insights into the development of PCOS-related drugs.
Collapse
Affiliation(s)
- Gan Qiao
- Department of Pharmacology, School of Pharmacy, Nucleic Acid Medicine of Luzhou Key Laboratory, Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jinshan Xing
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xin Luo
- Department of Pharmacology, School of Pharmacy, Nucleic Acid Medicine of Luzhou Key Laboratory, Central Nervous System Drug Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chunxiang Zhang
- Nucleic Acid Medicine of Luzhou Key Laboratory, Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Jingyan Yi
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Nucleic Acid Medicine of Luzhou Key Laboratory, Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
21
|
Fitz VW, Mahalingaiah S. Optimization of assisted reproductive technology outcomes in patients with polycystic ovarian syndrome: updates and unanswered questions. Curr Opin Endocrinol Diabetes Obes 2022; 29:547-553. [PMID: 36218224 DOI: 10.1097/med.0000000000000780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW Narrative review of recent literature on optimization of assisted reproduction technology outcomes in patients with polycystic ovarian syndrome (PCOS). RECENT FINDINGS The key areas of focus include pre cycle treatment with the goal of cohort synchronization, methods of ovulation suppression and trigger medication. There is no definitive evidence that precycle treatment with combined oral contraceptives (COCs) or progestins improve or negatively impact in vitro fertilization outcomes in patients with PCOS. The reviewed evidence supports consideration of progestins as suppression of premature ovulation in patients with PCOS as an alternative to gonadotropin releasing hormone (GnRH) antagonist if a freeze all protocol is planned. There is limited prospective evidence in PCOS populations regarding use of a dual trigger using GnRH agonist and human chorionic gonadotropin (hCG). SUMMARY This review has implications for clinical practice regarding ovarian stimulation protocols for patients with PCOS. We also identified areas of research need including the further exploration of the value of pre cycle COC or progestin use in a PCOS population, also the use of GnRH agonist in combination with hCG in a well defined PCOS population and using GnRH agonist trigger alone as a control.
Collapse
Affiliation(s)
- Victoria W Fitz
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Massachusetts General Hospital
| | - Shruthi Mahalingaiah
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Massachusetts General Hospital
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
22
|
Mirza FG, Tahlak MA, Rjeili RB, Hazari K, Ennab F, Hodgman C, Khamis AH, Atiomo W. Polycystic Ovarian Syndrome (PCOS): Does the Challenge End at Conception? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph192214914. [PMID: 36429632 PMCID: PMC9690374 DOI: 10.3390/ijerph192214914] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 05/14/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent condition that not only has the potential to impede conception but also represents the most common endocrine dysfunction in fertile women. It is considered a heterogeneous and multifaceted disorder, with multiple reproductive and metabolic phenotypes which differently affect the early- and long-term syndrome's risks. Undoubtedly, the impact of PCOS on infertility has attracted most of the attention of healthcare providers and investigators. However, there is growing evidence that even after conception is achieved, PCOS predisposes the parturient to several adverse pregnancy outcomes including a high risk of pregnancy-induced hypertension, spontaneous abortion, gestational diabetes, preeclampsia, and preterm birth, which increase the risks of stillbirth and neonatal death. Fetal growth abnormalities may also be more common, but the relationship is less well defined. This narrative review aims to summarize current knowledge regarding these conditions as they interplay with PCOS and concludes that although there appears to be an increase in these complications during the pregnancy of women with PCOS, there is a need for further research to clarify the possible confounding impact of obesity. Implications for clinical practice and future research are outlined.
Collapse
Affiliation(s)
- Fadi G. Mirza
- Latifa Women and Children Hospital, Dubai P.O. Box 9115, United Arab Emirates
- Department of Obstetrics and Gynaecology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14, Dubai Healthcare City, Dubai P.O. Box 505055, United Arab Emirates
- Department of Obstetrics and Gynaecology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Muna A. Tahlak
- Latifa Women and Children Hospital, Dubai P.O. Box 9115, United Arab Emirates
| | - Rachelle Bou Rjeili
- Faculty of Medicine, American University of Beirut, Beruit P.O. Box 11-0236, Lebanon
| | - Komal Hazari
- Latifa Women and Children Hospital, Dubai P.O. Box 9115, United Arab Emirates
| | - Farah Ennab
- Department of Obstetrics and Gynaecology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14, Dubai Healthcare City, Dubai P.O. Box 505055, United Arab Emirates
| | - Charlie Hodgman
- School of Biosciences, University of Nottingham, Loughborough LE12 5RD, UK
| | - Amar Hassan Khamis
- Department of Obstetrics and Gynaecology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14, Dubai Healthcare City, Dubai P.O. Box 505055, United Arab Emirates
| | - William Atiomo
- Department of Obstetrics and Gynaecology, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Building 14, Dubai Healthcare City, Dubai P.O. Box 505055, United Arab Emirates
- Correspondence:
| |
Collapse
|
23
|
Jiang Y, Yuan JC, Song G, Zhang XH, Miao SB, Wu XH. Pregnancy outcome and follow-up of offspring of donor oocytes recipient from PCOS patients. BMC Pregnancy Childbirth 2022; 22:779. [PMID: 36261799 PMCID: PMC9580140 DOI: 10.1186/s12884-022-05114-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 10/11/2022] [Indexed: 11/10/2022] Open
Abstract
Background The use of donated oocytes (DO) for in vitro fertilization(IVF) treatment in patients with infertility is generally recognized, and females with polycystic ovarian syndrome (PCOS) can participate in oocyte donation programs as donor patients. However, the pregnancy outcomes and offspring follow-up in patients with PCOS as the recipients are unclear. This study was to compare the pregnancy outcomes and follow-up of offspring in PCOS and non-PCOS receptor. Methods This was a retrospective cohort study of 62 patients undergoing the oocyte reception program were separated into 2 groups: Group I, PCOS oocyte recipients (n = 30); Group II, non-PCOS recipients (n = 32). Medical records were reviewed, and rates of fertilization, cleavage, high-quality embryos and blastocysts were compared between PCOS and non-PCOS groups. Rates of implantation, pregnancy, ectopic pregnancy, early abortion, multiple pregnancies, and offspring outcomes were calculated using the first single vitrified-warmed blastocyst transfer (SVBT) analysis between the groups. Results The average recipient age and body mass index (BMI) of PCOS and non-PCOS patients was (36.3 ± 2.6 vs. 36.2 ± 2.8, and 23.4 ± 3.9 vs. 23.7 ± 4.0), respectively (P > 0.05). The fertilization, cleavage, high-quality embryos and blastocyst rates were not significantly different between the PCOS and non-PCOS groups. Rates of implantation, pregnancy, ectopic pregnancy, early abortion, and multiple pregnancies were not significantly different in SVBT between the PCOS and non-PCOS groups. The incidence of complications, such as pre-eclampsia or gestational diabetes, between PCOS and non-PCOS groups was similar (11.8% vs.11.1%, 5.9% vs.5.5%; P > 0.05). Preterm births were also similar (11.8% vs.16.7%, P > 0.05). Donor oocytes are more likely to be delivered via cesarean Sect. (80.0% vs. 86.7%: P > 0.05). The mean gestational age, birth weight, and height were comparable between the 2 groups during full-term delivery. Conclusion There was no difference in the pregnancy outcomes and follow-up of the offspring between the PCOS and non-PCOS groups.
Collapse
Affiliation(s)
- Yan Jiang
- The Center for Reproductive Medicine and Infertility, The Fourth Hospital of Shijiazhuang, Shijiazhuang Obstetrics and Gynecology Hospital affiliated to Hebei Medical University, 206 Zhong-Shan-Dong, Shijiazhuang, Hebei, 050011, People's Republic of China.
| | - Jing-Chuan Yuan
- The Center for Reproductive Medicine and Infertility, The Fourth Hospital of Shijiazhuang, Shijiazhuang Obstetrics and Gynecology Hospital affiliated to Hebei Medical University, 206 Zhong-Shan-Dong, Shijiazhuang, Hebei, 050011, People's Republic of China
| | - Ge Song
- The Center for Reproductive Medicine and Infertility, The Fourth Hospital of Shijiazhuang, Shijiazhuang Obstetrics and Gynecology Hospital affiliated to Hebei Medical University, 206 Zhong-Shan-Dong, Shijiazhuang, Hebei, 050011, People's Republic of China
| | - Xu-Hui Zhang
- The Center for Reproductive Medicine and Infertility, The Fourth Hospital of Shijiazhuang, Shijiazhuang Obstetrics and Gynecology Hospital affiliated to Hebei Medical University, 206 Zhong-Shan-Dong, Shijiazhuang, Hebei, 050011, People's Republic of China
| | - Sui-Bing Miao
- The Center for Reproductive Medicine and Infertility, The Fourth Hospital of Shijiazhuang, Shijiazhuang Obstetrics and Gynecology Hospital affiliated to Hebei Medical University, 206 Zhong-Shan-Dong, Shijiazhuang, Hebei, 050011, People's Republic of China
| | - Xiao-Hua Wu
- The Center for Reproductive Medicine and Infertility, The Fourth Hospital of Shijiazhuang, Shijiazhuang Obstetrics and Gynecology Hospital affiliated to Hebei Medical University, 206 Zhong-Shan-Dong, Shijiazhuang, Hebei, 050011, People's Republic of China
| |
Collapse
|
24
|
Bellemare V, Rotshenker-Olshinka K, Nicholls L, Digby A, Pooni A, Kadour-Peero E, Son WY, Dahan MH. Among high responders, is oocyte development potential different in Rotterdam consensus PCOS vs non-PCOS patients undergoing IVF? J Assist Reprod Genet 2022; 39:2311-2316. [PMID: 36029372 PMCID: PMC9596635 DOI: 10.1007/s10815-022-02598-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022] Open
Abstract
PURPOSE To evaluate the oocyte potential to develop to blastocyst in Rotterdam consensus PCOS in women with hyper-responses requiring freeze-all embryos. METHODS Retrospective, single-academic center, cohort study of 205 patients who underwent freeze-all antagonist IVF cycles for OHSS risk between 2013 and 2019. Women in the PCOS group (n = 88) were diagnosed per the 2003 Rotterdam criteria. Control patients (n = 122) had no evidence of hyperandrogenism or menstrual disturbance. Data was compared by t-tests, chi-squared tests, or multivariate logistic regression (SPSS). Frozen blastocysts were Gardner's grade BB or better. RESULTS There was no difference in terms of number of oocytes collected (PCOS vs non-PCOS 27.7 ± 9.4 vs 25.9 ± 8.2, p = 0.157), number of MII (20.7 ± 8.0 vs 19.1 ± 6.6, p = 0.130), number of 2PN fertilized (15.6 ± 7.4 vs 14.4 ± 5.9, p = 0.220), and number of frozen blastocysts (7.8 ± 4.9 vs 7.1 ± 3.8, p = 0.272). In addition, fertilization rates (74 ± 17% vs 75 ± 17%, p = 0.730), blastulation rates per 2PN (51 ± 25% vs 51 ± 25%, p = 0.869), and blastulation rates per mature oocytes (37 ± 18% vs 37 ± 15%, p = 0.984) were all comparable between PCOS and controls, respectively. Moreover, there was no difference when comparing PCOS and controls in pregnancy rates (45/81 vs 77/122, p = 0.28) and clinical pregnancy rates (34/81 vs 54/122, p = 0.75), respectively. Multivariate logistic regression controlling for confounders failed to alter these results. CONCLUSION PCOS subjects do not seem to have altered oocyte potential as measured by number of MII oocytes collected, fertilization, and blastulation rates when compared to high-responder controls, with similar magnitude of stimulation.
Collapse
Affiliation(s)
| | | | - Laura Nicholls
- Fertility Clinic, McGill University Health Center, Montreal, QC, Canada
| | - Alyson Digby
- Fertility Clinic, McGill University Health Center, Montreal, QC, Canada
| | - Amrita Pooni
- Fertility Clinic, McGill University Health Center, Montreal, QC, Canada
| | | | - Weon-Young Son
- Fertility Clinic, McGill University Health Center, Montreal, QC, Canada
| | - Michael H Dahan
- Fertility Clinic, McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
25
|
Acharya KS, Harris BS, Weber JM, Truong T, Pieper C, Eaton JL. Impact of increasing antimüllerian hormone level on in vitro fertilization fresh transfer and live birth rate. F S Rep 2022; 3:223-230. [PMID: 36212572 PMCID: PMC9532892 DOI: 10.1016/j.xfre.2022.06.005] [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: 09/22/2021] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 11/30/2022] Open
Abstract
Objective The objective of our study was to assess the association between AMH and live birth among women with elevated AMH undergoing first fresh IVF. Serum antimüllerian hormone (AMH) correlates with oocyte yield during in vitro fertilization (IVF). However, there are limited data regarding IVF outcomes in women with elevated AMH levels. Design Retrospective cohort study using the Society for Assisted Reproductive Technology Clinical Outcomes Reporting System database from 2012-2014. Setting Fertility clinics reporting to Society for Assisted Reproductive Technology. Patients First, fresh, autologous IVF cycles with elevated AMH levels (≥5.0 ng/mL). Subanalyses were performed to examine patients with or without polycystic ovary syndrome (PCOS). Interventions None. Main Outcome Measures Odds of live birth. Results Our cohort included 10,615 patients with elevated an AMH level, including 2,707 patients with PCOS only. The adjusted odds of live birth per initiated cycle were significantly lower per each unit increase in the AMH level (odds ratio, 0.97; 95% confidence interval, 0.96-0.98). Increasing AMH level was associated with increased cancellation of fresh transfer (odds ratio, 1.12; 95% confidence interval, 1.10-1.15) up to an AMH level of 12 ng/mL. The decrease in the live birth rate appears to be caused by the increasing incidence of cancellation of fresh transfer because the live birth rate per completed transfer was maintained. Similar trends were observed in the PCOS and non-PCOS subanalyses. Conclusions Among patients with AMH levels of ≥5 ng/mL undergoing fresh, autologous IVF, each unit increase in AMH level is associated with a 3% decrease in odds of live birth because of the increased incidence of fresh embryo transfer cancellation.
Collapse
Affiliation(s)
- Kelly S. Acharya
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina
| | - Benjamin S. Harris
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina
| | - Jeremy M. Weber
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | - Tracy Truong
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | - Carl Pieper
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, North Carolina
| | - Jennifer L. Eaton
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina
- Women & Infants Fertility Center, Providence, Rhode Island
| |
Collapse
|
26
|
Zhang Q, Ren J, Wang F, Pan M, Cui L, Li M, Qu F. Mitochondrial and glucose metabolic dysfunctions in granulosa cells induce impaired oocytes of polycystic ovary syndrome through Sirtuin 3. Free Radic Biol Med 2022; 187:1-16. [PMID: 35594990 DOI: 10.1016/j.freeradbiomed.2022.05.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/29/2022] [Accepted: 05/14/2022] [Indexed: 12/23/2022]
Abstract
Mitochondrial function and glucose metabolism play important roles in bidirectional signaling between granulosa cells (GCs) and oocytes. However, the factors associated with mitochondrial function and glucose metabolism of GCs in polycystic ovary syndrome (PCOS) are poorly understood, and their potential downstream effects on oocyte quality are still unknown. The aim of this study was to investigate whether there are alterations in mitochondrial-related functions and glucose metabolism in ovarian GCs of women with PCOS and the role of Sirtuin 3 (SIRT3) in this process. Here, we demonstrated that women with PCOS undergoing in vitro fertilization and embryo transfer had significantly lower rates of metaphase II oocytes, two-pronuclear fertilization, cleavage, and day 3 good-quality embryos. Germinal vesicle- and metaphase I-stage oocytes from women with PCOS exhibited increased mitochondrial reactive oxygen species (ROS), decreased mitochondrial membrane potential, and downregulation of glucose-6-phosphate dehydrogenase. GCs from women with PCOS presented significant alterations in mitochondrial morphology, amount, and localization, decreased membrane potential, reduced adenosine triphosphate (ATP) synthesis, increased mitochondrial ROS and oxidative stress, and insufficient oxidative phosphorylation (OXPHOS) together with decreased glycolysis. SIRT3 expression was significantly decreased in GCs of PCOS patients, and knockdown of SIRT3 in KGN cells could mimic the alterations in mitochondrial functions and glucose metabolism in PCOS GCs. SIRT3 knockdown changed the acetylation status of NDUFS1, which might induce altered mitochondrial OXPHOS, the generation of mitochondrial ROS, and eventually defects in the cellular insulin signaling pathway. These findings suggest that SIRT3 deficiency in GCs of PCOS patients may contribute to mitochondrial dysfunction, elevated oxidative stress, and defects in glucose metabolism, which potentially induce impaired oocytes in PCOS.
Collapse
Affiliation(s)
- Qing Zhang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Jun Ren
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Fangfang Wang
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Manman Pan
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Long Cui
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China
| | - Mingqian Li
- Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang, 310012, China
| | - Fan Qu
- Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310006, China.
| |
Collapse
|
27
|
Effect of dominant follicle status at the time of retrieval on the clinical outcomes in natural cycle IVF combined with immature oocyte treatment. Aging (Albany NY) 2022; 14:4728-4738. [PMID: 35674776 PMCID: PMC9217702 DOI: 10.18632/aging.204106] [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: 07/02/2021] [Accepted: 05/24/2022] [Indexed: 11/25/2022]
Abstract
Objective: It is commonly believed that the oocytes from small follicles are unhealthy when a dominant follicle (DF) is recruited in the ovaries, especially when the DF is ovulated. This study aims to confirm whether the presence or ovulation of DF at the time of retrieval affects the clinical outcome of the natural cycle IVF with in vitro maturation (NC-IVF/M) treatment. Methods: Data were collected from 446 women with regular menstrual cycle and 536 retrieval cycles using NC-IVF/M treatment. The cycles were divided into three groups based on the results of the oocyte retrieval cycle. Group A covers the collection of oocytes from the DF and small follicles; Group B incorporates failed oocyte retrieval from DF and then the oocytes are retrieved only from small follicles; and Group C includes the retrieval of oocytes only from small follicles accompanied with an ovulated DF. Furthermore, Group B and C have subgroups to include whether in vivo matured oocytes were obtained from small follicles. Following aspiration of DF and small follicles, mature oocytes were inseminated on the date of retrieval by intracytoplasmic sperm injection (ICSI) and the immature oocytes were matured in vitro. If the immature oocytes were matured in vitro, they were inseminated using ICSI, and then the embryos obtained from in vivo and in vitro matured oocytes were transferred accordingly. Results: The oocytes from DF were successfully retrieved in 445 cycles (83.0%), failed to be retrieved in 54 cycles (10.1%) and ovulated in 37 cycles (6.9%). In Group A, an average of 2.0 ± 1.7 mature oocytes were retrieved, which was significantly higher than the average of Group B, with 1.3 ± 1.3 matured oocytes and Group C, with an average of 1.1 ± 1.5 matured oocytes (P < 0.01). However, the average number of immature oocytes retrieved from each group show no difference among the three groups. There was no significant difference in maturation rates of immature oocytes, fertilization rates among the three groups. The clinical pregnancy rate per transfer cycle is 34.5%, 34.6% and 25.7% in Group A, B and C, respectively. No significant differences were observed in embryonic development and implantation capacity in Group B and C in comparison to Group A. And there was no significant difference in clinical pregnancy, implantation, live birth and miscarriage rates among the three groups. No significant differences were observed in the developmental and implantation capacity according to with or without in vivo matured oocytes were retrieved in Group B and Group C. Conclusion: The presence or ovulation of the dominant follicle from the ovaries does not significantly influence the developmental and implantation capacity of immature oocytes retrieved from small follicles, suggesting that NC-IVF/M is a promising treatment option for women without ovarian stimulation.
Collapse
|
28
|
Mina A, Boutzios G, Papoutsis I, Kaparos G, Christopoulos P, Kousta E, Mastrominas M, Athanaselis S, Mastorakos G. Bisphenol A correlates with fewer retrieved oocytes in women with tubal factor infertility. Hormones (Athens) 2022; 21:305-315. [PMID: 35524040 DOI: 10.1007/s42000-022-00370-1] [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: 06/18/2021] [Accepted: 04/12/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE Serum and urinary bisphenol A (BPA) concentrations have been associated negatively with the number of retrieved oocytes after in vitro fertilization (IVF). The impact of BPA upon women with polycystic ovary syndrome (PCOS) and women with tubal factor infertility (TFI), following IVF, was investigated. To this purpose, associations among serum and urinary and follicular fluid (FF) BPA concentrations and the number of retrieved and fertilized oocytes and comparisons between pregnancy rates were evaluated. METHODS This was a cross-sectional study conducted at a university-affiliated assisted conception unit between January and November 2019, including 93 women of reproductive age (PCOS: 45; TFI: 48) following IVF. Unconjugated FF and serum BPA concentrations and total urinary BPA concentration were measured using a novel gas chromatography-mass spectrometry method. The number of retrieved and fertilized oocytes and pregnancy rate were documented and evaluated. RESULTS The number of oocytes retrieved from PCOS women was greater than that of 21 TFI women, independently of BMI. Lower FF BPA concentrations were found in all PCOS women and in overweight/obese PCOS compared to TFI women (0.50, 0.38, and 1.13 ng/mL, respectively). In TFI women, FF BPA concentrations correlated negatively with the number of retrieved oocytes. Serum and FF and urinary BPA concentrations did not significantly affect the number of fertilized oocytes and pregnancy rate in both groups. CONCLUSION FF BPA concentrations were lower in all PCOS women and in overweight/obese PCOS than in TFI women. In TFI women, FF BPA concentrations correlated negatively with retrieved oocytes. Confirmation of these findings might lead to moderation of use of BPA-containing products by women undergoing IVF.
Collapse
Affiliation(s)
- Areti Mina
- Endocrine Unit, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, 76 Vas. Sofias Str, PO Box 11528, Athens, Greece
- Department of Forensic Medicine and Toxicology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str, PO Box 11527, Athens, Greece
| | - Georgios Boutzios
- Endocrine Unit, Department of Pathophysiology, Medical School, Laiko University Hospital, National and Kapodistrian University of Athens, 75 Mikras Asias Str, PO Box 11527, Athens, Greece
| | - Ioannis Papoutsis
- Department of Forensic Medicine and Toxicology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str, PO Box 11527, Athens, Greece
| | - George Kaparos
- Hormonal and Biochemical Laboratory, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, 76 Vas. Sofias Str, PO Box 11528, Athens, Greece
| | - Panagiotis Christopoulos
- Endocrine Unit, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, 76 Vas. Sofias Str, PO Box 11528, Athens, Greece
| | - Eleni Kousta
- Endocrine Unit, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, 76 Vas. Sofias Str, PO Box 11528, Athens, Greece
| | - Minas Mastrominas
- Embryogenesis Assisted Conception Unit, Kifisias 49 Avenue, PO Box 15123, Athens, Greece
| | - Sotirios Athanaselis
- Department of Forensic Medicine and Toxicology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Str, PO Box 11527, Athens, Greece
| | - George Mastorakos
- Endocrine Unit, Medical School, Aretaieion University Hospital, National and Kapodistrian University of Athens, 76 Vas. Sofias Str, PO Box 11528, Athens, Greece.
| |
Collapse
|
29
|
Yaka M, ÇİL N, Kabukçu C, Şenol H, Mete GA. The Relationship Between Embryo Development and Apoptotic Gene Expression in Cumulus Cells of Oocytes Collected from Patients with Poor Ovarian Response and Polycystic Ovary Syndrome. Reprod Biomed Online 2022; 45:935-942. [DOI: 10.1016/j.rbmo.2022.06.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 10/17/2022]
|
30
|
Comparison of two endometrial preparation methods for frozen-thawed embryo transfer in anovulatory PCOS patients: impact on miscarriage rate. J Gynecol Obstet Hum Reprod 2022; 51:102399. [PMID: 35489711 DOI: 10.1016/j.jogoh.2022.102399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 04/07/2022] [Accepted: 04/15/2022] [Indexed: 11/23/2022]
Abstract
OBJECTIVE - Some studies have suggested that patients with polycystic ovary syndrome (PCOS) are at high risk of miscarriage. However, this still remains controversial. Several potential factors might explain this association: obesity, hyperinsulinemia and hyperandrogenism. Artificial and stimulated cycles appear to be comparable for endometrial preparation in frozen-thawed embryo transfer (FET) in PCOS patients. Only a few studies have assessed miscarriage rates specifically in PCOS. We have evaluated the impact of endometrial preparation on FET outcomes in anovulatory PCOS patients. METHODS - A retrospective cohort study was conducted at the Lille University Hospital, including 255 FET cycles in 134 PCOS patients between January 2011 and December 2017. PCOS was defined by the presence of at least two of the three Rotterdam's criteria. Patients were under 35 years old. Two endometrial preparation protocol were studied: stimulated cycle (gonadotropins on the second day of the cycle and luteal phase support including natural progesterone 600 mg/day) and artificial cycle (6 mg oral estradiol valerate and 800 mg micronized vaginal progesterone daily). RESULTS - 137 FET were performed under stimulated cycle and 118 FET under artificial cycle. Early pregnancy rates (30% versus 37.3%, p = NS), miscarriage rates (22% versus 25%, p = NS) and live birth rates (23.4% versus 26.3%, p = NS) were similar. CONCLUSIONS - In anovulatory PCOS women, the type of endometrial preparation does not influence FET outcomes, specifically regarding the miscarriage rate.
Collapse
|
31
|
Ravisankar S, Murphy MJ, Redmayne-Titley N, Davis B, Luo F, Takahashi D, Hennebold JD, Chavez SL. Long-term Hyperandrogenemia and/or Western-style Diet in Rhesus Macaque Females Impairs Preimplantation Embryogenesis. Endocrinology 2022; 163:bqac019. [PMID: 35192701 PMCID: PMC8962721 DOI: 10.1210/endocr/bqac019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Indexed: 11/19/2022]
Abstract
Hyperandrogenemia and obesity are common in women with polycystic ovary syndrome, but it is currently unclear how each alone or in combination contribute to reproductive dysfunction and female infertility. To distinguish the individual and combined effects of hyperandrogenemia and an obesogenic diet on ovarian function, prepubertal female rhesus macaques received a standard control (C) diet, testosterone (T) implants, an obesogenic Western-style diet (WSD), or both (T + WSD). After 5 to 6 years of treatment, the females underwent metabolic assessments and controlled ovarian stimulations. Follicular fluid (FF) was collected for steroid and cytokine analysis and the oocytes fertilized in vitro. Although the T + WSD females exhibited higher insulin resistance compared to the controls, there were no significant differences in metabolic parameters between treatments. Significantly higher concentrations of CXCL-10 were detected in the FF from the T group, but no significant differences in intrafollicular steroid levels were observed. Immunostaining of cleavage-stage embryos revealed multiple nuclear abnormalities in the T, WSD, and T + WSD groups. Single-cell DNA sequencing showed that while C embryos contained primarily euploid blastomeres, most cells in the other treatment groups were aneuploid. Despite yielding a higher number of mature oocytes, T + WSD treatment resulted in significantly reduced blastocyst formation rates compared to the T group. RNA sequencing analysis of individual blastocysts showed differential expression of genes involved in critical implantation processes between the C group and other treatments. Collectively, we show that long-term WSD consumption reduces the capacity of fertilized oocytes to develop into blastocysts and that the addition of T further impacts gene expression and embryogenesis.
Collapse
Affiliation(s)
- Sweta Ravisankar
- Department of Cell, Developmental & Cancer Biology; Graduate Program in Molecular & Cellular Biosciences; Oregon Health & Science University School of Medicine; Portland, OR, USA
- Division of Reproductive & Developmental Sciences; Oregon National Primate Research Center; Beaverton, OR, USA
| | - Melinda J Murphy
- Division of Reproductive & Developmental Sciences; Oregon National Primate Research Center; Beaverton, OR, USA
| | - Nash Redmayne-Titley
- Division of Reproductive & Developmental Sciences; Oregon National Primate Research Center; Beaverton, OR, USA
| | - Brett Davis
- Knight Cardiovascular Institute; Oregon Health & Science University, Portland, OR, USA
| | - Fangzhou Luo
- Division of Reproductive & Developmental Sciences; Oregon National Primate Research Center; Beaverton, OR, USA
| | - Diana Takahashi
- Division of Cardiometabolic Health, Oregon National Primate Research Center; Beaverton, OR, USA
| | - Jon D Hennebold
- Division of Reproductive & Developmental Sciences; Oregon National Primate Research Center; Beaverton, OR, USA
- Department of Obstetrics & Gynecology; Oregon Health & Science University School of Medicine; Portland, OR, USA
| | - Shawn L Chavez
- Division of Reproductive & Developmental Sciences; Oregon National Primate Research Center; Beaverton, OR, USA
- Department of Obstetrics & Gynecology; Oregon Health & Science University School of Medicine; Portland, OR, USA
- Department of Molecular & Medical Genetics; Oregon Health & Science University School of Medicine; Portland, OR, USA
| |
Collapse
|
32
|
Chappell NR, Gibbons WE, Blesson CS. Pathology of hyperandrogenemia in the oocyte of polycystic ovary syndrome. Steroids 2022; 180:108989. [PMID: 35189133 PMCID: PMC8920773 DOI: 10.1016/j.steroids.2022.108989] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 02/07/2022] [Accepted: 02/16/2022] [Indexed: 12/01/2022]
Abstract
Polycystic ovary syndrome (PCOS) is the most common ovulatory disorder in the world and is associated with multiple adverse outcomes. The phenotype is widely varied, with several pathologies contributing to the spectrum of the disease including insulin resistance, obesity and hyperandrogenemia. Of these, the role of hyperandrogenemia and the mechanism by which it causes dysfunction remains poorly understood. Early studies have shown that androgens may affect the metabolic pathways of a cell, and this may pose hazards at the level of the mitochondria. As mitochondria are strictly maternally inherited, this would provide an exciting explanation not only to the pathophysiology of PCOS as a disease, but also to the inheritance pattern. This review seeks to summarize what is known about PCOS and associated adverse outcomes with focus on the role of hyperandrogenemia and specific emphasis on the oocyte.
Collapse
Affiliation(s)
- Neil R Chappell
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine. One Baylor Plaza, Houston 77030, TX, USA; Family Fertility Center, Texas Children's Hospital, Houston 77030, TX, USA
| | - William E Gibbons
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine. One Baylor Plaza, Houston 77030, TX, USA; Family Fertility Center, Texas Children's Hospital, Houston 77030, TX, USA
| | - Chellakkan S Blesson
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine. One Baylor Plaza, Houston 77030, TX, USA; Family Fertility Center, Texas Children's Hospital, Houston 77030, TX, USA.
| |
Collapse
|
33
|
Man L, Lustgarten Guahmich N, Kallinos E, Caiazza B, Khan M, Liu ZY, Patel R, Torres C, Pepin D, Yang HS, Bodine R, Zaninovic N, Schattman G, Rosenwaks Z, James D. Chronic superphysiologic AMH promotes premature luteinization of antral follicles in human ovarian xenografts. SCIENCE ADVANCES 2022; 8:eabi7315. [PMID: 35263130 PMCID: PMC8906729 DOI: 10.1126/sciadv.abi7315] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 01/14/2022] [Indexed: 05/19/2023]
Abstract
Anti-Müllerian hormone (AMH) is produced by growing ovarian follicles and provides a diagnostic measure of reproductive reserve in women; however, the impact of AMH on folliculogenesis is poorly understood. We cotransplanted human ovarian cortex with control or AMH-expressing endothelial cells in immunocompromised mice and recovered antral follicles for purification and downstream single-cell RNA sequencing of granulosa and theca/stroma cell fractions. A total of 38 antral follicles were observed (19 control and 19 AMH) at long-term intervals (>10 weeks). In the context of exogenous AMH, follicles exhibited a decreased ratio of primordial to growing follicles and antral follicles of increased diameter. Transcriptomic analysis and immunolabeling revealed a marked increase in factors typically noted at more advanced stages of follicle maturation, with granulosa and theca/stroma cells also displaying molecular hallmarks of luteinization. These results suggest that superphysiologic AMH alone may contribute to ovulatory dysfunction by accelerating maturation and/or luteinization of antral-stage follicles.
Collapse
Affiliation(s)
- Limor Man
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nicole Lustgarten Guahmich
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Eleni Kallinos
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Barbara Caiazza
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Monica Khan
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Zong-Ying Liu
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ritaben Patel
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Carmen Torres
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - David Pepin
- Department of Surgery, Harvard Medical School, Boston, MA 02215, USA
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA 02214, USA
| | - He S. Yang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Richard Bodine
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nikica Zaninovic
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
- Tri-Institutional Stem Cell Derivation Laboratory, Weill Cornell Medicine, New York, NY 10065, USA
| | - Glenn Schattman
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Zev Rosenwaks
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
| | - Daylon James
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY 10065, USA
- Tri-Institutional Stem Cell Derivation Laboratory, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
34
|
Uk A, Decanter C, Grysole C, Keller L, Béhal H, Silva M, Dewailly D, Robin G, Barbotin AL. Polycystic ovary syndrome phenotype does not have impact on oocyte morphology. Reprod Biol Endocrinol 2022; 20:7. [PMID: 34986863 PMCID: PMC8729101 DOI: 10.1186/s12958-021-00874-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 12/07/2021] [Indexed: 11/15/2022] Open
Abstract
PURPOSE The primary objective of the present study of women participating in an ICSI program was to determine whether the morphologic quality of oocytes was related to the polycystic ovary syndrome (PCOS) phenotype. METHODS We performed a retrospective cohort study in the IVF unit at the Lille University Medical Center (Lille, France) between 2006 and 2015. Oocyte morphology (fragmented first polar body, abnormal zona pellucida, large perivitelline space, material in perivitelline space, abnormal shape of oocyte, granular cytoplasm and intracytoplasmic vacuoles) was evaluated in PCOS women and according to different subgroup (depending on the presence or absence of the cardinal features polycystic ovarian morphology (PCOM), hyperandrogenism (HA), and oligo-anovulation (OA)). RESULTS A total of 1496 metaphase II oocytes (n = 602 for phenotype A combining PCOM + HA + OA, n = 462 oocytes for phenotype C: PCOM + HA, and n = 432 for phenotype D: PCOM + OA) were assessed. The phenotypes A, C and D did not differ significantly with regard to the proportion of normal oocytes (adjusted percentages (95%CI): 35.2% (31.5 to 39.1%), 25.8% (21.9 to 29.9%) and 34.0% (29.7 to 38.6%), respectively: adjusted p = 0.13). Likewise, there were no significant intergroup differences in oocyte morphology. The ICSI outcome was not significantly associated with the PCOS phenotype. CONCLUSION The present study is the first to show that the PCOS phenotype (notably the presence vs. absence of OA and/or HA) is not significantly associated with the morphological quality of oocytes.
Collapse
Affiliation(s)
- Audrey Uk
- CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Jeanne de Flandre Hospital, Lille, France
- Inserm UMR-S 1172, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille, France
- Department of Medicine, University of Lille, Lille, France
| | - Christine Decanter
- CHU Lille, Department of Endocrine Gynecology and Reproductive Medicine, Jeanne de Flandre Hospital, Lille, France
- Inserm EA 4308 Gametogenèse et Qualité du Gamète, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Lille, France
| | - Camille Grysole
- CHU Lille, Department of Endocrine Gynecology and Reproductive Medicine, Jeanne de Flandre Hospital, Lille, France
| | - Laura Keller
- CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Jeanne de Flandre Hospital, Lille, France
| | - Hélène Béhal
- Univ. Lille, CHU Lille, ULR 2694 - METRICS: Évaluation des Technologies de Santé et des Pratiques Médicales, Lille, France
| | - Mauro Silva
- Inserm UMR-S 1172, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille, France
| | | | - Geoffroy Robin
- Department of Medicine, University of Lille, Lille, France
- CHU Lille, Department of Endocrine Gynecology and Reproductive Medicine, Jeanne de Flandre Hospital, Lille, France
- Inserm EA 4308 Gametogenèse et Qualité du Gamète, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Lille, France
| | - Anne-Laure Barbotin
- CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Jeanne de Flandre Hospital, Lille, France.
- Inserm UMR-S 1172, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille, France.
- Department of Medicine, University of Lille, Lille, France.
| |
Collapse
|
35
|
Wang H, Li H, Zhu J, Xu J, Jiang Y, Chen W, Sun Y, Yang Q. The Effect of Sperm DNA Fragmentation on In Vitro Fertilization Outcomes for Women With Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2022; 13:822786. [PMID: 35712248 PMCID: PMC9196864 DOI: 10.3389/fendo.2022.822786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a prevalent endocrine disease in reproductive women associated with poor pregnancy outcomes. In modern society, people pay more attention to the female factor, but it is uncertain whether sperm quality is another factor affecting pregnancy outcomes of patients with PCOS. METHOD The effect of sperm DNA fragmentation (SDF) on oocyte fertilization, embryonic development, and pregnancy outcomes for patients with PCOS who underwent in vitro fertilization (IVF) treatment was studied. A total of 141 PCOS patients and 332 control patients undergoing IVF treatment were recruited from January 2017 to December 2019. All female patients were designated into two groups according to the Rotterdam criteria. Each group was divided into two sets, DNA fragmentation index (DFI) ≤15% and DFI > 15%, on the basis of sperm DFI. RESULT There were no differences in basic clinical characteristics between couples with a sperm DFI ≤ 15% or DFI > 15%. For control patients, no differences were observed in IVF outcomes. However, for PCOS patients, although there were no differences in the fertilization (60.4% vs. 59.9%, p = 0.831), high-quality embryo (68.5% vs. 67.9% p = 0.832), clinical pregnancy (78.4% vs. 66.7%, p = 0.148), and abortion (12.5% vs. 11.5%, p = 1.000) rates, a significantly lower high-quality blastocyst formation rate (26.3% vs. 16.3%, p = 0.023) was observed for couples with a sperm DFI > 15% compared with a sperm DFI ≤ 15%. CONCLUSION For PCOS patients undergoing IVF, oocytes fertilized using sperm with higher DFI led to a lower high-quality blastocyst formation rate but had no influence on fertilization, high-quality embryo, clinical pregnancy, and miscarriage rates.
Collapse
Affiliation(s)
- Huan Wang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Li
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Zhu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jianmin Xu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuqing Jiang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenhui Chen
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Yingpu Sun, ; Qingling Yang,
| | - Qingling Yang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Provincial Obstetrical and Gynecological Diseases (Reproductive Medicine) Clinical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Yingpu Sun, ; Qingling Yang,
| |
Collapse
|
36
|
Jiang X, Lu X, Cai M, Liu Y, Guo Y. Impact of dyslipidemia on the cumulative pregnancy outcomes after first ovarian stimulation. Front Endocrinol (Lausanne) 2022; 13:915424. [PMID: 36017313 PMCID: PMC9395644 DOI: 10.3389/fendo.2022.915424] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/15/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To investigate the cumulative live birth rate (CLBR) according to lipid metabolism in patients with or without polycystic ovarian syndrome (PCOS) undergoing their first complete in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) cycles. PATIENTS A total of 1,470 patients with PCOS and 3,232 patients without PCOS who underwent their first complete IVF/ICSI cycles from January 2016 to June 2018 were included. During a minimum of 2 years of follow-up, they had achieved at least one live birth or used all available embryos. The cumulative pregnancy outcomes were compared based on the patients' blood lipid parameters, including triglycerides (TG), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) and high-density lipoprotein cholesterol (HDL-C), in the two populations. Patients with an abnormal level of one or more of these four indicators were considered the dyslipidemia group. Patients whose four indicators were normal were considered the control group. RESULTS Among 1,470 patients with PCOS, the cumulative pregnancy outcomes were similar in the dyslipidemia group and control group. Logistic regression analysis showed that the TC levels were significantly negatively associated with the cumulative live birth rate (CLBR) after adjustment for confounding factors such as age and BMI (aOR 0.81, 95% CI 0.66-0.98, P<0.05). Among the 3,232 patients without PCOS, there was no significant difference in the cumulative pregnancy outcomes between the dyslipidemia group and the control group. No significant correlations were found in other logistic regression analyses. CONCLUSIONS TC negatively impacts the CLBR after first ovarian stimulation in PCOS patients. PCOS patients with dyslipidemia caused by elevated TC may have a poor CLBR.
Collapse
Affiliation(s)
- Xue Jiang
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, Zhengzhou, China
| | - Xinle Lu
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, Zhengzhou, China
| | - Mingshu Cai
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, Zhengzhou, China
| | - Yu Liu
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, Zhengzhou, China
| | - Yihong Guo
- Center of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Reproduction and Genetics, Zhengzhou, China
- *Correspondence: Yihong Guo,
| |
Collapse
|
37
|
Ma Y, Zheng L, Wang Y, Gao Y, Xu Y. Arachidonic Acid in Follicular Fluid of PCOS Induces Oxidative Stress in a Human Ovarian Granulosa Tumor Cell Line (KGN) and Upregulates GDF15 Expression as a Response. Front Endocrinol (Lausanne) 2022; 13:865748. [PMID: 35634503 PMCID: PMC9132262 DOI: 10.3389/fendo.2022.865748] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/07/2022] [Indexed: 11/13/2022] Open
Abstract
Polycystic ovarian ovary syndrome (PCOS) is the main cause of ovulatory infertility and a common reproductive endocrine disease of women in reproductive age. In addition, nearly half of PCOS patients are associated with obesity, and their total free fatty acids tend to increase. Arachidonic acid (AA) is a polyunsaturated fatty acid. Oxidation products of AA reacting with various enzymes[cyclooxygenases (COX), lipoxygenases (LOX), cytochrome P450s (CYP)] can change cellular mitochondrial distribution and calcium ion concentration, and increase reactive oxygen species (ROS) production. In this study, we analyzed the follicular fluid fatty acids and found higher levels of C20:4n6 (AA) in PCOS patients than in normal control subjects. Also, to determine whether AA induces oxidative stress (OS) in the human ovarian granulosa tumor cell line (KGN) and affects its function, we treated KGN cells with or without reduced glutathione (GSH) and then stimulated them with AA. The results showed that AA significantly reduced the total antioxidant capacity (TAC) and activity of antioxidant enzymes and increased the malondialdehyde (MDA), ROS and superoxide anion(O2-)levels in KGN cells. In addition, AA was also found to impair the secretory and mitochondrial functions of KGN cells and induce their apoptosis. We further investigated the downstream genes affected by AA in KGN cells and its mechanism of action. We found that AA upregulated the expression of growth differentiation factor 15 (GDF15), which had a protective effect on inflammation and tissue damage. Therefore, we investigated whether AA-induced OS in KGN cells upregulates GDF15 expression as an OS response.Through silencing of GDF15 and supplementation with recombinant GDF15 (rGDF15), we found that GDF15, expressed as an OS response, protected KGN cells against AA-induced OS effects, such as impairment of secretory and mitochondrial functions and apoptosis. Therefore, this study suggested that AA might induce OS in KGN cells and upregulate the expression of GDF15 as a response to OS.
Collapse
Affiliation(s)
- Yalan Ma
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Lianwen Zheng
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Yeling Wang
- Cardiovascular Medicine Department, The First Hospital of Jilin University, Changchun, China
| | - Yiyin Gao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Ying Xu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Ying Xu,
| |
Collapse
|
38
|
Yao QY, Yuan XQ, Liu C, Du YY, Yao YC, Wu LJ, Jiang HH, Deng TR, Guo N, Deng YL, Zeng Q, Li YF. OUP accepted manuscript. Hum Reprod 2022; 37:1297-1310. [PMID: 35259255 DOI: 10.1093/humrep/deac040] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 02/09/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Qing-Yun Yao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Xiao-Qiong Yuan
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Chong Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yao-Yao Du
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yang-Cheng Yao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Lin-Jing Wu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Hua-Hua Jiang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Tao-Ran Deng
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Na Guo
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yan-Ling Deng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Qiang Zeng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Yu-Feng Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| |
Collapse
|
39
|
Huang TC, Huang MZ, Seow KM, Yang IJ, Pan SP, Chen MJ, Hwang JL, Chen SU. Progestin primed ovarian stimulation using corifollitropin alfa in PCOS women effectively prevents LH surge and reduces injection burden compared to GnRH antagonist protocol. Sci Rep 2021; 11:22732. [PMID: 34815477 PMCID: PMC8611037 DOI: 10.1038/s41598-021-02227-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/09/2021] [Indexed: 12/28/2022] Open
Abstract
Utilizing corifollitropin alfa in GnRH antagonist (GnRHant) protocol in conjunction with GnRH agonist trigger/freeze-all strategy (corifollitropin alfa/GnRHant protocol) was reported to have satisfactory outcomes in women with polycystic ovary syndrome (PCOS). Although lessening in gonadotropin injections, GnRHant were still needed. In addition to using corifollitropin alfa, GnRHant was replaced with an oral progestin as in progestin primed ovarian stimulation (PPOS) to further reduce the injection burden in this study. We try to investigate whether this regimen (corifollitropin alfa/PPOS protocol) could effectively reduce GnRHant injections and prevent premature LH surge in PCOS patients undergoing IVF/ICSI cycles. This is a retrospective cohort study recruiting 333 women with PCOS, with body weight between 50 and 70 kg, undergoing first IVF/ICSI cycle between August 2015 and July 2018. We used corifollitropin alfa/GnRHant protocol prior to Jan 2017 (n = 160), then changed to corifollitropin alfa/PPOS protocol (n = 173). All patients received corifollitropin alfa 100 μg on menstruation day 2/3 (S1). Additional rFSH was administered daily from S8. In corifollitropin alfa/GnRHant group, cetrorelix 0.25 mg/day was administered from S5 till the trigger day. In corifollitropin alfa/PPOS group, dydrogesterone 20 mg/day was given from S1 till the trigger day. GnRH agonist was used to trigger maturation of oocyte. All good quality day 5/6 embryos were frozen, and frozen-thawed embryo transfer (FET) was performed on subsequent cycle. A comparison of clinical outcomes was made between the two protocols. The primary endpoint was the incidence of premature LH surge and none of the patients occurred. Dydrogesterone successfully replace GnRHant to block LH surge while an average of 6.8 days of GnRHant injections were needed in the corifollitropin alfa/GnRHant group. No patients suffered from ovarian hyperstimulation syndrome (OHSS). The other clinical outcomes including additional duration/dose of daily gonadotropin administration, number of oocytes retrieved, and fertilization rate were similar between the two groups. The implantation rate, clinical pregnancy rate, and live birth rate in the first FET cycle were also similar between the two groups. In women with PCOS undergoing IVF/ICSI treatment, corifollitropin alfa/PPOS protocol could minimize the injections burden with comparable outcomes to corifollitropin alfa/GnRHant protocol.
Collapse
Affiliation(s)
- Ting-Chi Huang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei, Taiwan
| | - Mei-Zen Huang
- Department of Nursing, National Tainan Junior College of Nursing, Tainan, Taiwan
| | - Kok-Min Seow
- Department of Obstetrics and Gynecology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ih-Jane Yang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei, Taiwan
| | - Song-Po Pan
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei, Taiwan
| | - Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei, Taiwan
| | - Jiann-Loung Hwang
- Department of Obstetrics and Gynecology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Taipei IVF, Center for Reproduction and Genetics, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Taipei Medical University, Taipei, Taiwan
| | - Shee-Uan Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, No. 7 Chung-Shan South Road, Taipei, Taiwan.
| |
Collapse
|
40
|
Anti-Müllerian Hormone in Pathogenesis, Diagnostic and Treatment of PCOS. Int J Mol Sci 2021; 22:ijms222212507. [PMID: 34830389 PMCID: PMC8619458 DOI: 10.3390/ijms222212507] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 01/19/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among reproductive-aged women. It is characterized by chronic anovulation, hyperandrogenism, and the presence of polycystic ovary in ultrasound examination. PCOS is specified by an increased number of follicles at all growing stages, mainly seen in the preantral and small antral follicles and an increased serum level of Anti-Müllerian Hormone (AMH). Because of the strong correlation between circulating AMH levels and antral follicle count on ultrasound, Anti-Müllerian Hormone has been proposed as an alternative marker of ovulatory dysfunction in PCOS. However, the results from the current literature are not homogeneous, and the specific threshold of AMH in PCOS and PCOM is, therefore, very challenging. This review aims to update the current knowledge about AMH, the pathophysiology of AMH in the pathogenesis of PCOS, and the role of Anti-Müllerian Hormone in the treatment of this syndrome.
Collapse
|
41
|
Compromised Cumulus-Oocyte Complex Matrix Organization and Expansion in Women with PCOS. Reprod Sci 2021; 29:836-848. [PMID: 34748173 DOI: 10.1007/s43032-021-00775-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/14/2021] [Indexed: 10/19/2022]
Abstract
The cumulus-oocyte complex (COC) matrix plays a critical role in the ovulation and fertilization process and a major predictor of oocyte quality. Proteomics studies of follicular fluid showed differential expression of COC matrix proteins in women with polycystic ovary syndrome (PCOS), indicating altered COC matrix in these women. In the present study, we aimed to understand COC matrix gene induction in humans and its probable dysfunction in women with PCOS. Animal studies have shown that amphiregulin (AREG) and growth differentiation factor-9 (GDF-9) are important in the induction of COC matrix genes which are involved in cumulus expansion. The effects of AREG and GDF-9 on expression of tumor necrosis factor alpha induced protein 6 (TNFAIP6) and hyaluronan synthase 2 (HAS2) on human cumulus granulosa cells (CGCs) and murine COC expansion were evaluated. Further time-dependent effects of growth factor supplementation on these gene expressions in CGCs from PCOS and control women were compared. Follicular fluid from PCOS showed reduced COC matrix expansion capacity, using murine COCs. Expression of COC matrix genes TNFAIP6 and HAS2 were significantly reduced in CGCs of PCOS. Treatment of CGCs with AREG and GDF-9 together induced expression of both these genes in controls and could only restore HAS2 but not TNFAIP6 expression in PCOS. Our results suggest that the reduced potential of follicular fluid to support COC expansion, altered expression of structural constituents, and intrinsic defects in granulosa cells of women with PCOS may contribute to the aberrant COC organization and expansion in PCOS, thus affecting fertilization.
Collapse
|
42
|
Li J, Chen H, Gou M, Tian C, Wang H, Song X, Keefe DL, Bai X, Liu L. Molecular Features of Polycystic Ovary Syndrome Revealed by Transcriptome Analysis of Oocytes and Cumulus Cells. Front Cell Dev Biol 2021; 9:735684. [PMID: 34552933 PMCID: PMC8450412 DOI: 10.3389/fcell.2021.735684] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 08/09/2021] [Indexed: 01/21/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is typically characterized by a polycystic ovarian morphology, hyperandrogenism, ovulatory dysfunction, and infertility. Furthermore, PCOS patients undergoing ovarian stimulation have more oocytes; however, the poor quality of oocytes leads to lower fertilization and implantation rates, decreased pregnancy rates, and increased miscarriage rates. The complex molecular mechanisms underlying PCOS and the poor quality of oocytes remain to be elucidated. We obtained matched oocytes and cumulus cells (CCs) from PCOS patients, compared them with age-matched controls, and performed RNA sequencing analysis to explore the transcriptional characteristics of their oocytes and CCs. Moreover, we validated our newly confirmed candidate genes for PCOS by immunofluorescence. Unsupervised clustering analysis showed that the overall global gene expression patterns and transposable element (TE) expression profiles of PCOS patients tightly clustered together, clearly distinct from those of controls. Abnormalities in functionally important pathways are found in PCOS oocytes. Notably, genes involved in microtubule processes, TUBB8 and TUBA1C, are overexpressed in PCOS oocytes. The metabolic and oxidative phosphorylation pathways are also dysregulated in both oocytes and CCs from PCOS patients. Moreover, in oocytes, differentially expressed TEs are not uniformly dispersed in human chromosomes. Endogenous retrovirus 1 (ERV1) elements located on chromosomes 2, 3, 4, and 5 are rather highly upregulated. Interestingly, these correlate with the most highly expressed protein-coding genes, including tubulin-associated genes TUBA1C, TUBB8P8, and TUBB8, linking the ERV1 elements to the occurrence of PCOS. Our comprehensive analysis of gene expression in oocytes and CCs, including TE expression, revealed the specific molecular features of PCOS. The aberrantly elevated expression of TUBB8 and TUBA1C and ERV1 provides additional markers for PCOS and may contribute to the compromised oocyte developmental competence in PCOS patients. Our findings may also have implications for treatment strategies to improve oocyte maturation and the pregnancy outcomes for women with PCOS.
Collapse
Affiliation(s)
- Jie Li
- The State Key Laboratory of Medicinal Chemical Biology, Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, China
| | - Haixia Chen
- The Center for Reproductive Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Mo Gou
- The State Key Laboratory of Medicinal Chemical Biology, Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, China
| | - Chenglei Tian
- The State Key Laboratory of Medicinal Chemical Biology, Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, China
| | - Huasong Wang
- The State Key Laboratory of Medicinal Chemical Biology, Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, China
| | - Xueru Song
- The Center for Reproductive Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - David L Keefe
- Department of Obstetrics and Gynecology, NYU Langone Medical Center, New York, NY, United States
| | - Xiaohong Bai
- The Center for Reproductive Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Lin Liu
- The State Key Laboratory of Medicinal Chemical Biology, Department of Cell Biology and Genetics, College of Life Sciences, Nankai University, Tianjin, China
| |
Collapse
|
43
|
Palomba S. Is fertility reduced in ovulatory women with polycystic ovary syndrome? An opinion paper. Hum Reprod 2021; 36:2421-2428. [PMID: 34333641 DOI: 10.1093/humrep/deab181] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 06/22/2021] [Indexed: 01/13/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility; however, whether women with PCOS and spontaneous or drug-induced ovulatory cycles have the same reproductive potential as non-PCOS controls is a matter of debate. In the present opinion paper, the author takes the opportunity to summarize the collective evidence supporting the hypothesis of reduced fertility potential in women with PCOS, regardless of ovulatory status, and speculate that reduced reproductive potential may be caused by altered oocytes, embryo and endometrial competence, and infertility-related co-morbidities as well as an increased risk of pregnancy complications.
Collapse
Affiliation(s)
- Stefano Palomba
- Obstetrics and Gynecology, University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
44
|
Berntsen S, Nøhr B, Grøndahl ML, Petersen MR, Andersen LF, Englund AL, Knudsen UB, Prætorius L, Zedeler A, Nielsen HS, Pinborg A, Freiesleben NLC. In vitro fertilisation (IVF) versus intracytoplasmic sperm injection (ICSI) in patients without severe male factor infertility: study protocol for the randomised, controlled, multicentre trial INVICSI. BMJ Open 2021; 11:e051058. [PMID: 34168037 PMCID: PMC8231059 DOI: 10.1136/bmjopen-2021-051058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
INTRODUCTION Over the last decades, the use of intracytoplasmic sperm injection (ICSI) has increased, even among patients without male factor infertility. The increase has happened even though there is no evidence to support that ICSI results in higher live birth rates compared with conventional in vitro fertilisation (IVF) in cases with nonmale factor infertility. The lack of robust evidence on an advantage of using ICSI over conventional IVF in these patients is problematic since ICSI is more invasive, complex and requires additional resources, time and effort. Therefore, the primary objective of the IVF versus ICSI (INVICSI) study is to determine whether ICSI is superior to standard IVF in patients without severe male factor infertility. The primary outcome measure is first live birth from fresh and frozen-thawed transfers after one stimulated cycle. Secondary outcomes include fertilisation rate, ongoing pregnancy rate, birth weight and congenital anomalies. METHODS AND ANALYSIS This is a two-armed, multicentre, randomised, controlled trial. In total, 824 couples/women with infertility without severe male factor will be recruited and allocated randomly into two groups (IVF or ICSI) in a 1:1 ratio. Participants will be randomised in variable block sizes and stratified by trial site and age. The main inclusion criteria are (1) no prior IVF/ICSI treatment, (2) male partner sperm with an expected count of minimum 2 million progressive motile spermatozoa following density gradient purification on the day of oocyte pick up and (3) age of the woman between 18 and 42 years. ETHICS AND DISSEMINATION The study will be performed in accordance with the ethical principles in the Helsinki Declaration. The study is approved by the Scientific Ethical Committee of the Capital Region of Denmark. Study findings will be presented, irrespectively of results at international conferences and submitted for publication in peer-reviewed journals. TRIAL REGISTRATION NUMBER NCT04128904. Pre-results.
Collapse
Affiliation(s)
- Sine Berntsen
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre Hospital, Hvidovre, Denmark
| | - Bugge Nøhr
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Herlev, Herlev Hospital, Herlev, Denmark
| | - Marie Louise Grøndahl
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Herlev, Herlev Hospital, Herlev, Denmark
| | - Morten Rønn Petersen
- The Fertility Clinic, Copenhagen University Hospital Rigshospitalet, Rigshospitalet, Copenhagen, Denmark
| | - Lars Franch Andersen
- Department of Obstetrics and Gynaecology, Copenhagen University Hospital North Zealand, North Zealand Hospital, Hilleroed, Denmark
| | - Anne Lis Englund
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Zealand University Hospital Koege, Zealand University Hospital Koge, Koege, Denmark
| | - Ulla Breth Knudsen
- Department of Obstetrics and Gynaecology, The Fertility Clinic, The Regional Hospital Horsens, Regional Hospital Horsens, Horsens, Denmark
| | - Lisbeth Prætorius
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre Hospital, Hvidovre, Denmark
| | - Anne Zedeler
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre Hospital, Hvidovre, Denmark
| | - Henriette Svarre Nielsen
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre Hospital, Hvidovre, Denmark
- Institute of Clinical Medicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Anja Pinborg
- The Fertility Clinic, Copenhagen University Hospital Rigshospitalet, Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Medicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Nina La Cour Freiesleben
- Department of Obstetrics and Gynaecology, The Fertility Clinic, Copenhagen University Hospital Hvidovre, Hvidovre Hospital, Hvidovre, Denmark
- Institute of Clinical Medicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
45
|
Salemi S, Yahyaei A, Vesali S, Ghaffari F. Endometrial preparation for vitrified-warmed embryo transfer with or without GnRH-agonist pre-treatment in patients with polycystic ovary syndrome: a randomized controlled trial. Reprod Biomed Online 2021; 43:446-452. [PMID: 34340936 DOI: 10.1016/j.rbmo.2021.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 10/21/2022]
Abstract
RESEARCH QUESTION What are the effects on pregnancy outcome in patients with polycystic ovary syndrome (PCOS) in endometrial preparation cycles for vitrified-warmed embryo transfer with or without gonadotrophin releasing hormone (GnRH) agonist pre-treatment? DESIGN A total of 212 patients with PCOS referred to Royan Institute, Tehran, Iran, between 20 August 2017 to 20 June 2018 were included. The patients were randomly assigned to receive oestradiol after downregulation with GnRH agonist (group A) or without GnRH agonist down-regulation (group B). RESULTS A total of 188 patients with PCOS completed the trial, 93 patients in group A and 95 patients in group B. Basal oestradiol and LH levels were significantly higher in group B (26.66 versus 41.61, P = 0.01 and 0.93 versus 5.33, P < 0.0001, respectively). Clinical pregnancy rates were not significantly different in both groups (31.2% versus 33.7%). Similarly, no significant differences were found between groups A and B in miscarriage (9.7% versus 11.6%), implantation (0.58 versus 0.51) and live birth (21.7% versus 22.1%) rates and for medical complications during pregnancy and neonatal anomalies. CONCLUSIONS Our findings indicate that endometrial preparation for frozen-thawed embryo transfer with and without ovarian suppression by GnRH agonist provides similar results.
Collapse
Affiliation(s)
- Shabnam Salemi
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Number 12, East Hafez Avenue, Banihashem Street, Resalat Highway Tehran, Iran
| | - Azar Yahyaei
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Number 12, East Hafez Avenue, Banihashem Street, Resalat Highway Tehran, Iran
| | - Samira Vesali
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR Tehran, Iran
| | - Firouzeh Ghaffari
- Department of Endocrinology and Female Infertility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Number 12, East Hafez Avenue, Banihashem Street, Resalat Highway Tehran, Iran.
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW To provide an overview of mitochondrial functional alterations in women with polycystic ovary syndrome (PCOS). RECENT FINDINGS Although numerous studies have focused on PCOS, the pathophysiological mechanisms that cause this common disease remain unclear. Mitochondria play a central role in energy production, and mitochondrial dysfunction may underlie several abnormalities observed in women with PCOS. Recent studies associated mtDNA mutations and low mtDNA copy number with PCOS, and set out to characterize the potential protective role of mitochondrial and endoplasmic reticulum unfolded protein responses (UPR and UPR). SUMMARY Mitochondrial dysfunction likely plays a role in the pathogenesis of PCOS by increasing reactive oxygen (ROS) and oxidative stress. This occurs in a metabolic milieu often affected by insulin resistance, which is a common finding in women with PCOS, especially in those who are overweight or obese. Mutations in mtDNA and low mtDNA copy number are found in these patients and may have potential as diagnostic modalities for specific PCOS phenotypes. More recently, UPR and UPR are being investigated as potential cellular rescue mechanisms in PCOS, the failure of which may lead to apoptosis, and contribute to decreased reproductive potential.
Collapse
|
47
|
Bulsara J, Patel P, Soni A, Acharya S. A review: Brief insight into Polycystic Ovarian syndrome. ENDOCRINE AND METABOLIC SCIENCE 2021. [DOI: 10.1016/j.endmts.2021.100085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
48
|
Yang G, Yao G, Xu Z, Fan H, Liu X, He J, Kong Y, Kong D, Bai Y, He Q, Zhang T, Zhang J, Sun Y. Expression Level of ADAMTS1 in Granulosa Cells of PCOS Patients Is Related to Granulosa Cell Function, Oocyte Quality, and Embryo Development. Front Cell Dev Biol 2021; 9:647522. [PMID: 33912563 PMCID: PMC8075003 DOI: 10.3389/fcell.2021.647522] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/22/2021] [Indexed: 11/27/2022] Open
Abstract
A disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS1) is an extracellular matrix metalloproteinase that plays an important role in the process of ovulation. According to previous studies, the expression level of ADAMTS1 in the granulosa cells of polycystic ovarian syndrome (PCOS) patients and the mechanism for regulating oocyte quality and embryonic development potential are still unclear. Our research clarified that ADAMTS1 was significantly increased in granulosa cells of PCOS patients as compared to ovulatory controls. After silencing ADAMTS1 in granulosa cells, cell proliferation and E2 secretion were significantly inhibited, which may be related to the down-regulation of B-cell lymphoma 2 (Bcl2) family genes and key genes involved in E2 synthesis. Through retrospective analysis of the clinical data, it was found that the expression level of ADAMTS1 was significantly positively correlated to the oocyte maturation rate and good-quality embryo rate in PCOS patients. The downregulation of ADAMTS1 in primary granulosa cells lead to the changes in the expression of marker genes for oocyte and embryonic quality. By using immunofluorescence staining, it was found ADAMTS1 was expressed in various stages of pre-implantation embryo but its expression level gradually decreases with the development of the embryo. In addition, the silence of ADAMTS1 in 3PN zygotes significantly prolonged the development time of the zygote to the morula stage. This is, to our knowledge, the first time to explored the mechanism by which ADAMST1 is involved in affecting the quality of oocytes and embryonic development potential, which will provide new evidence for further understanding of the follicular microenvironment and embryo development.
Collapse
Affiliation(s)
- Guang Yang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guidong Yao
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ziwen Xu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huiying Fan
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xingui Liu
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiahuan He
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yue Kong
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Deqi Kong
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yucheng Bai
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qina He
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tongwei Zhang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junya Zhang
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingpu Sun
- Center for Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
49
|
In vitro fertilization outcomes in women with polycystic ovary syndrome: A meta-analysis. Eur J Obstet Gynecol Reprod Biol 2021; 259:146-152. [PMID: 33676123 DOI: 10.1016/j.ejogrb.2021.02.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/22/2021] [Accepted: 02/22/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE During the past three decades, applying IVF for infertility treatment PCOS women has increased significantly, and the landscape of treatment strategies has changed dramatically. However, early review of IVF on PCOS have insufficiently accounted for efficacy and safety of the technic. With abundant studies in recent years, there is a need to reconcile these new data. MATERIAL AND METHODS To compare reproductive and obstetric outcomes of IVF between women with and without PCOS, a meta-analysis of 95 studies involving more than 21289 PCOS patients and 43036 controls was performed. RESULTS Despite longer stimulation duration (WMD = 0.34 day, 95 % CI: 0.09, 0.59) and lower dose of Gn required (WMD = -361.3 IU, 95 % CI: -442.3, -280.4), more oocytes (WMD = 3.67, 95 % CI: 3.14-4.21) and matured oocytes (WMD = 2.16, 95 % CI: 1.52-2.80) per cycle were obtained from PCOS women. There were no statistically significant differences for cleavage, high-grade embryo and implantation rate. Although similar pregnancy and live birth rates per cycle were achieved in PCOS and non-PCOS women after IVF, women with PCOS still suffered from significantly increased risks of miscarriage (OR = 1.44, 95 % CI: 1.20-1.72), biochemical pregnancy loss (OR = 1.89, 95 % CI: 1.48-2.41), and OHSS (OR = 3.58, 95 % CI: 2.86-4.48), in addition to lower fertilization rate (OR = 0.79, 95 % CI: 0.71-0.88). Adverse obstetric outcomes including ectopics pregnancy and multiple pregnancies are comparable between two groups. The overall cycle cancellation rate was significantly higher among PCOS women with OR of 2.55 (95 % CI: 1.67-3.89), and concern over OHSS or hyper-response constitute the main cause. Similar results were also observed after stratified analysis. CONCLUSIONS Our results support the effectiveness of IVF for infertility treatment among PCOS patients. However, options to minimize adverse outcomes regarding to lower fertilization, miscarriage, biochemical pregnancy loss and OHSS are required. Further studies elucidating detailed mechanism underlying these adverse outcomes could be of great importance to improve the experience of IVF treatment.
Collapse
|
50
|
Infertility management in women with polycystic ovary syndrome: a review. Porto Biomed J 2021; 6:e116. [PMID: 33532657 PMCID: PMC7846416 DOI: 10.1097/j.pbj.0000000000000116] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/22/2020] [Indexed: 11/25/2022] Open
Abstract
Polycystic ovary syndrome is the most common endocrine disorder in women and a major cause of anovulatory infertility. Various medical options are used, alone or in combination, to treat subfertility associated with polycystic ovary syndrome. This narrative review was conducted to provide an update and summarize the available evidence on the management of polycystic ovary syndrome related infertility. A wide literature search was performed and preferably randomized controlled trials and systematic reviews were included. Management is often centered on lifestyle changes. Pharmacological ovulation induction is the next step, with recommended use of letrozole, clomiphene citrate or gonadotropins. When it fails, assisted reproductive technologies or laparoscopic ovarian drilling are frequently advised. Combination treatment with metformin is often recommended. More recent alternative and adjunctive treatments have been suggested, like inositol, vitamin D, bariatric surgery and acupuncture, but further research is needed for recommendation.
Collapse
|