Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
BMC Womens Health ; 24(1): 315, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824522

ABSTRACT

BACKGROUND: Sleep health and obesity may affect the risk of female infertility. However, few studies focused on the interaction of obesity and sleep health on the female infertility risk. This study aimed to evaluate the combined impact of trouble sleeping / sleep duration and overweight/obesity/ abdominal obesity on the risk of female infertility. METHODS: The data for this cross-sectional study was obtained from National Health and Nutritional Examination Survey, which provided information on trouble sleeping, sleep duration, overweight/obesity, abdominal obesity, and confounding factors. Adopted weighted univariate and multivariate logistic regression models to explore the relationship between trouble sleeping, sleep duration, overweight/obesity, abdominal obesity, and the risk of infertility, respectively, and the combined effect of trouble sleeping and overweight/obesity, trouble sleeping and abdominal obesity, sleep duration and overweight/obesity, sleep duration and abdominal obesity, on the female infertility risk. RESULTS: This study included a total of 1,577 women, and 191 were diagnosed with infertility. Women with infertility had a higher proportion of people with overweight/obesity, abdominal obesity, sleep duration ≤ 7 h and trouble sleeping than those with non-infertility. The result indicated that trouble sleeping [odds ratio (OR) = 2.25, 95% confidence intervals (CI): 1.49-3.39], sleep duration ≤ 7 h (OR = 1.59, 95% CI: 1.03-2.48), and the combined impact of abdominal obesity and trouble sleeping (OR = 2.18, 95% CI: 1.28-3.72), abdominal obesity and sleep duration ≤ 7 h (OR = 2.00, 95% CI: 1.17-3.40), overweight/obesity and trouble sleeping (OR = 2.29, 95% CI: 1.24-4.26), and overweight/obesity and sleep duration ≤ 7 h (OR = 1.88, 95% CI: 1.01-3.49) were associated with increased odds of infertility, respectively. CONCLUSION: There was combined effects of trouble sleeping/sleep duration ≤ 7 h and overweight/obesity/ abdominal obesity on increased odds of female infertility.


Subject(s)
Infertility, Female , Nutrition Surveys , Obesity, Abdominal , Obesity , Sleep Wake Disorders , Humans , Female , Adult , Infertility, Female/epidemiology , Infertility, Female/etiology , Cross-Sectional Studies , Obesity/epidemiology , Obesity/complications , Obesity, Abdominal/epidemiology , Obesity, Abdominal/complications , Sleep Wake Disorders/epidemiology , Sleep Wake Disorders/complications , Sleep/physiology , Overweight/epidemiology , Overweight/complications , Risk Factors , Young Adult , United States/epidemiology
2.
Cell Death Dis ; 15(2): 161, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383507

ABSTRACT

Oxidative stress dysfunction has recently been found to be involved in the pathogenesis of premature ovarian insufficiency (POI). Previously, we found that advanced oxidation protein products (AOPPs) in plasma were elevated in women with POI and had an adverse effect on granulosa cell proliferation. However, the mechanism underlying the effects of AOPPs on autophagy-lysosome pathway regulation in granulosa cells remains unclear. In this study, the effect of AOPPs on autophagy and lysosomal biogenesis and the underlying mechanisms were explored by a series of in vitro experiments in KGN and COV434 cell lines. AOPP-treated rat models were employed to determine the negative effect of AOPPs on the autophagy-lysosome systems in vivo. We found that increased AOPP levels activated the mammalian target of rapamycin (mTOR) pathway, and inhibited the autophagic response and lysosomal biogenesis in KGN and COV434 cells. Furthermore, scavenging of reactive oxygen species (ROS) with N-acetylcysteine and blockade of the mTOR pathway with rapamycin or via starvation alleviated the AOPP-induced inhibitory effects on autophagy and lysosomal biogenesis, suggesting that these effects of AOPPs are ROS-mTOR dependent. The protein expression and nuclear translocation of transcription factor EB (TFEB), the key regulator of lysosomal and autophagic function, were also impaired by the AOPP-activated ROS-mTOR pathway. In addition, TFEB overexpression attenuated the AOPP-induced impairment of autophagic flux and lysosomal biogenesis in KGN and COV434 cells. Chronic AOPP stimulation in vivo also impaired autophagy and lysosomal biogenesis in granulosa cells of rat ovaries. The results highlight that AOPPs lead to impairment of autophagic flux and lysosomal biogenesis via ROS-mTOR-TFEB signaling in granulosa cells and participate in the pathogenesis of POI.


Subject(s)
Advanced Oxidation Protein Products , TOR Serine-Threonine Kinases , Humans , Rats , Female , Animals , Advanced Oxidation Protein Products/metabolism , Advanced Oxidation Protein Products/pharmacology , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lysosomes/metabolism , Granulosa Cells/metabolism , Mammals
3.
Article in English | MEDLINE | ID: mdl-37814781

ABSTRACT

Pituitary stalk interruption syndrome (PSIS) in female patients is mainly characterized by short stature, primary amenorrhea, absent or incomplete sexual maturation, and infertility. Successful pregnancies among these patients are rare. In this report, we describe a successful pregnancy and delivery in a 28-year-old Chinese woman with PSIS following in vitro fertilization and embryo transfer. The patient exhibited typical symptoms, including multiple pituitary hormone deficiency, typical triad signs in magnetic resonance imaging (MRI), undetectable serum gonadotropins and estradiol levels, and invisible antral follicles in both ovaries. During the first attempted controlled ovarian hyperstimulation cycle, 14 oocytes were retrieved and six embryos were acquired. Artificial endometrial preparation and frozen-thawed embryo transfer were performed, resulting in a clinical pregnancy after the transfer of a day 5 blastocyst. The patient was closely monitored throughout the pregnancy and multiple hormone dosages were modulated accordingly. She delivered a healthy boy by elective cesarean section, and the newborn developed normally during a 1-year follow-up period. This is the first report of a successful live birth in a woman with PSIS achieved through in vitro fertilization and frozen-thawed embryo transfer. A literature review on this topic is also presented.

4.
Clin Genet ; 104(5): 516-527, 2023 11.
Article in English | MEDLINE | ID: mdl-37461298

ABSTRACT

Premature ovarian insufficiency (POI) is a clinical syndrome of ovarian dysfunction characterized by cessation of menstruation occurring before the age of 40 years. The genetic causes of idiopathic POI remain unclear. Here we recruited a POI patient from a consanguineous family to screen for potential pathogenic variants associated with POI. Genetic variants of the pedigree were screened using whole-exome sequencing analysis and validated through direct Sanger sequencing. A homozygous variant in TUFM (c.524G>C: p.Gly175Ala) was identified in this family. TUFM (Tu translation elongation factor, mitochondrial) is a nuclear-encoded mitochondrial protein translation elongation factor that plays a critical role in maintaining normal mitochondrial function. The variant position was highly conserved among species and predicted to be disease causing. Our in vitro functional studies demonstrated that this variant causes decreased TUFM protein expression, leading to mitochondrial dysfunction and impaired autophagy activation. Moreover, we found that mice with targeted Tufm variant recapitulated the phenotypes of human POI. Thus, this is the first report of a homozygous pathogenic TUFM variant in POI. Our findings highlighted the essential role of mitochondrial genes in folliculogenesis and ovarian function maintenance.


Subject(s)
Primary Ovarian Insufficiency , Adult , Animals , Female , Humans , Mice , Consanguinity , Homozygote , Mitochondria/genetics , Mitochondria/pathology , Mutation , Primary Ovarian Insufficiency/pathology
5.
BMC Pregnancy Childbirth ; 23(1): 462, 2023 Jun 22.
Article in English | MEDLINE | ID: mdl-37349693

ABSTRACT

BACKGROUND: Premature ovarian insufficiency (POI) patients present with a chronic inflammatory state. Cell-free mitochondria DNA (cf-mtDNA) has been explored as a reliable biomarker for estimating the inflammation-related disorders, however, the cf-mtDNA levels in POI patients have never been measured. Therefore, in the presenting study, we aimed to evaluate the levels of cf-mtDNA in plasma and follicular fluid (FF) of POI patients and to determine a potential role of cf-mtDNA in predicting the disease progress and pregnancy outcomes. METHODS: We collected plasma and FF samples from POI patients, biochemical POI (bPOI) patients and control women. Quantitative real-time PCR was used to measure the ratio of mitochondrial genome to nuclear genome of cf-DNAs extracted from the plasma and FF samples. RESULTS: The plasma cf-mtDNA levels, including COX3, CYB, ND1 and mtDNA79, were significantly higher in overt POI patients than those in bPOI patients or control women. The plasma cf-mtDNA levels were weakly correlated with ovarian reserve, and could not be improved by regular hormone replacement therapy. The levels of cf-mtDNA in FF, rather than those in plasma, exhibited the potential to predict the pregnancy outcomes, although they were comparable among overt POI, bPOI and control groups. CONCLUSIONS: The increased plasma cf-mtDNA levels in overt POI patients indicated its role in the progress of POI and the FF cf-mtDNA content may hold the value in predicting pregnancy outcomes of POI patients.


Subject(s)
Cell-Free Nucleic Acids , Primary Ovarian Insufficiency , Pregnancy , Humans , Female , Primary Ovarian Insufficiency/genetics , Mitochondria/genetics , DNA, Mitochondrial , Biomarkers
6.
J Ovarian Res ; 16(1): 2, 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36600288

ABSTRACT

BACKGROUND: Premature ovarian insufficiency (POI) patients are predisposed to metabolic disturbances, including in lipid metabolism and glucose metabolism, and metabolic disorders appear to be a prerequisite of the typical long-term complications of POI, such as cardiovascular diseases or osteoporosis. However, the metabolic changes underlying the development of POI and its subsequent complications are incompletely understood, and there are few studies characterizing the disturbed metabolome in POI patients. The aim of this study was to characterize the plasma metabolome in POI by using ultrahigh-performance liquid chromatography-mass spectrometry (UHPLC-MS/MS) metabolomics and to evaluate whether these disturbances identified in the plasma metabolome relate to ovarian reserve and have diagnostic value in POI. METHODS: This observational study recruited 30 POI patients and 30 age- and body mass index (BMI)-matched controls in the Center for Reproductive Medicine, Department of Gynecology and Obstetrics, Nanfang Hospital, Southern Medical University, from January 2018 to October 2020. Fasting venous blood was collected at 9:00 am on days 2-4 of the menstrual cycle and centrifuged for analysis. An untargeted quantitative metabolomic analysis was performed using UHPLC-MS/MS. RESULTS: Our study identified 48 upregulated and 21 downregulated positive metabolites, and 13 upregulated and 48 downregulated negative metabolites in the plasma of POI patients. The differentially regulated metabolites were involved in pathways such as caffeine metabolism and ubiquinone and other terpenoid-quinone biosynthesis. Six metabolites with an AUC value > 0.8, including arachidonoyl amide, 3-hydroxy-3-methylbutanoic acid, dihexyl nonanedioate, 18-HETE, cystine, and PG (16:0/18:1), were correlated with ovarian reserve and thus have the potential to be diagnostic biomarkers of POI. CONCLUSION: This UHPLC-MS/MS untargeted metabolomics study revealed differentially expressed metabolites in the plasma of patients with POI. The differential metabolites may not only be involved in the aetiology of POI but also contribute to its major complications. These findings offer a panoramic view of the plasma metabolite changes caused by POI, which may provide useful diagnostic and therapeutic clues for POI disease.


Subject(s)
Menopause, Premature , Primary Ovarian Insufficiency , Female , Humans , Tandem Mass Spectrometry , Metabolome , Menstrual Cycle , Metabolomics
7.
Mol Cell Endocrinol ; 555: 111719, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35850487

ABSTRACT

In the present study, we focused on characterizing the proteome in granulosa cells in patients with biochemical premature ovarian insufficiency (bPOI) in order to identify differential proteins and investigate the fundamental mechanisms of POI. A total of 2688 proteins were identified based on the data-independent acquisition method, and 70 differentially expressed proteins were significant. Bioinformatic analyses, including gene expression pattern analysis, gene ontology enrichment analysis, Kyoto Encyclopedia of Genes and Genomes pathway analysis, and Search Tool for the Retrieval of Interacting Genes/Proteins analysis, revealed discrete modules and the underlying molecular mechanisms in bPOI. Importantly, we observed that Ras-related C3 botulinum toxin substrate 1 (RAC1) was downregulated in the granulosa cells of bPOI. Low expression of RAC1 may affect the development process of POI by affecting the proliferation, apoptosis, and hormone synthesis of granulosa cells. Downregulation of RAC1 expression in the KGN and COV434 cells inhibited cell proliferation, blocked cells in the G1/G0 phase, and promoted apoptosis. Western blot results showed that ß-catenin and cyclin D1 in the KGN and COV434 cells transfected with RAC1-siRNA were downregulated, while P21 and Bax were upregulated. Knocking down RAC1 in the KGN cells or adding the RAC1 enzyme inhibitor to the human luteinized granulosa cells (hLGC) inhibited the synthesis of E2, and the expression of aromatase and follicle-stimulating hormone receptor (FSHR) was reduced.


Subject(s)
Primary Ovarian Insufficiency , Proteomics , Apoptosis , Cell Proliferation , Female , Granulosa Cells , Humans , rac1 GTP-Binding Protein
8.
Oxid Med Cell Longev ; 2021: 6634718, 2021.
Article in English | MEDLINE | ID: mdl-34367464

ABSTRACT

The mechanism underlying the role of oxidative stress and advanced oxidation protein products (AOPPs) in the aetiology of premature ovarian insufficiency (POI) is poorly understood. Here, we investigated the plasma AOPP level in POI patients and the effects of AOPPs on granulosa cells both in vitro and in vivo. KGN cells were treated with different AOPP doses, and cell cycle distribution, intracellular reactive oxygen species (ROS), and protein expression levels were measured. Sprague-Dawley (SD) rats were treated daily with PBS, rat serum albumin, AOPP, or AOPP+ N-acetylcysteine (NAC) for 12 weeks to explore the effect of AOPPs on ovarian function. Plasma AOPP concentrations were significantly higher in both POI and biochemical POI patients than in controls and negatively correlated with anti-Müllerian hormone and the antral follicle count. KGN cells treated with AOPP exhibited G1/G0-phase arrest. AOPP induced G1/G0-phase arrest in KGN cells by activating the ROS-c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinase (MAPK)-p21 pathway. Pretreatment with NAC, SP600125, SB203580, and si-p21 blocked AOPP-induced G1/G0-phase arrest. In SD rats, AOPP treatment increased the proportion of atretic follicles, and NAC attenuated the adverse effects of AOPPs in the ovary. In conclusion, we provide mechanistic evidence that AOPPs may induce cell cycle arrest in granulosa cells via the ROS-JNK/p38 MAPK-p21 pathway and thus may be a novel biomarker of POI.


Subject(s)
Advanced Oxidation Protein Products/metabolism , Cell Cycle Checkpoints , Cyclin-Dependent Kinase Inhibitor p21/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Primary Ovarian Insufficiency/pathology , Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Advanced Oxidation Protein Products/genetics , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , G1 Phase , Gene Expression Regulation , Granulosa Cells/metabolism , Granulosa Cells/pathology , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Primary Ovarian Insufficiency/genetics , Primary Ovarian Insufficiency/metabolism , Prognosis , Rats , Rats, Sprague-Dawley , Resting Phase, Cell Cycle , p38 Mitogen-Activated Protein Kinases/genetics
9.
Mol Cell Endocrinol ; 535: 111392, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34246727

ABSTRACT

Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder in reproductive-aged women, and its pathogenesis is still under debate. Recent studies suggest crucial roles for microRNAs (miRNAs) in PCOS development. The let-7 family miRNAs constitute the most abundant miRNAs in human granulosa cells (GCs), and plays an important role in follicular development. However, research on the let-7e implications of the non-hyperandrogenic (non-HA) phenotype remains unclear. This study aimed at determining the role of let-7e in the progression of PCOS. We performed quantitative real-time PCR to examine the levels of let-7e in fifty-two non-HA PCOS patients and fifty-two controls. A receiver operating characteristic (ROC) curve were used to reveal the diagnostic value of let-7e in non-HA PCOS. Using an immortalized human granulosa cell line, KGN, we investigated the influence of let-7e on cell proliferation and autophagy. Our data substantiated the expression of let-7e was significantly increased in non-HA PCOS group, and associated with an increased antral follicle count. The ROC curve indicated a major separation between non-HA PCOS group and the control group. Let-7e knockdown suppressed cell proliferation and enhanced cell autophagy by activating p21 pathway. Conversely, let-7e overexpression promoted cell proliferation and inhibited cell autophagy by suppressing p21 pathway. Our results indicate that increased let-7e levels in non-HA PCOS GCs may contribute to excessive follicular activation and growth, thereby involving in the pathogenesis of PCOS. Let-7e may thus be a potential therapeutic target in non-HA PCOS.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/genetics , Granulosa Cells/cytology , Hyperandrogenism/genetics , MicroRNAs/genetics , Polycystic Ovary Syndrome/genetics , Adult , Autophagy , Case-Control Studies , Cell Line , Cell Proliferation , Female , Granulosa Cells/metabolism , Humans , Signal Transduction , Up-Regulation
10.
RNA Biol ; 17(12): 1798-1810, 2020 12.
Article in English | MEDLINE | ID: mdl-32559120

ABSTRACT

Polycystic ovary syndrome (PCOS) causes anovulatory infertility in women of reproductive age, but etiopathogenesis of PCOS remains undetermined. Taurine up-regulated 1 (TUG1), an evolutionarily conserved long non-coding RNA, performs various biological functions; however, the role of TUG1 in PCOS remains unclear. Herein, TUG1 expression was assayed in granulosa cells (GCs) of 100 patients with PCOS and 100 control participants. Receiver operating characteristic (ROC) curve analysis was conducted to determine the diagnostic value of TUG1 in PCOS. TUG1 expression was also silenced in KGN cells to explore the role of TUG1 in cellular proliferation, apoptosis, cell-cycle progression, autophagy, and steroidogenesis. We found that TUG1 levels were dramatically increased in the PCOS group compared with those of the control group; this increased expression was related to a rising antral follicle count (R = 0.209, P < 0.001 versus control). The ROC curve indicated a significant separation between PCOS group and the control group (AUC: 0.702; 95% CI: 0.630-0.773; P < 0.001). TUG1 showed a predominantly nuclear localization in human GCs. TUG1 knockdown reduced cellular proliferation, and promoted MAPKs pathway-dependent apoptosis and P21-dependent autophagy, but may not affect cell-cycle progression. TUG1 knockdown increased aromatase expression and oestradiol biosynthesis. Our results indicate that increased TUG1 expression in PCOS GCs may contribute to excessive follicular activation and growth, and may disrupt the selection of dominant follicle. Our study shows that TUG1 can be used as a diagnostic biomarker for PCOS.


Subject(s)
Gene Expression Regulation , Polycystic Ovary Syndrome/genetics , RNA, Long Noncoding/genetics , Adult , Apoptosis/genetics , Aromatase/metabolism , Autophagy/genetics , Biomarkers , Case-Control Studies , Cell Proliferation , Cells, Cultured , Disease Susceptibility , Estradiol/biosynthesis , Female , Follicle Stimulating Hormone/blood , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Granulosa Cells/metabolism , Humans , Insulin/administration & dosage , Insulin/therapeutic use , MAP Kinase Signaling System , Ovarian Follicle/metabolism , Ovarian Follicle/pathology , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/drug therapy , Polycystic Ovary Syndrome/metabolism , ROC Curve , Young Adult
11.
Reprod Biol Endocrinol ; 18(1): 52, 2020 May 25.
Article in English | MEDLINE | ID: mdl-32450894

ABSTRACT

BACKGROUND: The major difference between a natural cycle and an artificially prepared cycle is the lack of luteinizing hormone (LH) peak in the latter. The LH/hCG receptors were identified to express in human endometrium and evidences of experiments also suggested the beneficial role of hCG in embryo implantation, indicating that the LH peak might be of clinical significance and the activation of LH/hCG receptors in the endometrium could improve embryo implantation. Hence, we postulated that the addition of hCG prior to secretory transformation in an artificial cycle might improve pregnancy outcomes. METHODS: This retrospective cohort study was conducted at a Reproductive Medicine Center between 2016 and 2018. Patients aged ≤43 years at the (index) oocyte retrieval and undergoing artificially prepared frozen-thawed embryo transfer (FET) with at least one good-quality embryo transferred were included. The cycles were divided into two groups: The hCG group (n = 337) received an intramuscular injection of 10,000 IU hCG before secretory transformation; the control group (n = 364) performed FET without hCG administration. The primary endpoint was live birth delivery rate (LBR), secondary outcomes included implantation rate, clinical pregnancy rate (CPR) and ongoing pregnancy rate (OPR). RESULTS: The LBR (49.9% vs 39.6%, P < 0.01), CPR (61.4% vs 50.5%, P < 0.01) and OPR (52.8% vs 43.1%, P < 0.05) were statistically significantly higher in the hCG group than the control group. The superiority in LBR after hCG administration remained significant after adjusting for confounding factors (OR 1.613, 95% CI 1.173-2.217; P < 0.01). In the subgroup analysis, the improvement in LBR was statistically significant after hCG administration for cleavage-stage embryo transfer cycles (51.2% vs 42.3%, P < 0.05), whereas for blastocyst transfer cycles, the improvement in LBR was not (45.7% vs 31.3%, P > 0.05). CONCLUSIONS: Intramuscular hCG injection prior to secretory transformation may benefit LBR in patients undergoing artificially prepared FET cycles. But it should be noted that nonsignificant tendency towards higher LBR was observed after hCG administration in patients undergoing blastocyst transfer. So, future prospective randomized controlled studies are required to confirm, especially for blastocyst transfer cycles.


Subject(s)
Chorionic Gonadotropin/administration & dosage , Embryo Transfer/methods , Fertility Agents, Female/administration & dosage , Luteal Phase/drug effects , Adult , Cohort Studies , Cryopreservation , Drug Administration Schedule , Embryo Implantation/drug effects , Embryo, Mammalian , Female , Freezing , Humans , Injections, Intramuscular , Pregnancy , Pregnancy Outcome , Pregnancy Rate , Retrospective Studies
12.
Epigenomics ; 12(4): 319-332, 2020 02.
Article in English | MEDLINE | ID: mdl-32081025

ABSTRACT

Aim: To identify the expression profiles and potential functions of circular RNAs (circRNAs) in granulosa cells (GCs) from women with biochemical premature ovarian insufficiency (bPOI). Patients & methods: CircRNAs microarray analysis was performed to GCs from 8 patients with bPOI and 8 control women, followed by qRT-PCR in 15 paired samples. CircRNA-miRNA networks and the prediction of their enriched signaling pathways were conducted by bioinformatics analysis. Results: A total of 133 upregulated and 424 downregulated circRNAs was identified in women with bPOI. We constructed circRNA-miRNA networks and found that the most predominantly enriched signaling pathways were the FoxO signaling pathway and cellular senescence. Conclusion: CircRNAs are differentially expressed in bPOI, which might contribute to the pathogenesis of bPOI.


Subject(s)
Granulosa Cells/metabolism , Primary Ovarian Insufficiency/genetics , RNA, Circular/metabolism , Adult , Female , Gene Expression Profiling , Humans , MicroRNAs/metabolism , Primary Ovarian Insufficiency/diagnosis , Primary Ovarian Insufficiency/metabolism
13.
Mol Cell Endocrinol ; 498: 110540, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31421163

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility in reproductive-aged women; however, its etiology remains poorly understood. This study aimed to reveal the role of miR-29a in PCOS. MiR-29a levels were measured in the granulosa cells (GCs) of forty-seven PCOS patients and forty-seven controls. A receiver operating characteristic (ROC) curve was drawn to evaluate the diagnostic value of miR-29a in non-hyperandrogenism PCOS. MiR-29a was overexpressed in KGN and COV434 cells to examine its roles in proliferation, cell-cycle progression, and steroidogenesis. MiR-29a was significantly down-regulated in PCOS patients, and associated with an increased antral follicle count. The ROC curve showed a major separation between PCOS patients and controls. MiR-29a overexpression in KGN and COV434 cells inhibited cell proliferation, arrested cell-cycle progression, and decreased aromatase expression and estradiol production. These findings suggest that miR-29a is involved in GC proliferation and steroidogenesis, providing insights into PCOS pathogenesis.


Subject(s)
Aromatase/metabolism , Biomarkers/metabolism , Cell Proliferation , Estradiol/metabolism , Granulosa Cells/pathology , MicroRNAs/genetics , Polycystic Ovary Syndrome/pathology , Adult , Apoptosis , Aromatase/genetics , Case-Control Studies , Cell Cycle , Cells, Cultured , Female , Follow-Up Studies , Granulosa Cells/metabolism , Humans , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Prognosis
14.
Curr Med Sci ; 39(2): 278-284, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31016522

ABSTRACT

This observational study included 21 patients at remarkably high risk of ovarian hyperstimulation syndrome (OHSS), characterized by more than 30 follicles measuring ≥11 mm in diameter on trigger day and/or pre-trigger peak estradiol exceeding 10 000 pg/mL, which was also the feature of women with established severe early OHSS followed by gonadotrophin-releasing hormone agonist (GnRHa) trigger and freeze-all policy that previously have been reported. All patients received a second dose of GnRHa 12 h after the first GnRHa trigger combined with administration of GnRH antagonist at 0.25 mg/day for a period of 3 days from the day of oocyte retrieval onwards. The in vitro fertilization (IVF) outcomes may be preferable compared with a bolus of GnRHa trigger and none of the included patients developed moderate-to-severe OHSS. Moreover, patients' symptoms, reproductive hormone levels and ultrasound findings were improved significantly. This new strategy seems to be efficacious and could be a further supplement of GnRHa trigger with or without applying freeze-all strategy to completely prevent early-onset moderate to severe OHSS, especially for the patients characterized by ≤30 follicles measuring ≥11 mm in diameter on trigger day and/or pre-trigger peak estradiol exceeding 10 000 pg/mL. Further studies should be performed to compare this regimen with conventional methods of OHSS prevention.


Subject(s)
Estradiol/metabolism , Fertility Agents, Female/administration & dosage , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/agonists , Ovarian Follicle/drug effects , Ovarian Hyperstimulation Syndrome/prevention & control , Adult , Female , Fertilization in Vitro/methods , Gonadotropin-Releasing Hormone/administration & dosage , Humans , Infertility, Female/drug therapy , Infertility, Female/metabolism , Oocyte Retrieval/methods , Ovulation Induction/methods , Pregnancy , Pregnancy Rate
15.
Mol Hum Reprod ; 25(1): 17-29, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30371869

ABSTRACT

STUDY QUESTION: Is there a specific mechanism underlying the association between lung adenocarcinoma transcript 1 (MALAT1) and endometriosis-related infertility? SUMMARY ANSWER: The down-regulation of MALAT1 in endometriosis granulosa cells (GCs) may have an adverse effect on the growth and development of oocytes by inhibiting GC proliferation, due to cell cycle-dependent mechanisms that enhance P21 expression through activation of the extracellular signal-regulated kinase (ERK)/mitogen-activated protein kinase (MAPK) pathway. WHAT IS KNOWN ALREADY: The association between endometriosis and infertility is well supported throughout the literature, and endometriosis per se and its surgical treatment have an adverse effect on the ovarian reserve and on oocyte development. MALAT1, one of the most extensively expressed and evolutionarily conserved transcripts, has been implicated to play a role in human development and many diseases. However, little is known about the role of MALAT1 long non-coding RNA (lncRNA) in endometriosis and its associated infertility. STUDY DESIGN, SIZE, DURATION: We measured MALAT1 lncRNA expression levels in GCs from 52 endometriosis patients and 52 controls. Also, MALAT1 was knocked down in a human GC tumor-derived cell line, KGN, to investigate the role of MALAT1 and its molecular mechanism in cell proliferation. PARTICIPANTS/MATERIALS, SETTING, METHODS: GCs were collected from women with or without endometriosis undergoing IVF or ICSI treatment. All endometriosis patients were diagnosed by laparoscopy or laparotomy, and control patients were limited to male factor or tubal disease and had a normal ovarian reserve. Quantitative real-time PCR (qRT-PCR) was used to measure the differential expression levels of MALAT1 lncRNA between endometriosis patients and controls. The receiver operating characteristic (ROC) curve was drawn to evaluate the diagnostic values of MALAT1 in endometriosis. In the KGN cell line, MALAT1 was knocked down with locked nucleic acid GapmeRs. Cell counting kit-8 assays, ethynyl-2-deoxyuridine assays and flow cytometry were used to study the role of MALAT1 in cell proliferation and cell-cycle progression, and western blotting was performed to detect the potential underlying mechanism. MAIN RESULTS AND THE ROLE OF CHANCE: We first found that MALAT1 lncRNA was significantly down-regulated in endometriosis GCs and was associated with the antral follicle count (R = 0.376, P < 0.001 versus control). In addition, MALAT1 lncRNA levels were significantly lower in the GCs of infertile women with advanced stages of endometriosis (P = 0.01 versus control). The ROC curves illustrated strong separation between all the endometriosis patients and the control group (AUC: 0.705; 95% CI: 0.606-0.804; P < 0.001), Stage I-II and control group (AUC: 0.651; 95% CI: 0.536-0.767; P = 0.016), and Stage III-IV and control group (AUC: 0.827; 95% CI: 0.718-0.936; P < 0.001). MALAT1 lncRNA was primarily localized in the nuclei of GCs. We found a negative correlation between MALAT1 lncRNA and P21 mRNA in the GCs from patients (R = -0.628; P < 0.001). MALAT1 knockdown in KGN cells inhibited cell proliferation and cell-cycle progression. In addition, MALAT1 knockdown induced an increase in both the mRNA and protein levels of P21, and of P53, phosphorylated ERK1/2 (p-ERK1/2) and phosphorylated c-Jun N-terminal protein kinase (p-JNK) protein levels, as well as causing a decrease in cyclin dependent kinase 2 (CDK2), cyclin D1 and p-P38 MAPK protein levels. Furthermore, inhibition of the ERK/MAPK pathway with U0126, the up-regulation of p-ERK1/2, P21 and P53, and the down-regulation of CDK2 and cyclin D1 by the knockdown of MALAT1 were all attenuated by MALAT1 knockdown. Therefore, MALAT1 may regulate GC proliferation through P21/P53-dependent control of the cell cycle, and the ERK/MAPK pathway participates in this process. LARGE SCALE DATA: None. LIMITATIONS, REASONS FOR CAUTION: The hormonal treatment used in IVF and surgical removal of endometriotic lesions may have altered MALAT1 expression in GCs. The ovarian granulosa-like tumor cell line, KGN, was used for further functional and mechanistic studies due to the difficulties in obtaining human GCs in sizable amounts and maintaining primary cultures. WIDER IMPLICATIONS OF THE FINDINGS: Our finding represents the first example of an lncRNA-based mechanism in endometriosis GCs. Women with endometriosis show altered MALAT1 expression levels in GCs that may impair fertility by regulating the function of GCs. Therefore, analysis of MALAT1 and its molecular mechanisms of action provide new insights into the pathogenesis of endometriosis and its associated infertility. STUDY FUNDING/COMPETING INTEREST(s): This work was supported by the National Natural Science Foundation of China (grant number: 81671524) and the National key research and development program of China (grant number: 2017YFC1001100). The authors declare there is no conflict of interest.


Subject(s)
Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Endometriosis/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Granulosa Cells/metabolism , MAP Kinase Signaling System/physiology , RNA, Long Noncoding/metabolism , Cell Line , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation , Endometriosis/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Humans , MAP Kinase Signaling System/genetics , RNA, Long Noncoding/genetics
16.
Curr Med Sci ; 38(3): 505-512, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30074219

ABSTRACT

Increasing evidence suggests that epigenetic dysfunction may influence the stability of normal pregnancy. The ten-eleven translocation (TET) family and 5-hydroxymethylcytosine (5-hmC) were found to be linked with epigenetic reprogramming. The present study aimed to examine the expression of the TET family and 5-hmC in the villi of human embryos and compared their expression between normal pregnancy and early pregnancy loss (EPL). Embryonic villi were collected from normal pregnant women (control) experiencing medical abortion and from EPL patients at gestation ages of 6, 7 and 8 weeks. The mRNAs of TET family were analysed using quantitative polymerase chain reaction (qPCR), and TET proteins using Western blotting and immunohistochemical analysis. The MethylFlash™ Kit was used to quantify the absolute amount of 5-methylcytosine (5-mC) and 5-hmC. Our results showed that the expression of the TETs and 5-hmC in the normal villus decreased with increasing gestational age. Immunohistochemistry revealed that the TET proteins were expressed in the cytoplasm of trophoblasts and their expression was the highest in the 6-week tissue samples, which was consistent with the qPCR and Western blot results. The expression of TET1, TET2, and TET3 was lower in the villi in EPL group than in normal pregnancy group (P<0.05 for all). It was concluded that the TET family and 5-hmC are critical in epigenetic reprogramming of human embryo. The findings also suggest that a deficiency of TETs in the villus might be associated with human EPL.


Subject(s)
5-Methylcytosine/analogs & derivatives , Abortion, Spontaneous/metabolism , Proto-Oncogene Proteins/metabolism , Trophoblasts/metabolism , 5-Methylcytosine/metabolism , Adult , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases/genetics , Dioxygenases/metabolism , Female , Humans , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Pregnancy , Proto-Oncogene Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trophoblasts/pathology
17.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(5): 601-605, 2018 May 20.
Article in Chinese | MEDLINE | ID: mdl-29891459

ABSTRACT

OBJECTIVE: To explore whether a high serum estradiol (E2) level before progesterone administration adversely affects the pregnancy outcomes of frozen-thawed embryo transfer (FET) cycles. METHODS: We retrospectively analyzed 205 hormone replacement therapy (HRT)-FET cycles in our Center between February, 2017 and August, 2017. With a cutoff value of serum E2 level of 600 pg/mL before progesterone administration, the cases were divided into high E2 level group and control group with normal E2 level, and the clinical characteristics and pregnancy outcomes were compared between the two groups. RESULTS: No significant difference was found between the two groups in the patients'age during IVF/ICSI cycle, body mass index (BMI) or endometrial thickness at the time of FET (P>0.05). The patients with high E2 levels had a significantly younger age (P<0.05) and a significantly longer duration of estradiol administration than those in the control group (P<0.05). The clinical pregnancy rates, ongoing pregnancy rates, early miscarriage rates, late abortion rates and live birth rates were all comparable between the two groups (P>0.05). After controlling for the compounding factors including the age at FET cycle and the duration of estradiol administration, all these pregnancy outcomes were still comparable between the two groups. CONCLUSION: A high serum E2 level before progesterone administration does not adversely affect the pregnancy outcomes of HRT-FET cycles.


Subject(s)
Embryo Transfer , Estradiol/blood , Pregnancy Outcome , Progesterone/administration & dosage , Progestins/administration & dosage , Age Factors , Estrogen Replacement Therapy/statistics & numerical data , Female , Humans , Pregnancy , Pregnancy Rate , Retrospective Studies
18.
Nan Fang Yi Ke Da Xue Xue Bao ; 37(10): 1418-1422, 2017 Oct 20.
Article in Chinese | MEDLINE | ID: mdl-29070478

ABSTRACT

OBJECTIVE: We report a case of ovarian function fluctuation during long-term follow-up in a patient with premature ovarian insufficiency (POI). The patient finally obtained clinical pregnancy with subsequent uneventful full-term delivery after several intracytoplasmic sperm injection-embryo transfer (ICSI-ET) cycles. This case demonstrates that hormone replacement therapy (HRT) and assisted reproductive therapy should be applied as soon as possible to young patients with POI who have a strong desire for pregnancy in the absence of contraindications. This strategy helps such patients obtain pregnancy and delivery before the exhaustion of ovarian function.


Subject(s)
Embryo Transfer , Primary Ovarian Insufficiency/therapy , Sperm Injections, Intracytoplasmic , Female , Hormone Replacement Therapy , Humans , Pregnancy , Pregnancy Rate
19.
Endocrinology ; 158(11): 3890-3899, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28938484

ABSTRACT

Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility in women of reproductive age, and its etiology remains poorly understood. Altered activities of long noncoding RNAs (lncRNAs) have been associated with human diseases and development. However, the roles of lncRNAs are unknown in reproductive medicine. We investigated the potential role of lncRNAs in the pathogenesis of PCOS, using human granulosa cells (GCs) and the KGN cell line. We used microarrays to compare lncRNA expression profiles in GCs from seven patients with PCOS and seven matched women. GC samples were collected during 2014 to 2016 from infertile women in Guangzhou, China. Quantitative real-time polymerase chain reaction was used to measure levels of the lncRNA HCG26 in GCs from 53 patients with PCOS and 50 controls. HCG26 was knocked down with locked nucleic acid GapmeRs in KGN cells to examine its role in cell proliferation, aromatase and follicle-stimulating hormone receptor gene expression, and estradiol production. A total of 862 lncRNA transcripts and 998 messenger RNA transcripts were differentially expressed (greater than or equal to twofold change; P < 0.05) in PCOS GCs compared with those of controls. HCG26 levels were upregulated in patients with PCOS and were associated with antral follicle count. HCG26 knockdown in KGN cells inhibited cell proliferation and cell-cycle progression and increased aromatase gene expression and estradiol production. Our study reports the lncRNA profiles in GCs from patients who have PCOS and those from healthy women and suggests that dysregulated lncRNAs may play vital roles in GC proliferation and steroidogenesis, providing insights into the pathogenesis of PCOS.


Subject(s)
Polycystic Ovary Syndrome/genetics , RNA, Long Noncoding/physiology , Adult , Case-Control Studies , Cell Line , Female , Gene Expression Profiling , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Infertility, Female/genetics , Microarray Analysis , Young Adult
20.
J Huazhong Univ Sci Technolog Med Sci ; 37(3): 313-318, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28585134

ABSTRACT

Proper chromosome separation in both mitosis and meiosis depends on the correct connection between kinetochores of chromosomes and spindle microtubules. Kinetochore dysfunction can lead to unequal distribution of chromosomes during cell division and result in aneuploidy, thus kinetochores are critical for faithful segregation of chromosomes. Centromere protein A (CENP-A) is an important component of the inner kinetochore plate. Multiple studies in mitosis have found that deficiencies in CENP-A could result in structural and functional changes of kinetochores, leading to abnormal chromosome segregation, aneuploidy and apoptosis in cells. Here we report the expression and function of CENP-A during mouse oocyte meiosis. Our study found that microinjection of CENP-A blocking antibody resulted in errors of homologous chromosome segregation and caused aneuploidy in eggs. Thus, our findings provide evidence that CENP-A is critical for the faithful chromosome segregation during mammalian oocyte meiosis.


Subject(s)
Centromere Protein A/genetics , Chromosome Segregation , Chromosomes, Mammalian/metabolism , Kinetochores/metabolism , Meiosis , Spindle Apparatus/metabolism , Aneuploidy , Animals , Antibodies, Neutralizing/pharmacology , Apoptosis , Centromere Protein A/antagonists & inhibitors , Centromere Protein A/metabolism , Chromosomes, Mammalian/ultrastructure , Female , Gene Expression Regulation , Karyotyping , Kinetochores/ultrastructure , Mice , Mice, Inbred ICR , Microinjections , Microtubules/metabolism , Microtubules/ultrastructure , Oocytes/metabolism , Oocytes/ultrastructure , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spindle Apparatus/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...