Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.329
Filter
3.
Fertil Steril ; 121(5): 765-782, 2024 May.
Article in English | MEDLINE | ID: mdl-38163620

ABSTRACT

There is controversy regarding whether to treat subtle abnormalities of thyroid function in infertile female patients. This guideline document reviews the risks and benefits of treating subclinical hypothyroidism in female patients with a history of infertility and miscarriage, as well as obstetric and neonatal outcomes in this population.


Subject(s)
Asymptomatic Diseases , Hypothyroidism , Infertility, Female , Humans , Female , Hypothyroidism/diagnosis , Hypothyroidism/epidemiology , Hypothyroidism/complications , Hypothyroidism/therapy , Infertility, Female/therapy , Infertility, Female/epidemiology , Infertility, Female/diagnosis , Infertility, Female/physiopathology , Pregnancy , Risk Factors , Treatment Outcome
4.
J Obstet Gynaecol ; 43(1): 2144177, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36397660

ABSTRACT

This study evaluated the effect of intrauterine perfusion of autologous platelet-rich plasma (PRP) on pregnancy outcomes in women with recurrent implantation failure (RIF). Key biomedical databases were searched to identify relevant clinical trials and observational studies. Outcomes included clinical pregnancy rate, chemical pregnancy rate, implantation rate, live birth rate, and abortion rate. Data was extracted from ten studies (six randomised controlled trials, four cohort studies) involving 1555 patients. Pregnancy outcomes were improved in women treated with PRP compared to controls: clinical pregnancy rate (RR = 1.96, 95% CI [1.67, 2.31], p < 0.00001, I2 = 46%), chemical pregnancy rate (RR = 1.79, 95% CI [1.54, 2.08], p < 0.00001, I2 = 29%), implantation rate (RR = 1.90, CI [1.50, 2.41], p < 0.00001, I2 = 0%), live birth rate (RR = 2.83, CI [1.45, 5.52], p = 0.0007, I2 = 83%), abortion rate (RR = 0.40, 95% CI [0.18, 0.90], p = 0.03, I2 = 59%). These data imply PRP has potential to improve pregnancy outcomes in women with RIF, suggesting a promising role in assisted reproductive technology.IMPACT STATEMENTWhat is already known on this subject? Platelet-rich plasma (PRP) is an autologous blood product that contains platelets, various growth factors, and cytokines at concentrations above the normal baseline level. Recent studies have shown that intrauterine infusion of autologous PRP can improve pregnancy outcomes in infertile women.What do the results of this study add? This systematic review and meta-analysis of data from ten studies (n = 1555; 775 cases and 780 controls) investigated the effect of intrauterine perfusion of autologous PRP on pregnancy outcomes in women with recurrent implantation failure (RIF). Findings suggest that pregnancy outcomes, including clinical pregnancy rate, chemical pregnancy rate, implantation rate, live birth rate and abortion rate were improved in women treated with PRP compared to controls.What are the implications of these findings for clinical practice and/or further research? RIF remains a challenge for researchers, clinicians, and patients. Our study identified PRP as a potential intervention in assisted reproduction. As an autologous blood preparation, PRP eliminates the risk of an immune response and transmission of disease. PRP is low cost and effective and may represent a new approach to the treatment of patients with RIF.


Subject(s)
Abortion, Spontaneous , Blood Transfusion, Autologous , Embryo Implantation , Infertility, Female , Platelet-Rich Plasma , Uterus , Female , Humans , Pregnancy , Abortion, Spontaneous/prevention & control , Embryo Implantation/physiology , Infertility, Female/physiopathology , Infertility, Female/therapy , Live Birth , Platelet-Rich Plasma/physiology , Pregnancy Outcome , Pregnancy Rate , Uterus/physiopathology , Administration, Topical , Blood Transfusion, Autologous/methods
5.
Reprod Biol Endocrinol ; 20(1): 54, 2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35313911

ABSTRACT

BACKGROUND: Both mild and conventional controlled ovarian stimulation are the frequently used protocols for poor ovarian responders. However, there are some debates about which treatment is better. Moreover, little is known about the follicular physiology after the two ovarian stimulation protocols. This study was intended to investigate the features in granulosa cells and follicular fluid micro-environment after the two different ovarian stimulation protocols in poor responders. METHODS: Granulosa cells RNA were sequenced using Illumina Hiseq technology. Specific differently expressed genes and proteins were verified by real-time quantitative PCR and Western blot analysis. Moreover, hormone and cytokine concentrations in the follicular fluid were measured by electrochemiluminescence immunoassay and enzyme-linked immunoabsorbent assay. The correlation between the results of molecular experiments and the laboratory outcomes were analyzed by Spearman correlation analysis. RESULTS: The differentially expressed genes between the two groups were involved in 4 signaling pathways related to the follicular development; three proteins pertinent to the TGF-ß signaling pathway were expressed differently in granulosa cells between the two, and the constituents in the follicular fluid were also different. Further, a correlation between the TGF-ß signaling pathway and the good-quality embryo was observed. CONCLUSIONS: The present study made a comparison for the first time in the transcriptome of human granulosa cells and the follicular fluid micro-environment between poor responders with the conventional controlled ovarian stimulation or the mild ovarian stimulation, showing that the TGF-ß signaling pathway may correlate with the good-quality of embryos in the mild group, which may be instrumental to the choice of optimal management for IVF patients.


Subject(s)
Follicular Fluid/metabolism , Granulosa Cells/metabolism , Infertility, Female/genetics , Ovulation Induction/methods , Transcriptome , Adult , Case-Control Studies , Cellular Microenvironment/genetics , Female , Follicular Fluid/chemistry , Gene Expression Profiling , Granulosa Cells/chemistry , High-Throughput Nucleotide Sequencing , Humans , Infertility, Female/metabolism , Infertility, Female/physiopathology , Oocyte Retrieval , Ovarian Reserve/genetics , Ovulation/genetics , Sequence Analysis, DNA , Treatment Failure
6.
Fertil Steril ; 117(2): 408-418, 2022 02.
Article in English | MEDLINE | ID: mdl-35125179

ABSTRACT

OBJECTIVE: To estimate the probability of clinical or multiple pregnancy during ovulation induction (OI)/ovarian stimulation (OS). DESIGN: Secondary analysis of two multicenter randomized clinical trials (combined). SETTING: Multicenter. PATIENTS: A total of 750 women with polycystic ovary syndrome and 900 women with unexplained infertility. INTERVENTIONS: Ovulation induction/OS with either timed intercourse (polycystic ovary syndrome) or intrauterine insemination. MAIN OUTCOME MEASURES: Clinical and multiple pregnancy rates/cycle, cumulative pregnancy rates. Age, body mass index, parity, diagnosis, medication, markers of ovarian reserve, and ovarian response were considered in multivariable regression models for clinical, multiple, and cumulative pregnancy rates. Receiver operating characteristic curves were created for clinical and multiple pregnancy rates. RESULTS: Younger patient and partner age, treatment type, lower body mass index, and medication dose were all associated with clinical pregnancy. Variables associated with multiple pregnancy included the abovementioned variables (except age), in addition to diagnosis, parity, higher antral follicle count, antimüllerian hormone levels, and ovarian response. Gonadotropin use was associated with multiple pregnancy, with progressively increasing odds ratios (cycles 1-4). Receiver operating characteristic curves indicated the model's predictive power to be fair for clinical pregnancy (areas under the curve [95% confidence interval {CI}]: 0.78 [0.75-0.81] for cycle 1 and 0.70 [0.64-0.75] for cycle 4) and good-to-excellent for multiple pregnancy (areas under the curve [95% CI]: 0.78 [0.72-0.84] for cycle 1 and 0.86 [0.78-0.93] for cycle 4). Partner age, lower medication dose, parity, antimüllerian hormone levels, and diagnosis were associated with cumulative pregnancy rates. CONCLUSIONS: Using the majority of the factors known to predict the outcome of OI/OS cycles, we constructed an easy-to-use formula that may predict individualized chances of clinical and multiple pregnancy for commonly used fertility treatments (https://pregnancyprediction.medicine.yale.edu/CalDirect.html). CLINICAL TRIAL REGISTRATION NUMBERS: Assessing Multiple Intrauterine Gestations after Ovulation Stimulation NCT01044862; PPCOSII NCT00719186.


Subject(s)
Decision Support Techniques , Infertility, Female/therapy , Ovulation Induction , Polycystic Ovary Syndrome/therapy , Precision Medicine , Adult , Body Mass Index , Clinical Decision-Making , Coitus , Female , Fertility , Fertility Agents, Female/therapeutic use , Humans , Infertility, Female/diagnosis , Infertility, Female/physiopathology , Insemination, Artificial , Male , Maternal Age , Polycystic Ovary Syndrome/diagnosis , Polycystic Ovary Syndrome/physiopathology , Pregnancy , Pregnancy Rate , Randomized Controlled Trials as Topic , Risk Assessment , Risk Factors , Treatment Outcome
7.
Proc Natl Acad Sci U S A ; 119(8)2022 02 22.
Article in English | MEDLINE | ID: mdl-35169075

ABSTRACT

Thin endometrium has been widely recognized as a critical cause of infertility, recurrent pregnancy loss, and placental abnormalities; however, access to effective treatment is a formidable challenge due to the rudimentary understanding of the pathogenesis of thin endometrium. Here, we profiled the transcriptomes of human endometrial cells at single-cell resolution to characterize cell types, their communications, and the underlying mechanism of endometrial growth in normal and thin endometrium during the proliferative phase. Stromal cells were the most abundant cell type in the endometrium, with a subpopulation of proliferating stromal cells whose cell cycle signaling pathways were compromised in thin endometrium. Both single-cell RNA sequencing and experimental verification revealed cellular senescence in the stroma and epithelium accompanied by collagen overdeposition around blood vessels. Moreover, decreased numbers of macrophages and natural killer cells further exacerbated endometrial thinness. In addition, our results uncovered aberrant SEMA3, EGF, PTN, and TWEAK signaling pathways as causes for the insufficient proliferation of the endometrium. Together, these data provide insight into therapeutic strategies for endometrial regeneration and growth to treat thin endometrium.


Subject(s)
Endometrium/metabolism , Endometrium/pathology , Endometrium/physiology , Carrier Proteins/metabolism , Cytokine TWEAK/metabolism , Cytokines/metabolism , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , Epithelium , Female , Gene Expression/genetics , Humans , Infertility, Female/etiology , Infertility, Female/physiopathology , Semaphorin-3A/genetics , Semaphorin-3A/metabolism , Signal Transduction/genetics , Single-Cell Analysis , Stromal Cells/metabolism , Transcriptome/genetics
8.
Mol Biol Rep ; 49(2): 1519-1528, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34988892

ABSTRACT

Polycystic Ovarian Syndrome (PCOS) is an endocrine disorder commonly affecting the reproductive capacity of women leading to infertility. PCOS-related infertility is majorly due to anovulation; however, it is not the only cause. The defective endometrium causing recurrent miscarriage and implantation failure can also be accountable for infertility in PCOS women. The unusual levels of hormones and their receptors in the PCOS endometrium have a hostile effect during WOI, making the microenvironment unfavorable for embryo implantation. To date, many studies have been performed to determine the role of candidate genes in endometrial receptivity but very limited data is available using whole genome approach. This review aims at summarizing the existing studies on the basic aspects of endometrial receptivity in PCOS. The review focuses on aberrant levels of hormones and their receptors in the endometrium, affecting the receptivity. Additionally, it explores the novel approach reviewing the effect on treatment options administered for ovulation induction in PCOS on their endometrial receptivity. Overall, this review will help us to understand the molecular milieu in PCOS endometrium and its effect on the receptivity potential. However, to have a thorough understanding of the mechanistic approach of hormonal imbalance in PCOS on endometrial receptivity, there is a need to give more weightage to genome-wide studies in the future. The current review will further guide us to formulate future studies using whole genome technologies for the assessment of endometrial receptivity in different cohorts of PCOS women, which may have future diagnostic implementations.


Subject(s)
Endometrium/pathology , Infertility, Female/etiology , Polycystic Ovary Syndrome/physiopathology , Embryo Implantation , Endometrium/metabolism , Female , Humans , Infertility, Female/physiopathology , Ovulation Induction
9.
Sci Rep ; 12(1): 402, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013422

ABSTRACT

There is a lack of studies assessing how hearing impairment relates to reproductive outcomes. We examined whether childhood hearing impairment (HI) affects reproductive patterns based on longitudinal Norwegian population level data for birth cohorts 1940-1980. We used Poisson regression to estimate the association between the number of children ever born and HI. The association with childlessness is estimated by a logit model. As a robustness check, we also estimated family fixed effects Poisson and logit models. Hearing was assessed at ages 7, 10 and 13, and reproduction was observed at adult ages until 2014. Air conduction hearing threshold levels were obtained by pure-tone audiometry at eight frequencies from 0.25 to 8 kHz. Fertility data were collected from Norwegian administrative registers. The combined dataset size was N = 50,022. Our analyses reveal that HI in childhood is associated with lower fertility in adulthood, especially for men. The proportion of childless individuals among those with childhood HI was almost twice as large as that of individuals with normal childhood hearing (20.8% vs. 10.7%). The negative association is robust to the inclusion of family fixed effects in the model that allow to control for the unobserved heterogeneity that are shared between siblings, including factors related to the upbringing and parent characteristics. Less family support in later life could add to the health challenges faced by those with HI. More attention should be given to how fertility relates to HI.


Subject(s)
Fertility , Hearing Loss/epidemiology , Hearing , Infertility, Female/epidemiology , Infertility, Male/epidemiology , Persons With Hearing Impairments , Reproduction , Adolescent , Age Factors , Aged , Audiometry, Pure-Tone , Auditory Threshold , Child , Family Characteristics , Female , Hearing Loss/diagnosis , Hearing Loss/physiopathology , Humans , Infertility, Female/diagnosis , Infertility, Female/physiopathology , Infertility, Male/diagnosis , Infertility, Male/physiopathology , Longitudinal Studies , Male , Middle Aged , Norway/epidemiology , Reproductive Behavior , Risk Assessment , Risk Factors , Sex Factors , Time Factors
10.
Reprod Sci ; 29(2): 620-626, 2022 02.
Article in English | MEDLINE | ID: mdl-34363198

ABSTRACT

The Pre-IVF Treatment with a GnRH Antagonist in Women with Endometriosis (PREGnant) Trial (clinicaltrials.gov no. NCT04173169) was designed to test the hypothesis that 60-day pre-treatment with an oral GnRH antagonist in women with documented endometriosis and planning an IVF cycle will result in a superior live birth rate to placebo. Eight hundred fourteen women are required from 4 national sites. To determine the feasibility of using an electronic medical record (EMR)-based strategy to recruit 204 participants at the Colorado site, we conducted a survey of women within the UCHealth system. Eligible women, identified using relevant ICD-10 codes, were invited to complete a 6-question survey to assess planned utilization of IVF, potential interest in participation, and whether delays in treatment due to COVID-19 would influence their decision to participate. Of 6354 age-eligible women with an endometriosis diagnosis, 421 had a concurrent infertility diagnosis. After eliminating duplicates, 212 were emailed a survey; 76 (36%) responded, 6 of whom reported no endometriosis diagnosis. Of the remaining 70, 29 (41%) were planning fertility treatment; only 19 planned IVF. All 19 expressed interest in participation. COVID-19 delays in treatment were not considered as a factor affecting participation by 8/19; the remaining 11 felt that it would "somewhat" affect their decision. None reported that they would not consider participation because of COVID-19. EMR-based recruitment for an endometriosis clinical trial is feasible although the overall yield of participants is low. Delays in treatment due to COVID-19 did not appear to overly influence potential recruitment.


Subject(s)
COVID-19 , Endometriosis/therapy , Fertility Agents, Female/therapeutic use , Fertilization in Vitro , Health Knowledge, Attitudes, Practice , Hormone Antagonists/therapeutic use , Infertility, Female/therapy , Patient Selection , Research Subjects/psychology , Adolescent , Adult , Choice Behavior , Double-Blind Method , Electronic Health Records , Endometriosis/diagnosis , Endometriosis/physiopathology , Female , Fertility Agents, Female/adverse effects , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Hormone Antagonists/adverse effects , Humans , Infertility, Female/diagnosis , Infertility, Female/physiopathology , Live Birth , Pregnancy , Pregnancy Rate , Treatment Outcome , United States , Young Adult
11.
Clin Pharmacol Ther ; 111(2): 382-390, 2022 02.
Article in English | MEDLINE | ID: mdl-33826770

ABSTRACT

We reviewed the available animal and human reproductive function studies of recently approved noncytotoxic oncology drugs. We reviewed the oncofertility information in the prescribing information for the US Food and Drug Administration (FDA)-approved products and/or the product information and consumer medicine information for Therapeutic Goods Administration (TGA)-approved drugs of 32 novel oncology drugs approved between 2014 and 2018 in the United States and/or Australia supplemented by a literature review for additional reproductive effects. No human studies were available on the reproductive effects of all 32 drugs. A systematic literature review of animal reproductive toxicity studies provided only very limited data with nine drugs displaying impaired male fertility, three impaired female fertility, and nine producing impaired fertility in both male and female animals. Two drugs in the study are reported to have no demonstrable impact on fertility in animal reproductive toxicity studies and nine are reported to have unknown effects on fertility. Of the 32 newly listed drugs, only 4 had recommendations regarding potential human fertility risks and accordingly advised clinicians about fertility preservation procedures for patients. The lack of human data and limited animal reproductive toxicity data raises concerns about the potential impact of these novel oncology drugs on human fertility and reproductive function. Consequently, adequate oncofertility recommendations, including for fertility preservation procedures, counselling for psychological or cost implications, and future prognosis for fertility are hindered by this paucity of relevant data. More data on human reproductive effects of novel oncology drugs is urgently required to facilitate effective use of the growing array of oncofertility care options available.


Subject(s)
Antineoplastic Agents/adverse effects , Fertility Preservation , Fertility/drug effects , Infertility, Female/chemically induced , Infertility, Male/chemically induced , Animals , Female , Humans , Infertility, Female/physiopathology , Infertility, Female/therapy , Infertility, Male/physiopathology , Infertility, Male/therapy , Male , Risk Assessment , Risk Factors
12.
Toxicology ; 465: 153031, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34774661

ABSTRACT

Per- and poly-fluoroalkyl substances (PFAS) are widespread environmental contaminants frequently detected in drinking water supplies worldwide that have been linked to a variety of adverse reproductive health outcomes in women. Compared to men, reproductive health effects in women are generally understudied while global trends in female reproduction rates are declining. Many factors may contribute to the observed decline in female reproduction, one of which is environmental contaminant exposure. PFAS have been used in home, food storage, personal care and industrial products for decades. Despite the phase-out of some legacy PFAS due to their environmental persistence and adverse health effects, alternative, short-chain and legacy PFAS mixtures will continue to pollute water and air and adversely influence women's health. Studies have shown that both long- and short-chain PFAS disrupt normal reproductive function in women through altering hormone secretion, menstrual cyclicity, and fertility. Here, we summarize the role of a variety of PFAS and PFAS mixtures in female reproductive tract dysfunction and disease. Since these chemicals may affect reproductive tissues directly or indirectly through endocrine disruption, the role of PFAS in breast, thyroid, and hypothalamic-pituitary-gonadal axis function are also discussed as the interplay between these tissues may be critical in understanding the long-term reproductive health effects of PFAS in women. A major research gap is the need for mechanism of action data - the targets for PFAS in the female reproductive and endocrine systems are not evident, but the effects are many. Given the global decline in female fecundity and the ability of PFAS to negatively impact female reproductive health, further studies are needed to examine effects on endocrine target tissues involved in the onset of reproductive disorders of women.


Subject(s)
Endocrine Disruptors/adverse effects , Endocrine System Diseases/chemically induced , Environmental Exposure/adverse effects , Environmental Pollutants/adverse effects , Fertility/drug effects , Hydrocarbons, Fluorinated/adverse effects , Menstrual Cycle/drug effects , Reproduction/drug effects , Endocrine System Diseases/metabolism , Endocrine System Diseases/physiopathology , Female , Humans , Infertility, Female/chemically induced , Infertility, Female/metabolism , Infertility, Female/physiopathology , Pregnancy , Pregnancy Complications/chemically induced , Pregnancy Complications/metabolism , Pregnancy Complications/physiopathology , Prognosis , Risk Assessment , Risk Factors
14.
Front Endocrinol (Lausanne) ; 12: 690575, 2021.
Article in English | MEDLINE | ID: mdl-34867773

ABSTRACT

Protocols utilizing gonadotropin-releasing hormone (GnRH) antagonists have emerged as mainstream procedures for ovarian stimulation; however, GnRH increases the risk for periodic cancellation of embryos. Therefore, this study aimed to compare the pregnancy outcomes of a fixed GnRH antagonist protocol and a flexible progestin-primed ovarian stimulation (fPPOS) protocol in patients with asynchronous follicular development during controlled ovulation stimulation and to explore the feasibility of converting patients undergoing a fixed GnRH antagonist protocol to an fPPOS protocol. This was the first retrospective study exploring the fPPOS protocol in patients with asynchronous follicular development, and it was conducted in a public reproductive medicine center from January to December 2020. We included infertile women. All participants were scheduled to undergo administration of a GnRH antagonist on the fifth day of controlled ovulation stimulation. The study group included 129 women who were converted from the fixed GnRH antagonist protocol to the fPPOS protocol for their asynchronous follicular development, while the antagonist group consisted of 258 women (ratio 1:2) who proceeded with a fixed GnRH antagonist protocol. On the second or third day of the menstrual period, 100-300 IU/day gonadotropin injections were administered. For patients who were converted to the fPPOS protocol, medroxyprogesterone acetate tablets at 10 mg/day were started on the fifth day of stimulation or when only one leading follicle reached 14 mm and the other follicles were ≤10 mm in diameter, whichever came first. The rates of embryo implantation, clinical pregnancy, and early pregnancy loss were obtained. The number of oocytes retrieved and the number of high-quality embryos in the antagonist group were significantly higher than those in the fPPOS group (P = 0.039 and P = 0.025, respectively). No significant differences in the rates of embryo implantation, clinical pregnancy, and early pregnancy loss were observed between the two groups. Our study found that in patients who were scheduled for administration of GnRH antagonists but presented with asynchronous follicular development on the fifth stimulation day, it was feasible to switch to the fPPOS protocol.


Subject(s)
Hormone Antagonists/administration & dosage , Infertility, Female/therapy , Ovulation Induction/methods , Progestins/administration & dosage , Adult , Cohort Studies , Drug Administration Schedule , Female , Fertilization in Vitro/methods , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Humans , Infant, Newborn , Infertility, Female/physiopathology , Male , Ovarian Follicle/drug effects , Ovarian Follicle/physiopathology , Ovulation/drug effects , Ovulation/physiology , Pregnancy , Pregnancy Outcome , Retrospective Studies , Sperm Injections, Intracytoplasmic/methods , Treatment Outcome
15.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34768846

ABSTRACT

The diagnosis of endometriosis and fertility disorders is difficult; therefore, it is necessary to look for reliable biomarkers. Analysis of the molecular status of fibronectin as a key player in repair and wound healing processes, as well as in coagulation and fibrinolysis pathways, is justified. ELISA and SDS-agarose immunoblotting were applied to determine the fibronectin concentration and presence and occurrence of soluble FN-fibrin complexes in the blood plasma of women with endometriosis (n = 38), fertility disorders (n = 28) and the healthy group (n = 25). The concentration of fibronectin in the blood plasma of women with endometriosis (292.61 ± 96.17 mg/L) and fertility disorders (287.53 ± 122.68 mg/L) was significantly higher than in the normal group (226.55 ± 91.98 mg/L). The presence of FN-fibrin complexes of 750, 1000, 1300, 1600 and 1900 kDa in the plasma of women with endometriosis and fertility disorders was shown. The presence of FN-fibrin complexes with a molecular mass of more than 1300 kDa in women with endometriosis and infertility and the complete absence of these complexes in healthy women may indicate an increased and chronic activation of coagulation mechanisms in these patients. The presence of complexes of high molecular mass may be one of the biomarkers of fertility disorders in women.


Subject(s)
Endometriosis/metabolism , Fibronectins/metabolism , Infertility, Female/metabolism , Adult , Biomarkers , Endometriosis/diagnosis , Endometriosis/physiopathology , Female , Fibrin/metabolism , Fibrin Fibrinogen Degradation Products/analysis , Fibronectins/analysis , Fibronectins/blood , Fibronectins/physiology , Humans , Infertility, Female/physiopathology , Middle Aged , Plasma/chemistry
16.
J Assist Reprod Genet ; 38(12): 3243-3249, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34846627

ABSTRACT

PURPOSE: Gonadotropin-resistant ovary syndrome (GROS) is a rare endocrine disorder that causes hypergonadotropic hypogonadism, amenorrhea, and infertility. This study reports live birth in two women with GROS who underwent fertility treatment with in vitro maturation (IVM). METHODS: Both patients had primary infertility, amenorrhea (primary and secondary), typical secondary sexual characters, elevated gonadotropin levels, normal ovarian reserve, normal chromosomal characteristics, and previous nonresponsiveness gonadotropin stimulations. One patient had polymorphism of the follicle-stimulating hormone receptor, which is a predictor of poor ovarian response. Given unresponsiveness to exogenous gonadotropin stimulations, IVM with human chorionic gonadotropin priming (hCG-IVM) was performed in both patients. All transferrable embryos were vitrified. RESULTS: Both patients achieved pregnancy after their first frozen embryos transfer, and each delivered a healthy baby boy. CONCLUSIONS: These results suggest that IVM should be a first-line therapeutic option for patients with GROS.


Subject(s)
Chorionic Gonadotropin/metabolism , Infertility, Female/physiopathology , Ovary/physiology , Primary Ovarian Insufficiency/physiopathology , Adult , Embryo Transfer/methods , Female , Fertilization in Vitro/methods , Humans , In Vitro Oocyte Maturation Techniques/methods , Infertility, Female/metabolism , Live Birth , Ovary/metabolism , Pregnancy , Pregnancy, Multiple/metabolism , Pregnancy, Multiple/physiology , Primary Ovarian Insufficiency/metabolism , Receptors, FSH/metabolism
17.
Biomed Res Int ; 2021: 6464686, 2021.
Article in English | MEDLINE | ID: mdl-34746304

ABSTRACT

OBJECTIVE: To explore the mechanisms of follicular fluids (FFs) on granulose cell (GC) apoptosis in endometriosis-associated infertility. MATERIALS AND METHODS: 60 infertile women were enrolled. The FFs from 30 endometriosis-associated infertility (EI) patients were collected and processed by ELISA hormone assay and proteomic profiling. The ovary GCs collected from 30 tubal-associated infertility (TI) patients were cultured in follicular fluids of endometriosis-associated infertility patients (EI-FFs), and the apoptosis mechanisms were explored by flow cytometry assay, real-time PCR, Western blotting, and protein-protein interaction (PPI) network analysis. RESULTS: Our results showed that the expression of 22 specific proteins was significantly different in the FFs from EI and TI patients, and the level of testosterone and anti-Müllerian hormone was not obviously different between the two groups. EI-FFs could accelerate the apoptosis process of granulose cells of tubal-associated infertility patients (TI-GCs) by regulating the expression of 5 apoptosis-related proteins including BCL2, BAX, CASP3, CASP9, and TP53. The correlation of these 22 specific proteins and 5 apoptosis-related proteins was analyzed by PPI, and 5 protein biomarkers (INS, CXCL10, ICAM1, WIF1, and TNFRSF13C) and 5 signaling pathways (cytokine-cytokine receptor interaction, apoptosis, regulation of actin cytoskeleton, MAPK, and p53 signaling pathway) were predicted. CONCLUSION: This research clarified the effect and explored the mechanisms of EI-FFs on the apoptosis of TI-GCs and indicated the protein biomarkers and signaling pathways for further study.


Subject(s)
Follicular Fluid/metabolism , Granulosa Cells/metabolism , Infertility, Female/physiopathology , Adult , Apoptosis/physiology , Case-Control Studies , Endometriosis/metabolism , Endometriosis/physiopathology , Female , Follicular Fluid/physiology , Humans , Infertility, Female/metabolism , Proteomics
18.
Fertil Steril ; 116(5): 1255-1265, 2021 11.
Article in English | MEDLINE | ID: mdl-34607703

ABSTRACT

Diagnostic evaluation for infertility in women should be conducted in a systematic, expeditious, and cost-effective manner to identify all the relevant factors with an initial emphasis on the least invasive methods for detecting the most common causes of infertility. The purpose of this committee opinion is to provide a critical review of the current methods and procedures for the evaluation of in fertile women, and it replaces the document of the same name, last published in 2015 (Fertil Steril 2015;103:e44-50). This guidance is intended for any provider evaluating women for infertility.


Subject(s)
Fertility , Infertility, Female/diagnosis , Ovary/physiopathology , Cervix Uteri/abnormalities , Cervix Uteri/physiopathology , Fallopian Tubes/physiopathology , Female , Humans , Infertility, Female/etiology , Infertility, Female/physiopathology , Infertility, Female/therapy , Ovarian Reserve , Ovulation , Predictive Value of Tests , Pregnancy , Pregnancy Rate , Reproductive Techniques, Assisted , Risk Factors , Treatment Outcome
19.
Front Endocrinol (Lausanne) ; 12: 745199, 2021.
Article in English | MEDLINE | ID: mdl-34646238

ABSTRACT

Objective: To analyze the correlation between ovarian reserve and thyroid function in women with infertility. Methods: Retrospective analysis of the data of 496 infertility patients who visited the clinic between January 2019 and December 2020. According to the TSH level, it is grouped into <2.5 mIU/L, 2.5~4.0mIU/L and ≥4.0 mIU/L or according to the positive/negative thyroid autoimmune antibody. The relationship was assessed through the ovarian reserve, thyroid function, and anti-Müllerian hormone (AMH) levels in infertile patients. On the other hand, the patients are divided into groups according to age (≤29 years old, 30-34 years old and ≥35 years old), basic FSH (<10 IU/L and ≥10 IU/L), and AMH levels. The ovarian reserve was evaluated through the AMH and the antral follicle count (AFC). Results: The average age of the patients was 30.31 ± 4.50 years old, and the average AMH level was 5.13 ± 4.30 ng/mL. 3.63% (18/496) of patients had abnormal TSH levels (normal: 0.35-5.5 mIU/L), the positive rate of thyroid peroxidase antibody (TPOAb) was 14.52% (72/496), the positive rate of anti-thyroglobulin antibody (TgAb) was 16.94% (84/496), and the positive rate of TPOAb and TgAb was 10.48% (52/496). After grouping according to TSH level or thyroid autoimmune antibody positive/negative grouping, the analysis found that there was no statistical significance in age, AMH level and basic FSH level among the groups (P>0.05). There were no significant differences in the levels of TSH, FT3, and FT4 among different ages, AMH, and FSH levels (P>0.05). Conclusion: There is no significant correlation between ovarian reserve and thyroid function in infertile women.


Subject(s)
Infertility, Female , Ovarian Reserve/physiology , Thyroid Hormones/blood , Adult , Age Factors , Anti-Mullerian Hormone/blood , China/epidemiology , Female , Follicle Stimulating Hormone/blood , Humans , Infertility, Female/blood , Infertility, Female/epidemiology , Infertility, Female/pathology , Infertility, Female/physiopathology , Retrospective Studies , Thyroid Diseases/blood , Thyroid Diseases/epidemiology , Thyroid Diseases/physiopathology , Thyroid Function Tests , Thyroid Gland/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...