Andrea Candela, Vincenza De Gregorio, Vincenzo Genovese, Angela Travaglione, Mario Cimmino, Riccardo Talevi, Roberto Gualtieri
{"title":"Multi-oocyte follicles in the bovine ovary: occurrence, activation, and growth during early in vitro folliculogenesis in a dynamic culture bioreactor.","authors":"Andrea Candela, Vincenza De Gregorio, Vincenzo Genovese, Angela Travaglione, Mario Cimmino, Riccardo Talevi, Roberto Gualtieri","doi":"10.1007/s10815-025-03600-8","DOIUrl":"https://doi.org/10.1007/s10815-025-03600-8","url":null,"abstract":"<p><strong>Purpose: </strong>Multi-oocyte follicles (MOFs) have been identified in various mammals, including humans, yet their origin and function remain controversial. This study aimed to investigate the occurrence and early developmental dynamics of MOFs in bovine ovaries, comparing them to single-oocyte follicles (SOFs).</p><p><strong>Methods: </strong>Ovarian tissues from Bos taurus taurus individuals, including one case with an unusually high MOF incidence, were cultured in vitro under dynamic conditions. MOF frequency and their progression through early folliculogenesis stages were assessed histologically and via confocal microscopy.</p><p><strong>Results: </strong>MOFs were observed at varying frequencies, with one individual showing a notably high incidence (15.8%). In vitro culture confirmed that MOFs can activate and progress to secondary stages, similar to SOFs. However, differences in activation rates and oocyte number dynamics were noted between MOFs and SOFs, and between fresh and cultured tissues. No de novo formation of MOFs was detected in vitro.</p><p><strong>Conclusions: </strong>MOFs retain the capacity for early folliculogenesis comparable to SOFs, without increased atresia. Their stable frequency postculture supports a prenatal origin. These findings offer new insights into MOF biology and suggest a possible physiological relevance in mammalian reproductive systems.</p>","PeriodicalId":15246,"journal":{"name":"Journal of Assisted Reproduction and Genetics","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-08-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144804192","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Xin Yuan, Jiajie Li, Tang Deng, Li Yang, Yilun Wu, Liangchen Xiang, Fang Yuan, Xinrui Sun, Yi Xia, Yihong Yang, Yi Zhang, Fang Ma, Yan Wang
{"title":"Spermatozoal sialidases NEU1 and NEU3 are potential predictors for fertilization rate in IVF: a prospective cohort study.","authors":"Xin Yuan, Jiajie Li, Tang Deng, Li Yang, Yilun Wu, Liangchen Xiang, Fang Yuan, Xinrui Sun, Yi Xia, Yihong Yang, Yi Zhang, Fang Ma, Yan Wang","doi":"10.1007/s10815-025-03602-6","DOIUrl":"https://doi.org/10.1007/s10815-025-03602-6","url":null,"abstract":"<p><strong>Purpose: </strong>This study aimed to explore the roles of spermatozoal sialidase NEU1 and NEU3 in affecting the fertilization rate of in vitro fertilization (IVF), and whether sperm sialidase NEU1 and NEU3 can be used as predictors of IVF outcome.</p><p><strong>Material and methods: </strong>This was a prospective cohort study that collected semen samples from 194 IVF couples between January 2024 and April 2024. Patients were grouped based on oocyte maturity and routine semen analysis. Detection of sperm NEU1 and NEU3 was carried out in semen from the same ejaculate for IVF by using flow cytometry, followed by correlation analysis with fertilization rates.</p><p><strong>Results: </strong>NEU1 and NEU3 are independent indicators separate from regular semen parameters. With a metaphase II (MII) rate ≥80% and normal semen routine analysis, sperm NEU1 (P = 0.0300) and NEU3 (P = 0.0253) levels were significantly different demarcated by 80% fertilization rate. Lower NEU1 and NEU3 expression on sperm is associated with lower fertilization rates. ROC curve analyses displaying the diagnostic accuracy of NEU1 (P = 0.0306, AUC = 0.660) and NEU3 (P = 0.0259, AUC = 0.665). χ<sup>2</sup> test analyzes the fertilization rate and the different cut-off rate for NEU1 (χ<sup>2</sup> = 4.671, P = 0.0307) or NEU3 (χ<sup>2</sup> = 6.377, P = 0.0116).</p><p><strong>Conclusions: </strong>NEU1 and NEU3 on sperm can predict the IVF fertilization outcome in the group with oocyte MII rate ≥80%. The protein levels of NEU1 and NEU3 on sperm could be a new index for assisted reproductive technology (ART).</p>","PeriodicalId":15246,"journal":{"name":"Journal of Assisted Reproduction and Genetics","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-08-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144799212","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Min Wang, Lianwen Zheng, Shuai Ma, Ying Xu, Jingshun Zhang, Lulu Fu
{"title":"A comparison of the effects of progestin-primed ovarian stimulation protocol and gonadotropin-releasing hormone antagonist protocol in assisted reproductive technology: a systematic review and meta-analysis.","authors":"Min Wang, Lianwen Zheng, Shuai Ma, Ying Xu, Jingshun Zhang, Lulu Fu","doi":"10.1007/s10815-025-03612-4","DOIUrl":"https://doi.org/10.1007/s10815-025-03612-4","url":null,"abstract":"<p><strong>Objective: </strong>We aimed to compare the effects of the progestin-primed ovarian stimulation (PPOS) protocol and gonadotropin-releasing hormone antagonist (GnRH-A) protocol in women with different ovarian reserves who underwent assisted reproductive technology (ART).</p><p><strong>Methods: </strong>We searched published studies in the Cochrane Library, Web of Science, Embase, PubMed, CNKI, and CBM databases. Patients in the experimental group underwent the PPOS protocol, and those in the control group underwent the GnRH-A protocol. Randomised controlled trials (RCTs) and non-randomised controlled trials (N-RCTs) of PPOS and GnRH-A protocols were collected. We searched the literature published until November 1, 2024. A subgroup analysis was performed for patients with different ovarian reserves.</p><p><strong>Results: </strong>This study included 36 studies. The primary outcome showed that the live birth rate was similar between the PPOS and GnRH-A groups. In the high ovarian response (HOR) patients, the incidence of ovarian hyperstimulation syndrome (OHSS) was significantly lower in the PPOS protocol group than in the GnRH-A group [OR = 0.24, 95% confidence interval (CI) = 0.12-0.48, p < 0.0001]. In the secondary outcomes, the endometrial thickness in the PPOS protocol group decreased compared with the GnRH-A group (mean difference (MD) = - 1.13, 95% CI = - 1.76 to - 0.51, p = 0.0004). In the HOR subgroup, gonadotropin (Gn) dose (MD = 222.88, 95% CI = 59.30-386.46, p = 0.008) and duration (MD = 0.70, 95% CI = 0.48-0.92, p < 0.00001) were increased in the PPOS protocol group compared with the GnRH-A protocol group. In the normal ovarian response (NOR) subgroup, the number of viable embryos in the PPOS protocol group was greater than that in the GnRH-A group (MD = 2.00, 95% CI = 0.10-3.90, p = 0.04).</p><p><strong>Conclusion: </strong>The PPOS protocol had similar clinical effects to the GnRH-A protocol. In HOR patients, the Gn duration and dose in the PPOS protocol group increased, whereas OHSS incidence was reduced. Meanwhile, in NOR patients, the number of viable embryos in the PPOS protocol group increased. The PPOS protocol can be widely promoted in clinical practice when patients do not choose to proceed with fresh embryo transfer due to their own circumstances.</p>","PeriodicalId":15246,"journal":{"name":"Journal of Assisted Reproduction and Genetics","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144799211","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Junfeng Li, Hang Xing, Jing Zhao, Yuan Chen, Yuqing Zhang, Alix Hamon, Rongxiang Li, Shaozhe Yang, Xiuhong Fu
{"title":"Enhancing frozen-thawed embryo transfer outcomes and treatment personalization through machine learning models.","authors":"Junfeng Li, Hang Xing, Jing Zhao, Yuan Chen, Yuqing Zhang, Alix Hamon, Rongxiang Li, Shaozhe Yang, Xiuhong Fu","doi":"10.1007/s10815-025-03598-z","DOIUrl":"https://doi.org/10.1007/s10815-025-03598-z","url":null,"abstract":"<p><strong>Background: </strong>Infertility affects millions globally, with significant social, emotional, and economic consequences. While frozen-thawed embryo transfer (FET) is a cornerstone of assisted reproductive technology, its clinical pregnancy success rates remain inconsistent (29.6-59.2%). Improving predictive accuracy and personalizing treatment strategies for FET outcomes could address critical unmet needs in reproductive medicine.</p><p><strong>Objective: </strong>To develop and validate machine learning models to accurately predict clinical pregnancy outcomes following FET and to simulate personalized treatment strategies based on individual patient profiles.</p><p><strong>Methods: </strong>A retrospective analysis of 1013 FET cycles across two medical centers was conducted. Four machine learning (ML) models-XGBoost, random forest, logistic regression, and deep neural networks-were trained using female-specific features, male-specific features, combined female and male features, and combined features supplemented with expert-selected clinical features. Model performance was evaluated via ROC AUC, sensitivity, and specificity. SHAP analysis identified key predictors, while decision curve analysis assessed clinical utility. Personalized FET strategies were simulated to evaluate the potential for tailored interventions.</p><p><strong>Results: </strong>The XGBoost model trained on combined features supplemented with expert-selected clinical features outperformed all other models, achieving the highest ROC AUC (0.7922) along with balanced sensitivity (0.7309) and specificity (0.7755). SHAP analysis highlighted embryo quality, female age, and anti-Müllerian hormone levels as top predictors. Decision curve analysis confirmed XGBoost's clinical utility, demonstrating optimal net benefit across decision thresholds by balancing true and false positives. Simulated personalized strategies based on model predictions showed potential to refine treatment protocols, enhancing pregnancy success rates through patient-specific adjustments.</p><p><strong>Conclusions: </strong>XGBoost-based ML models provide a robust, data-driven framework for predicting FET outcomes and personalizing treatment. By integrating key clinical and embryological factors, these models enable precision care strategies that optimize success rates and patient outcomes. This study underscores the transformative role of ML in advancing reproductive medicine, offering a pathway to improve decision-making and reduce the burden of infertility globally.</p>","PeriodicalId":15246,"journal":{"name":"Journal of Assisted Reproduction and Genetics","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144768684","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Huanmin Luo, Shuqing Li, Yuming Cao, Jinfeng Xu, Li Wang
{"title":"Identification of m5C-related genes and subclusters in recurrent pregnancy loss.","authors":"Huanmin Luo, Shuqing Li, Yuming Cao, Jinfeng Xu, Li Wang","doi":"10.1007/s10815-025-03580-9","DOIUrl":"https://doi.org/10.1007/s10815-025-03580-9","url":null,"abstract":"<p><strong>Purpose: </strong>Recurrent pregnancy loss (RPL), which occurs in 1-5% of couples and nearly half of the cases remain unexplained, is a complex condition influenced by multiple factors. Previous investigations have demonstrated the role of m5C-related genes (MRGs) in cancer prognosis and the significance of epigenetic modifications during pregnancy. However, the connection between MRGs and the pathogenesis of RPL remains elusive. This study endeavors to elucidate this relationship through bioinformatics approaches.</p><p><strong>Methods: </strong>Data of 48 endometrial tissue samples were obtained through GEO query. Twenty-one MRGs were analyzed. Multiple machine learning (ML) methods were applied to identify biomarkers. A nomogram was constructed, and further analyses like GSEA and scRNA-seq were carried out using the R software.</p><p><strong>Results: </strong>Five core biomarkers (DNMT1, SMUG1, ZBTB38, MBD4, and TDG) were pinpointed by ML methods, with the prediction model achieving an AUC of 0.953. Based on hub genes, 24 RPL samples were grouped into cluster A (n = 9) and cluster B (n = 15). The study revealed differences in immune cells and microenvironments, and the scRNA-seq analysis confirmed the connection between immune cells and m5C.</p><p><strong>Conclusion: </strong>This study identified five key m5C-related genes, unraveled their link to immune cells, and developed an accurate RPL diagnostic model. The RPL patients are innovatively divided into two clusters, and the difference of their immune microenvironment is analyzed. This study offers a fresh perspective for examining biomarkers and potential therapeutic targets for RPL.</p>","PeriodicalId":15246,"journal":{"name":"Journal of Assisted Reproduction and Genetics","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144768685","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Pregnancy and obstetric outcomes of frozen-thawed embryo transfer with embryos generated from COVID‑19 infection during controlled ovarian stimulation: a retrospective study.","authors":"Huayan Yin, Jingtang Zhang, Yuqi Zhou, Jieru Li, Longmei Wu, Qunshan Shen, Yunxia Cao, Bing Song","doi":"10.1007/s10815-025-03608-0","DOIUrl":"https://doi.org/10.1007/s10815-025-03608-0","url":null,"abstract":"<p><strong>Purpose: </strong>This investigation aimed to explore the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during controlled ovarian stimulation (COS) on the pregnancy and obstetric outcomes in frozen-thawed embryo transfer (FET) cycles. This area has not been thoroughly examined in previous research.</p><p><strong>Methods: </strong>This retrospective cohort study analyzed 126 couples with confirmed SARS-CoV-2 infection during COS and evaluated FET outcomes using embryos generated during virologically active exposure windows. Controls were noninfected women during COS, matched by age, number, and day of embryo transfer in the same time. Demographic and cycle characteristics and pregnancy and obstetric outcomes were examined for this retrospective cohort analysis.</p><p><strong>Results: </strong>This cohort study comprised 300 infertile couples, stratified into SARS-CoV-2-exposed (n = 126) and unexposed (n = 174) groups based on infection status during COS. The clinical pregnancy rates were 58.8% and 58.6%, respectively (P = 0.958). Other pregnancy features, including biochemical pregnancy, early miscarriage, ongoing pregnancy, and cumulative clinical pregnancy, were also comparable between the two groups. No significant intergroup differences were observed in obstetric outcomes, such as gestational age, cesarean section rates, and preterm birth rates.</p><p><strong>Conclusions: </strong>In FET cycles of patients with embryos generated from SARS-CoV-2 infection, no significant difference in pregnancy and obstetric outcomes was observed. These data substantiate the reproductive safety of FET following SARS-CoV-2 exposure during COS. Nevertheless, more studies are needed for longitudinal investigations to elucidate the transgenerational effects of SARS-CoV-2 on somatic maturation and neurodevelopmental trajectories in offspring.</p>","PeriodicalId":15246,"journal":{"name":"Journal of Assisted Reproduction and Genetics","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-08-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144768694","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Trish Dinh, Nichole Sanchez Diaz, Kelsey McLaughlin, John C Kingdom, Ellen M Greenblatt, Sascha Drewlo, John W Snelgrove
{"title":"Mid-pregnancy circulating placental growth factor (PlGF) and obstetrical outcomes following nonovulatory and ovulatory frozen embryo transfer cycles.","authors":"Trish Dinh, Nichole Sanchez Diaz, Kelsey McLaughlin, John C Kingdom, Ellen M Greenblatt, Sascha Drewlo, John W Snelgrove","doi":"10.1007/s10815-025-03605-3","DOIUrl":"https://doi.org/10.1007/s10815-025-03605-3","url":null,"abstract":"<p><strong>Purpose: </strong>We compared serum placental growth factor (PlGF) concentration in pregnancies from ovulatory vs. non-ovulatory FET cycles and evaluated associations with obstetrical and neonatal outcomes.</p><p><strong>Methods: </strong>We conducted a secondary analysis of a prospective single-center screening study at a large urban center in Canada where PlGF was performed between April 2020 and December 2022. Baseline characteristics were compared between FET ovulatory cycle and non-ovulatory cycle groups. We compared PlGF concentration in unadjusted and adjusted analyses using multivariable linear regression. Secondary outcomes were compared using chi square and Fisher exact tests for categorical variables and Wilcoxon rank sum tests for continuous variables.</p><p><strong>Results: </strong>We identified 340 patients, 246 (72.4%) in the non-ovulatory group and 94 (27.7%) in the ovulatory group. The median gestational age (IQR) at PlGF testing was 27.9 weeks (26.6-28.3) in the non-ovulatory group and 27.7 weeks (25.6-28.1) in the ovulatory group (p = 0.07). Univariable analysis comparing the difference in PlGF showed no association between groups [β - 51.7 (95% CI - 43.7-384.1), p = 0.44]. After adjusting for maternal age, race, parity, pre-pregnancy BMI, gestational diabetes, gestational age of PlGF test, donor egg cycle, and maternal and previous obstetrical comorbidities, the difference in PlGF concentration remained non-significant. Obstetrical and neonatal outcomes were similar between groups.</p><p><strong>Conclusion: </strong>Our findings indicate no significant difference in either mid-pregnancy circulating PlGF concentration or obstetrical outcomes comparing successful FETs in ovulatory vs. non-ovulatory cycles. Larger-scale prospective research should be prioritized to further elucidate the role of PlGF in predicting the potential risk of adverse pregnancy outcomes.</p>","PeriodicalId":15246,"journal":{"name":"Journal of Assisted Reproduction and Genetics","volume":" ","pages":""},"PeriodicalIF":2.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144764957","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ha T T Nguyen, Tam T M Luu, Linh T Đo, Tri C Nguyen, Diem T N Nguyen, Trang T M Ho, Hoa Giang, Thuy T H Dao, Bao G Huynh, Tuong M Ho, Lan N Vuong
{"title":"Non-invasive preimplantation genetic testing for aneuploidy using cell-free DNA in blastocyst culture medium.","authors":"Ha T T Nguyen, Tam T M Luu, Linh T Đo, Tri C Nguyen, Diem T N Nguyen, Trang T M Ho, Hoa Giang, Thuy T H Dao, Bao G Huynh, Tuong M Ho, Lan N Vuong","doi":"10.1007/s10815-025-03510-9","DOIUrl":"10.1007/s10815-025-03510-9","url":null,"abstract":"<p><strong>Purpose: </strong>This study evaluated the performance of preimplantation genetic testing for aneuploidies (PGT-A) using cell-free DNA (cfDNA) from spent culture media (SCM) of blastocyst embryos (non-invasive PGT-A; NiPGT-A) compared with conventional trophectoderm (TE) biopsy samples.</p><p><strong>Methods: </strong>This prospective study was conducted at IVFMD, My Duc Hospital, Vietnam, from August to December 2020, and included patients with an indication for PGT-A. The culture medium was replaced on day 3, and SCM from day 3 to the day of TE biopsy (days 5 or 6) of all biopsied blastocysts was tested using next-generation sequencing. The total concordance rate, sensitivity, and specificity of NiPGT-A versus PGT-A for detecting aneuploid embryos were calculated. Outcomes after single blastocyst transfer are also reported.</p><p><strong>Results: </strong>Forty-four couples participated; 100 paired TE PGT-A biopsies and SCM samples were evaluated. The whole-genome amplification success rate for SCM was 82%; 77 samples had clear NGS results and were further evaluated. The total concordance rate between NiPGT-A and PGT-A was 63.6%. For detecting aneuploidy, NiPGT-A had a sensitivity of 57.1%, specificity of 67.3%, positive predictive value of 50.0%, and negative predictive value of 73.3%. Of the 35 single euploidy embryo transfers, 8 had no NiPGT-A results, 21 were classified as NiPGT-A euploid, and 6 were classified as NiPGT-A aneuploid; the live birth rate was 51.4% (18/35). Four of the 6 NiPGT-A aneuploid blastocysts resulted in live births.</p><p><strong>Conclusions: </strong>cfDNA in SCM has the potential for NiPGT-A. However, the NiPGT-A process is unreliable enough to replace traditional PGT-A using TE biopsy.</p>","PeriodicalId":15246,"journal":{"name":"Journal of Assisted Reproduction and Genetics","volume":" ","pages":"2587-2595"},"PeriodicalIF":2.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12423013/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144119397","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sarah C Rubin, Kaleb Noruzi, Emily Chen, Alexis Greene, Martin Keltz
{"title":"Intraovarian platelet-rich plasma (PRP) infusion appeared to benefit low prognosis IVF patients; however, when compared to controls, no significant benefit could be confirmed.","authors":"Sarah C Rubin, Kaleb Noruzi, Emily Chen, Alexis Greene, Martin Keltz","doi":"10.1007/s10815-025-03530-5","DOIUrl":"10.1007/s10815-025-03530-5","url":null,"abstract":"<p><strong>Purpose: </strong>To assess whether in-office intraovarian PRP injections among patients with a history of a low euploid embryo yield following freeze-all PGT-A IVF cycle improves transferable embryo yield when compared to controls.</p><p><strong>Materials and methods: </strong>A retrospective case-control study, between March 2022 and December 2024 where all poor outcome patients who underwent in-office intraovarian PRP injection prior to a repeat freeze-all PGT-A IVF cycle were compared to controls with no intervention between cycles. The primary outcome was transferable embryo yield. Secondary outcomes included: blastocyst yield, aneuploidy rate, and euploid yield.</p><p><strong>Results: </strong>Thirty-two patients met the inclusion criteria and were compared to 309 controls. Mean age for the PRP and control group were 38.3 ± 3.1 and 38.4 ± 3.7 (p = 0.91) respectively. Mean AMH for the PRP group was 1.6 ± 1.1 ng/mL as compared to 1.5 ± 1.0 ng/mL (p = 0.34). Following PRP the blastocyst, euploid and transferable embryo yield increased; however, when compared to controls, there were no differences in IVF outcomes. For patients with an AMH ≥ 1, the euploid yield and transferable embryo yield increased by fivefold; however, there was no difference when compared to the control. For patients with an AMH < 1, there were no differences in euploid or transferable embryo yield following PRP.</p><p><strong>Conclusion: </strong>Although there was an improvement in transferable embryos yielded following PRP, no difference was observed compared to the control group, as the control with a poor initial cycle experienced regression to the mean which may also be a source of bias in the PRP group.</p>","PeriodicalId":15246,"journal":{"name":"Journal of Assisted Reproduction and Genetics","volume":" ","pages":"2719-2726"},"PeriodicalIF":2.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12422996/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144199243","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Juncheng Yao, Yuanyuan Zhang, Tingting Lan, Yutao Li
{"title":"Autophagy-related biomarkers in non-obstructive azoospermia: insights from transcriptomics and single-cell sequencing.","authors":"Juncheng Yao, Yuanyuan Zhang, Tingting Lan, Yutao Li","doi":"10.1007/s10815-025-03559-6","DOIUrl":"10.1007/s10815-025-03559-6","url":null,"abstract":"<p><strong>Objective: </strong>Autophagy, by modulating cellular degradation and recycling processes, affects sperm cell survival and differentiation and may be linked to the pathophysiology of non-obstructive azoospermia (NOA). However, the role of autophagy-related genes (ARGs) in NOA has not yet been fully explored.</p><p><strong>Methods: </strong>Transcriptomic datasets for NOA from public databases were used. By combining differential expression analysis between NOA and obstructive azoospermia (OA), weighted gene co-expression network analysis (WGCNA), and machine learning. algorithms, biomarkers were identified. Their performance was evaluated with ROC curves. Enrichment and immune analyses explored mechanisms, and validation was done via datasets (GSE9210 and GSE145467) and RT-qPCR. Single-cell level mechanisms were also studied.</p><p><strong>Results: </strong>A total of 321 differentially expressed genes (DEGs) were screened, related to germ cell development and nucleocytoplasmic transport. Four biomarkers (ATP6V1E2, UBQLN2, FYCO1, and ITPR1) from machine learning had good diagnostic performance. FYCO1 positively correlated with T follicular helper cells and negatively with regulatory T cells. Single-gene GSEA showed it was enriched in gametogenesis and cell adhesion molecules. Single-cell analysis revealed ATP6V1E2 had the highest expression in testicular gamete cells, while UBQLIN2, FYCO1, and ITPR1 in smooth muscle cells RT-qPCR results matched the dataset trends.</p><p><strong>Conclusion: </strong>This study screened four ARGs (ATP6V1E2, UBQLN2, FYCO1, and ITPR1) as biomarkers for NOA, which could serve as potential diagnostic tools and therapeutic targets, potentially providing new treatment strategies.</p>","PeriodicalId":15246,"journal":{"name":"Journal of Assisted Reproduction and Genetics","volume":" ","pages":"2773-2791"},"PeriodicalIF":2.7,"publicationDate":"2025-08-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12423381/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144528147","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}