{"title":"Morphometric analysis of neoplastic cell clusters in high-grade serous ovarian cancer ascites identifies a promising prognostic factor: a retrospective study.","authors":"Benoît Thibault, Romina D'Angelo, Samy Rigal, Mélanie White-Koning, Guillaume Bataillon, Julie Guillermet-Guibert, Céline Basset","doi":"10.1186/s13048-025-01653-y","DOIUrl":"https://doi.org/10.1186/s13048-025-01653-y","url":null,"abstract":"<p><p>High-grade serous carcinoma of the ovary is the most frequent intraperitoneal malignancy in women. It is associated with a poor prognostic outcome owing to the late appearance of clinical signs leading to a delayed diagnosis, and with resistance to platinum-based chemotherapy. One of the clinical signs is the development of ascites. The detection of neoplastic cells in ascites fluid is important as it indicates tumor progression and is associated with shorter survival. Microscopic cytospin analysis of this fluid reveals the cytological and architectural features of the neoplastic cells, allowing the pathologist to identify rapidly the malignancy and the histologic type. In association with immunocytochemistry, this process ensures a definite diagnosis and provides a specific etiology. Our objective was to provide proof-of-principle that the automatized analysis of general cytomorphological criteria, such as carcinomatous cell clustering, in malignant ascites fluid is of prognostic value in high-grade serous carcinoma. We performed a retrospective analysis of the ascites fluid of 24 advanced-stage high-grade serous ovarian cancer patients naïve of treatment. We found that the low number of neoplastic cell clusters in fluid was significantly associated with shorter overall and progression-free survival after adjusting for WHO performance status, Sugarbaker score, age and BMI. These results were independent of the peritoneal implantation of neoplastic cells. We believe this is a promising strategy to improve high-grade serous carcinoma diagnostics using a more informative but simple analysis of ascites tumor cell morphology.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"74"},"PeriodicalIF":3.8,"publicationDate":"2025-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143811639","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}
Jingfang Liu, Beier Wu, Shihan Wan, Yanlu Jin, Li Yang, Meijuan Wu, Jie Xing, Jiejie Zhang, Xin Chen, Aijun Yu
{"title":"Upregulation of TRPS1 promotes proliferation, migration, and invasion in ovarian clear cell carcinoma and correlates with poor patient prognosis.","authors":"Jingfang Liu, Beier Wu, Shihan Wan, Yanlu Jin, Li Yang, Meijuan Wu, Jie Xing, Jiejie Zhang, Xin Chen, Aijun Yu","doi":"10.1186/s13048-025-01603-8","DOIUrl":"10.1186/s13048-025-01603-8","url":null,"abstract":"<p><strong>Objective: </strong>Tricho-rhino-phalangeal syndrome-1 (TRPS1), an atypical GATA transcription factor, plays a critical role in diverse physiological and pathological processes and holds potential as a biomarker for diseases and targeted tumor therapies. This study explores TRPS1 expression in ovarian clear cell carcinoma (OCCC), its correlation with patient prognosis, and its involvement in OCCC pathogenesis.</p><p><strong>Research objectives and methods: </strong>To investigate TRPS1 expression, we analyzed ovarian tissues from 50 OCCC patients and 25 normal tissues (from patients with uterine leiomyoma) via immunohistochemistry. Statistical methods, including Chi-square tests, Kaplan-Meier survival analysis, and Cox regression, were employed to evaluate the correlation between TRPS1 expression and clinicopathological parameters. In OCCC cell lines (TOV21G and ES-2), TRPS1 expression was quantified using qRT-PCR and Western blot. Functional studies were conducted by silencing TRPS1 in TOV21G cells with small interfering RNA and inducing overexpression in ES-2 cells using a plasmid. Cellular proliferation and migration were assessed through CCK-8, colony formation, and Transwell assays. Finally, Western blot analysis was performed to investigate the link between TRPS1 and EMT-related molecular pathways.</p><p><strong>Results: </strong>TRPS1 protein expression was significantly higher in OCCC tissues compared to normal tissues and was positively associated with lymph node metastasis and advanced clinical stage. High TRPS1 expression was linked to shorter overall and recurrence-free survival in OCCC patients. In vitro, TRPS1 knockdown suppressed cell proliferation, migration, and invasion, accompanied by reduced levels of invasion-promoting proteins (N-cadherin, MMP2, MMP9) and increased expression of the invasion-inhibiting protein E-cadherin. Conversely, TRPS1 overexpression promoted the expression of invasion-promoting proteins.</p><p><strong>Conclusions: </strong>TRPS1 is overexpressed in OCCC and is associated with poor prognosis, serving as an independent predictor of patient outcomes. Its elevated expression enhances OCCC cell proliferation, migration, and invasion by regulating proteins involved in the epithelial-to-mesenchymal transition (EMT) pathway. These findings highlight TRPS1 as a critical player in OCCC pathogenesis and a potential biomarker and therapeutic target for disease management.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"73"},"PeriodicalIF":3.8,"publicationDate":"2025-04-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143803646","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}
Megha Nair, Arun Samidurai, Anindita Das, Sham S Kakar, Rakesh C Kukreja
{"title":"Ovarian cancer and the heart: pathophysiology, chemotherapy-induced cardiotoxicity, and new therapeutic strategies.","authors":"Megha Nair, Arun Samidurai, Anindita Das, Sham S Kakar, Rakesh C Kukreja","doi":"10.1186/s13048-025-01636-z","DOIUrl":"10.1186/s13048-025-01636-z","url":null,"abstract":"<p><p>Ovarian Cancer (OC) is recognized as the most lethal gynecologic malignancy, characterized by numerous genetic mutations that trigger uncontrolled cellular growth and replication. Emerging evidence suggests that non-coding RNAs including miRNAs and lncRNAs significantly influence OC through their multiple roles including tumor initiation, progression, metastasis, immune evasion, and chemoresistance, making them promising diagnostic markers and therapeutic targets. The primary approach to treating OC typically involves cytoreductive surgery followed by chemotherapy. However, the chemotherapeutic agents, particularly the anthracyclines such as doxorubicin (DOX), are known for their cardiotoxic effects, which can range from acute to chronic, potentially leading to heart failure and death. To enhance the overall treatment response and to minimize cardiotoxicity, alternative strategies have been explored. These include the use of liposomal doxorubicin (DOXIL) as a substitute for DOX, various radiotherapies, immunotherapies, and the co-administration of angiotensin-converting enzyme inhibitors and/or beta-blockers. Phosphodiesterase-5 inhibitors (PDE5i) have also demonstrated efficacy in reducing cardiotoxicity linked to cancer treatments and in promoting apoptosis in cancer cells across multiple cancer types. Although there is no current clinical trial directly examining the impact of PDE5i on reducing cardiotoxicity in OC, however emerging therapies such as Withaferin A, PARP inhibitors, and nanoparticle combination therapy show promise. Additional research is essential to develop treatments that are both effective against OC and less harmful to the heart.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"72"},"PeriodicalIF":3.8,"publicationDate":"2025-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11971845/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143788538","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}
Donglan Yuan, Ting Guo, Xiaotong Zhu, Weiwei Song, Dengyun Nie, Hong Yu
{"title":"Icariside II induces ferroptosis through the down-regulation of SLC7A11 in ovarian cancer.","authors":"Donglan Yuan, Ting Guo, Xiaotong Zhu, Weiwei Song, Dengyun Nie, Hong Yu","doi":"10.1186/s13048-025-01650-1","DOIUrl":"10.1186/s13048-025-01650-1","url":null,"abstract":"<p><strong>Background: </strong>Ovarian cancer (OV) is the leading cause of death among gynecological malignancies. This study aimed to investigate the influence of Icariside II on OV in vitro and in vivo and to elucidate whether Icariside II induces ferroptosis in OV cells by regulating SLC7A11 expression.</p><p><strong>Methods: </strong>SKOV3 cells and OV nude mice were treated with Icariside II, a control-plasmid or an SLC7A11-plasmid. EdU assay, flow cytometry, wound-healing assay, and Transwell assays were used to assess cell proliferation, apoptosis, migration, and invasion respectively. Total iron, Fe<sup>2+</sup> levels, and intracellular lipid reactive oxygen species (ROS) stimulation were evaluated in both cells and tissues. Levels of cysteine (Cys), glutathione (GSH), and glutathione peroxidase 4 (GPX4) were also analyzed. Ferroptosis markers, including Ptgs2, Chac1, SLC7A11, and apoptosis-associated genes (Bax and Bcl-2), were detected using qRT-PCR, western blotting, and immunohistochemistry (IHC). SLC7A11 expression in OV was explored using data from The Cancer Genome Atlas (TCGA), and validated with IHC staining.</p><p><strong>Results: </strong>Icariside II-induced ferroptosis in OV cells was confirmed by elevated Fe<sup>2+</sup> and total iron levels, enhanced lipid ROS levels, higher Ptgs2 and Chac1 mRNA levels, and reduced levels of SLC7A11, Cys, GSH, and GPX4 in both in vitro and in vivo models. These effects were partially reversed by the SLC7A11-plasmid. Moreover, Icariside II suppressed SKOV3 cell proliferation, inhibited cells migration and invasion, and promoted apoptosis by downregulating SLC7A11 expression. Furthermore, we found that SLC7A11 expression was upregulated in OV tissues compared to adjacent non-tumor tissues.</p><p><strong>Conclusion: </strong>Icariside II induces ferroptosis in OV by downregulating SLC7A11 expression in vitro and in vivo. Our study identified Icariside II as a promising therapeutic agent for the treatment of OV.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"71"},"PeriodicalIF":3.8,"publicationDate":"2025-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11971851/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143788482","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}
Suqin Zhu, Zhiqing Huang, Xiaojing Chen, Wenwen Jiang, Yuan Zhou, Beihong Zheng, Yan Sun
{"title":"Construction and evaluation of machine learning-based prediction model for live birth following fresh embryo transfer in IVF/ICSI patients with polycystic ovary syndrome.","authors":"Suqin Zhu, Zhiqing Huang, Xiaojing Chen, Wenwen Jiang, Yuan Zhou, Beihong Zheng, Yan Sun","doi":"10.1186/s13048-025-01654-x","DOIUrl":"10.1186/s13048-025-01654-x","url":null,"abstract":"<p><strong>Objective: </strong>To investigate the determinants affecting live birth outcomes in fresh embryo transfer among polycystic ovary syndrome (PCOS) patients using various machine learning (ML) algorithms and to construct predictive models, offering novel insights for enhancing live birth rates in this specific group.</p><p><strong>Methods: </strong>A sum of 1,062 fresh embryo transfer cycles involving PCOS patients were analyzed, with 466 resulting in live births. The dataset was split randomly into training and testing subsets at a 7:3 ratio. Least absolute shrinkage and selection operator and recursive feature elimination methods were utilized for feature selection within the training data. A grid search strategy identified the optimal parameters for seven ML models: decision tree (DT), K-nearest neighbors (KNN), light gradient boosting machine (LightGBM), naive Bayes model(NBM), random forest (RF), support vector machine (SVM) and extreme gradient boosting (XGBoost). The evaluation of model effectiveness incorporated diverse metrics, encompassing area under the curve (AUC), accuracy, positive predictive value, negative predictive value, F1 score, and Brier score. Calibration curves and decision curve analysis were employed to ascertain the optimal model. Furthermore, Shapley additive explanations were applied to elucidate the importance of predictor variables in the top-performing model.</p><p><strong>Results: </strong>The AUC values of DT, KNN, LightGBM, NBM, RF, SVM and XGBoost models in the training set were 0.813, 1.000, 0.724, 0.791, 1.000, 0.819 and 0.853, respectively. Corresponding values in the testing set were 0.773, 0.719, 0.705, 0.764, 0.794, 0.806 and 0.822. XGBoost emerged as the most effective ML model. SHAP analysis revealed that variables encompassing embryo transfer count, embryo type, maternal age, infertility duration, body mass index, serum testosterone (T) levels, and progesterone (P) levels on the day of human chorionic gonadotropin administration were pivotal predictors of live birth outcomes in individuals with PCOS receiving fresh embryo transfer.</p><p><strong>Conclusion: </strong>This study developed a live birth prediction model tailored for PCOS fresh embryo transfer cycles, leveraging ML algorithms to compare the efficacy of multiple models. The XGBoost model demonstrated superior predictive capacity, enabling prompt and precise identification of critical risk factors influencing live birth outcomes in PCOS patients. These findings offer actionable insights for clinical intervention, guiding strategies to improve pregnancy outcomes in this population.</p><p><strong>Clinical trial number: </strong>Not applicable.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"70"},"PeriodicalIF":3.8,"publicationDate":"2025-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11969817/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143788309","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}
Xiaopei Li, Yan Li, Bumei Zhang, Jianmei Wang, Yang Yang, Yongrui Du
{"title":"Plasma cytokine levels and PCOS risk: Mendelian randomization analysis reveals IL6R as a preventive factor.","authors":"Xiaopei Li, Yan Li, Bumei Zhang, Jianmei Wang, Yang Yang, Yongrui Du","doi":"10.1186/s13048-025-01647-w","DOIUrl":"10.1186/s13048-025-01647-w","url":null,"abstract":"<p><strong>Background: </strong>Polycystic ovary syndrome is a prevalent gynecological condition affecting primarily women of childbearing age. It is characterized by elevated androgen levels, ovulatory dysfunction, and morphological abnormalities. Despite extensive research from various perspectives, the etiology and pathogenesis of PCOS remain unclear. While controversial, many believe that individuals with PCOS exhibit a chronic low-grade inflammatory state. Cytokines play diverse roles in the initiation and progression of inflammation, contributing to this inflammatory milieu. Therefore, the aim of this study was to utilize publicly available genome-wide association study data to explore the potential causal relationship between cytokines and PCOS.</p><p><strong>Methods: </strong>To accurately investigate the causal relationship between cytokines and PCOS, we initially defined cytokines using the GeneCrad and then identified cytokines in two independent large-scale plasma proteins. Subsequently, we employed a two-sample Mendelian randomization analysis framework. A series of quality control procedures were implemented to select eligible instrumental variables closely associated with the exposure. MR analysis was conducted using genome-wide association studies of PCOS in two independent European ancestry groups. Cochran, s Q test, MR-Egger and intercept test were employed to assess heterogeneity and pleiotropy in PCOS. Co-localization analysis, summary-data-based Mendelian randomization analysis, and HEIDI testing were utilized to further corroborate the relationship between positive findings and PCOS. Finally, systematical Mendelian randomization analysis between healthy lifestyle factors and PCOS-related proteins was conducted to identify which proteins could act as interventional targets by lifestyle changes.</p><p><strong>Results: </strong>In our investigation, we performed Mendelian randomization analysis on 33 cytokines in relation to PCOS using data from the deCODE and the Fenland. Our findings revealed that the plasma level of IL6R emerges as a notable protective factor against PCOS, exhibiting a substantial effect size. Moreover, we identified CCL22 as a significant risk factor for PCOS, a finding that was similarly validated and supported by independent cohorts.</p><p><strong>Conclusion: </strong>Our Mendelian randomization analysis, leveraging genome-wide association study data from a sizable population cohort, unequivocally delineated a causal relationship between IL6R and PCOS. These results underscore the involvement of cytokines in the pathogenesis of PCOS and highlight their potential as promising therapeutic targets for addressing this intricate disease.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"69"},"PeriodicalIF":3.8,"publicationDate":"2025-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11966818/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143780277","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}
{"title":"Modified screening of MYC promotor region elements using the CRISPR library in ovarian cancer.","authors":"Akiko Yamamoto, Yosuke Tanaka, Sachiko Ishibashi, Masumi Ikeda, Keisuke Sugita, Masanori Ono, Hirotaka Nishi, Morito Kurata","doi":"10.1186/s13048-025-01644-z","DOIUrl":"10.1186/s13048-025-01644-z","url":null,"abstract":"<p><p>Ovarian cancer remains one of the most lethal gynecological malignancies owing to its high recurrence rate and chemotherapeutic resistance. MYC is a well-known proto-oncogene that is frequently amplified in ovarian cancer and has been implicated in drug resistance. Previously, we established a new promoter-reporter system combined with a CRISPR activation library to identify unknown MYC regulators, and M1AP was identified as a novel MYC regulator. However, considering the insufficient explanation for the absence of guide RNA (gRNA) of MYC, this present study explored methods to prevent the gRNA of MYC itself from binding. This study first modified the promoter-reporter vector to improve its quality, then conducted CRISPR screening and analyzed candidate genes as MYC promoter regulators using next-generation sequencing in OVSAHO ovarian cancer cells. Eighty-six genes had ≥ 1000 reads, and Pearson's correlation coefficient analysis was performed on the cBioPortal of the Cancer Genomics database. Fourteen genes were identified as candidate MYC regulators with positive and significant correlations with MYC. Seven genes, including CYP4v2, ASPH, ANP32D, PCED1A, ABI1, FUZ, and HOOK2, demonstrated significantly higher luciferase activity than the control genes. Four genes, including ABI1, PCED1A, HOOK2, and CYP4v2, activated the MYC promoter, which showed over twofold higher activity than the control when overexpressed using a vector. In conclusion, four genes that activate MYC promoters were identified in an ovarian cancer cell line using the CRISPR library system with a modified promoter-reporter tool. These results will prove helpful in the development of novel treatment strategies for ovarian cancer.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"68"},"PeriodicalIF":3.8,"publicationDate":"2025-04-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11963443/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143772716","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}
Zhaoping Tan, Tiancheng Wu, Mei Wang, Liang Chen, Yating Li, Ming Zhang, Yuanzhen Zhang, Lili Sun
{"title":"Downregulation of FASN in granulosa cells and its impact on ovulatory dysfunction in PCOS.","authors":"Zhaoping Tan, Tiancheng Wu, Mei Wang, Liang Chen, Yating Li, Ming Zhang, Yuanzhen Zhang, Lili Sun","doi":"10.1186/s13048-025-01645-y","DOIUrl":"10.1186/s13048-025-01645-y","url":null,"abstract":"<p><strong>Background: </strong>Polycystic ovary syndrome (PCOS) is a complicated endocrinological and anovulatory disorder in women. Mice exposed to dihydrotestosterone (DHT) exhibit a PCOS-like phenotype characterized by abnormal steroid hormone production and ovulation dysfunction. The present investigation aims to identify overlapping genes expressed in PCOS patients and a PCOS mouse model induced by DHT and to examine the function of key genes fatty acid synthase (FASN) in hormone production and ovulation dysfunction.</p><p><strong>Results: </strong>We examined 5 datasets of high-throughput mRNA transcription from the Gene Expression Omnibus (GEO) database, including 4 datasets from individuals with PCOS and 1 dataset from a DHT-induced mouse model. GO and KEGG enrichment analyses revealed these differentially expressed genes (DEGs) are primarily involved in ovarian steroidogenesis and fatty acid metabolism. The PPI network identified 12 hub genes. qRT-PCR verification in human granulosa cells showed differential expression of FASN, SCARB1, FABP5, RIMS2, and RAPGEF4 in PCOS patients (p < 0.05). FASN was downregulated in the granulosa cells (GCs) of PCOS patients (p < 0.05). FASN depletion reduced KGN cell proliferation (p < 0.001), decreased progesterone secretion (p < 0.05), and increased estradiol secretion (p < 0.05). Downregulation of FASN inhibited ovulation by suppressing ERK1/2 phosphorylation and the expression of C/EBPα and C/EBPβ. Lentivirus-mediated FASN downregulation in rat ovaries for one and four weeks impaired the super ovulatory response, reducing oocyte retrieval, estrous cycle, secretion of estrogen and progesterone, and luteinization.</p><p><strong>Conclusions: </strong>Our results provide new insights into PCOS pathogenesis and suggest that FASN could be a promising target for treating abnormal steroid hormone production and impaired ovulation in PCOS.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"67"},"PeriodicalIF":3.8,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11959749/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143764195","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}
Fereshteh Esfandiarinezhad, Xiaoshu Zhan, Seang Lin Tan, Julang Li, Benjamin K Tsang
{"title":"A primary insight into gut microbiome, MicroRNA and stemness, in a PCOS rat model.","authors":"Fereshteh Esfandiarinezhad, Xiaoshu Zhan, Seang Lin Tan, Julang Li, Benjamin K Tsang","doi":"10.1186/s13048-025-01648-9","DOIUrl":"10.1186/s13048-025-01648-9","url":null,"abstract":"<p><p>Polycystic ovary syndrome (PCOS) is a common endocrine disorder associated with reproductive and metabolic dysfunctions, including gut microbiome dysbiosis. This study aimed to examine the alterations in stemness in ovarian surface epithelium (OSE), gut microbiome microRNA expression in granulosa cells and plasma in a dihydrotestosterone (DHT)-induced rat model of PCOS. Female rats were administered DHT to induce PCOS, and the expression of stem cell markers in OSE was assessed to evaluate the impact on stemness. Alterations in the gut microbiome composition were assessed using 16S rRNA gene Long-Read sequencing and changes in the microRNA profile of granulosa cells and plasma were analyzed using qPCR. Our results demonstrated alterations in stemness markers and, a significant alteration in gut microbiome composition in DHT-induced rats compared to controls, characterized by shifts in the relative abundance of specific bacterial taxa, particularly Akkermansia muciniphila. Elevated levels of miR-574 and miR-378 were observed in plasma, whereas miR-21 and miR-574 showed increased expression in ovarian granulosa cells. Concurrently, increased expression of stem cell markers was observed in OSE, suggesting an enhancement of stemness in response to PCOS-like conditions. These findings imply a potential link between gut microbiome dysbiosis and increased ovarian stemness in PCOS, suggesting that the gut microbiome may contribute to ovarian dysfunction through modulation of stem cell activity. Understanding this interaction could provide novel insights into therapeutic targets in restoring ovarian function in PCOS patients.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"66"},"PeriodicalIF":3.8,"publicationDate":"2025-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11959994/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143764194","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}
Hongcui Zhao, Juan Wu, Yang Xu, Xiaofang Shen, Huanhuan Wang, Aihua Zhao, Fumin Cao, Xinna Chen
{"title":"Intra-ovarian platelet-rich plasma administration plus successive accumulated embryo transfer could be a promising strategy for poor ovarian response management: a before-after study.","authors":"Hongcui Zhao, Juan Wu, Yang Xu, Xiaofang Shen, Huanhuan Wang, Aihua Zhao, Fumin Cao, Xinna Chen","doi":"10.1186/s13048-025-01646-x","DOIUrl":"https://doi.org/10.1186/s13048-025-01646-x","url":null,"abstract":"<p><strong>Background: </strong>The management of poor responders is a significant challenge for both patients and clinicians. The aim of this study was to evaluate the effectiveness of intra-ovarian injection of Platelet-Rich Plasma (PRP) combined with successive accumulated embryo transfer in improving the outcomes of patients with Poor Ovarian Response(POR) based on POSEIDON criteria.</p><p><strong>Methods: </strong>This single-center, retrospective before-after study was conducted at a private reproductive center, involving 49 women diagnosed with POR, indicated by an AMH level of less than 1.2 ng/ml. The participants, comprising 13 group 3 and 36 group 4 POR patients, underwent intra-ovarian injections of PRP followed by the accumulation of embryos over three successive cycles of mild stimulation IVF/ICSI from May 2021 to May 2022, before proceeding to the embryo transfer phase. The ovarian reserve markers, oocyte and embryologic outcomes were compared in all patients before and after intra-ovarian injection of PRP. The cumulative clinical pregnancy and cumulative live birth outcomes were presented. Statistical analyses were performed using SPSS version 25. A p-value < 0.05 denoted statistical significance.</p><p><strong>Result(s): </strong>The mean age of all participants was 37.67 ± 4.15 years and their mean body mass index was 21.52 ± 2.80 kg/m<sup>2</sup>. Autologous intraovarian PRP therapy significantly increased AMH levels, AFC and decreased FSH levels. Autologous intraovarian PRP therapy accompanied with 3 successive cumulated cycles, significantly increased No. of accumulated embryos and blastocysts. This strategy also significantly reduced the rate of cancelled cycle. Following this strategy, of 44 cases with accumulated embryos/blastocysts transfer, 20 (45.45%) achieved clinical pregnancy, of which 15 (34.09%) resulted in live births and 5 (11.36%) ended in miscarriage.</p><p><strong>Conclusion(s): </strong>Intra-ovarian injection of PRP plus successive embryo accumulation following mild stimulation and accumulated embryo transfer appears to be an optimal strategy for POR management.</p><p><strong>Clinical trial number: </strong>Not applicable.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"64"},"PeriodicalIF":3.8,"publicationDate":"2025-03-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11951750/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143743039","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}