Geertruid J Brink, Jolijn W Groeneweg, Ariane A Sickinghe, Hans W Nijman, Luc R C W van Lonkhuijzen, Christianne A R Lok, Jurgen M J Piek, Eva Maria Roes, Cornelis D de Kroon, Ward Hofhuis, Geertruida N Jonges, Eelke H Gort, Petronella O Witteveen, Ronald P Zweemer
{"title":"Is it time to abandon staging surgery and prolonged follow-up in patients with primary adult-type granulosa cell tumor?","authors":"Geertruid J Brink, Jolijn W Groeneweg, Ariane A Sickinghe, Hans W Nijman, Luc R C W van Lonkhuijzen, Christianne A R Lok, Jurgen M J Piek, Eva Maria Roes, Cornelis D de Kroon, Ward Hofhuis, Geertruida N Jonges, Eelke H Gort, Petronella O Witteveen, Ronald P Zweemer","doi":"10.1186/s13048-025-01622-5","DOIUrl":"10.1186/s13048-025-01622-5","url":null,"abstract":"<p><strong>Background: </strong>As current literature does not provide sufficient data to support clear guidelines in patients with a rare adult-type granulosa cell tumor, we aim to investigate: (1) whether additional staging surgery following primary surgical treatment is necessary; (2) how long standard follow-up should be and (3) risk factors for disease recurrence.</p><p><strong>Methods: </strong>A national multicenter prospective study was initiated in April 2018. Patients with suspected or confirmed adult-type granulosa cell tumor were eligible. Data on staging, follow-up and risk factors were both retrospectively and prospectively collected from medical records, and patients were followed until April 2024 or until death. Descriptive statistical analysis and survival analysis were performed using Cox regression methods and Kaplan-Meier analyses.</p><p><strong>Results: </strong>In total, 208 patients with histopathologically confirmed adult-type granulosa cell tumor were included, with a median follow-up of 5.5 years (IQR: 2.2-12.3 years). Vaginal bleeding and abdominal pain were the most common symptoms at diagnosis. Median time until first recurrence was 4.2 years (range 2 months- 32 years). Additional staging surgery did not reduce the risk of recurrence. During follow-up, most patients had no symptoms at the time of detection of recurrence. No difference in overall survival was found between patients who were diagnosed with a recurrence during follow-up, and those who were no longer in follow-up and presented with symptoms.</p><p><strong>Conclusions: </strong>Staging surgery does not improve recurrence free survival in patients with adult-type granulosa cell tumor. Our results suggest that adult-type granulosa cell tumor patients can be discharged from follow-up of adult-type granulosa cell tumor after five years.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"37"},"PeriodicalIF":3.8,"publicationDate":"2025-02-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11846436/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143476608","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":"TAM-derived exosomal miR-589-3p accelerates ovarian cancer progression through BCL2L13.","authors":"Jianqing Wang, Yan Zhu, Yang He, Weiwei Shao","doi":"10.1186/s13048-025-01618-1","DOIUrl":"10.1186/s13048-025-01618-1","url":null,"abstract":"<p><strong>Background: </strong>Tumor-associated macrophages (TAM) are critical elements of intercellular communication in tumor microenvironment (TME), and exosomes are key mediators between tumor cells and the TME. According to previous reports, miRNAs exert a pivotal role in ovarian cancer (OC) development. The purpose of this work was to explore the function of TAM-derived exosomal miR-589-3p in OC development and elucidate the underlying molecular mechanisms.</p><p><strong>Methods: </strong>First, peripheral blood mononuclear cells (PBMC) were treated with IL-4 and IL-13 to polarize them into M2-type macrophages. Exosomes were separated from M2-type macrophages, and the physical properties of exosomes were evaluated using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). Next, quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) was applied to examine the expression of relevant genes. Subsequently, Targetscan and miRDB were utilized to predict miR-589-3p target genes, and then the interaction between miR-589-3p and BCL2L13 was verified by dual luciferase assay and RNA Binding Protein Immunoprecipitation (RIP) assay. Finally, Cell Counting Kit-8 (CCK-8) and flow cytometry experiments were employed to explore the changes in the proliferative and apoptotic abilities of OC cells.</p><p><strong>Results: </strong>In this research, we demonstrated that TAM-derived exosomes facilitated OC cell proliferation and suppressed OC cell apoptosis. Then, qRT-PCR results indicated that miR-589-3p were markedly elevated after co-culture of TAM-derived exosomes with OC cells. In addition, we discovered that miR-589-3p was bound to BCL-2-like protein 13 (BCL2L13), which was confirmed through luciferase assay and RIP assay. Furthermore, functional analysis displayed that TAM-derived exosomes treated with miR-589-3p inhibitor attenuated the promotion of OC cell progression by exosomes.</p><p><strong>Conclusion: </strong>TAM-derived exosomal miR-589-3p enhanced OC progression through BCL2L13, which offers a novel for OC therapy.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"36"},"PeriodicalIF":3.8,"publicationDate":"2025-02-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11846191/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143476610","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":"Physiopathology of polycystic ovary syndrome in endocrinology, metabolism and inflammation.","authors":"Pingping Su, Chao Chen, Yun Sun","doi":"10.1186/s13048-025-01621-6","DOIUrl":"10.1186/s13048-025-01621-6","url":null,"abstract":"<p><p>Polycystic ovary syndrome (PCOS) is a prevalent endocrine disorder characterized by elevated androgen levels, ovarian cysts, and impaired ovulation in females. This condition is closely linked with various reproductive health issues and has significant impacts on endocrine and metabolic pathways. Patients with PCOS commonly exhibit hyperandrogenaemia and insulin resistance, leading to complications such as acne, hirsutism, weight fluctuations, and metabolic disturbances, as well as an increased risk for type 2 diabetes, cardiovascular disease, and endometrial cancer. Although extensive research has identified several mechanistic aspects of PCOS, a thorough understanding of its pathophysiology remains incomplete. This review aims to provide a detailed analysis of the physiological and pathological aspects of PCOS, covering endocrine, metabolic, and inflammatory dimensions, to better elucidate its etiological framework.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"34"},"PeriodicalIF":3.8,"publicationDate":"2025-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11841159/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143468513","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":"Administration of dehydroepiandrosterone improves endometrial thickness in women undergoing IVF/ICSI: a systematic review and meta-analysis.","authors":"Ling Huang, Ying Gao, Shuo Liang, Mei Jiang","doi":"10.1186/s13048-024-01581-3","DOIUrl":"10.1186/s13048-024-01581-3","url":null,"abstract":"<p><strong>Background: </strong>The positive effects of dehydroepiandrosterone (DHEA) on oocyte and embryo quality improvement are often concerned. While the results on DHEA-induced endometrial improvement are controversial.</p><p><strong>Objective: </strong>To evaluate whether DHEA intervention improved endometrial function and reproductive outcomes during in vitro fertilization/intracytoplasmic sperm injection (IVF/ICSI) cycles and to thus provide clinical recommendations.</p><p><strong>Data sources: </strong>PubMed, Cochrane Library, EMBASE and Web of Science from database inception to 31 July 2024, without language restrictions. The references of conference proceedings and websites on clinical trials were manually checked.</p><p><strong>Study design: </strong>Systematic review and meta-analysis.</p><p><strong>Study eligibility criteria: </strong>Parallel-controlled randomized controlled trials (RCTs) design; women underwent IVF/ICSI, patients in the experimental group received adminstration with DHEA, whereas the control group received with or without placebo; and the outcomes included reproductive or endometrial function.</p><p><strong>Study appraisal and synthesis methods: </strong>RCTs evaluating the effects of DHEA on IVF/ICSI outcomes were included. Risk of bias and quality of evidence (QoE) were assessed according to the Cochrane Collaboration's tool and the Grading of Recommendations Assessment, Development and Evaluation system. Odds ratios (ORs) and mean differences (MDs) with 95% confidence intervals (CIs) were assessed by random-effects or fixed-effects models. Subgroup and meta-regression analyses were used to find sources of heterogeneity. Trial sequential analysis was used to judge the stability of the outcomes. Trial sequential analysis was used in order to control for random errors.</p><p><strong>Results: </strong>A total of 16 trials included 1973 women. DHEA treatment significantly increased endometrial thickness (MD = 0.93, CI: 0.27 to 1.60; low QoE), which helped improve clinical pregnancy rate (CPR) (OR = 1.34, 95% CI: 1.08 to 1.67; low QoE). DHEA administration also increased the quality of oocyte and embryo [including the number of oocytes retrieved (MD = 0.73, CI: 0.36 to 1.10; low QoE), oocytes fertilized (MD = 0.48, CI: 0.10 to 0.87; low QoE), transferred embryos (MD = 0.27, CI: 0.09 to 0.46; very low QoE), and high-quality embryos (MD = 0.65, CI: 0.27 to 1.03; low QoE)]. Subgroup and meta-regression analyses revealed that heterogeneity might be related to disease type, ovarian stimulation protocol, and addition time of DHEA treatment. There was insufficient evidence to reach a conclusion regarding the live birth rate/ongoing pregnancy rate, miscarriage rate, and MII oocyte number of DHEA. And no severe adverse effects were observed with DHEA administration. Due to the apparent improvemen in the CPR, women with thin endometrium might benefit from DHEA cotreament.</p><p><strong>Conclusions: </strong>Due to the limited","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"35"},"PeriodicalIF":3.8,"publicationDate":"2025-02-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11843996/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143468506","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":"Polycystic ovary syndrome and epithelial-mesenchymal transition: Mendelian randomization and single-cell analysis insights.","authors":"Dong Liu, Dan Liu, Kunyan Zhou","doi":"10.1186/s13048-025-01617-2","DOIUrl":"10.1186/s13048-025-01617-2","url":null,"abstract":"<p><strong>Background: </strong>The process of epithelial-mesenchymal transition (EMT) may promote fibrosis in ovarian tissue related to polycystic ovary syndrome (PCOS), thus affecting ovarian function and hormonal balance.</p><p><strong>Objective: </strong>This study aimed to explore key genes associated with EMT in PCOS and their potential molecular regulatory mechanisms, exclusively from the perspective of transcriptomics and single-cell RNA sequencing (scRNA-seq), combined with Mendelian Randomization (MR) analysis.</p><p><strong>Methods: </strong>The dataset for PCOS and EMT-related genes (EMT-RGs) were sourced from public databases. The key genes in this study were identified via differential expression analysis, MR, and evaluation of expression levels. Enrichment analysis and a series of functional analyses were conducted on these genes to further elucidate their potential mechanisms. Subsequently, using scRNA-seq data and validation of the expression of key genes, key cell group in PCOS were identified, followed by pseudo-time and cell communication analyses to provide deeper insights.</p><p><strong>Results: </strong>Three key genes, NUCB2 [odds ratio (OR) = 0.8634, 95% confidence interval (CI): 0.8145-0.9152, P < 0.0001], PGF (OR = 0.8393, 95% CI: 0.7185-0.9805, P < 0.05), and CRIM1 (OR = 0.7539, 95% CI: 0.6556-0.670, P < 0.0001), were identified as having a unidirectional causal association with PCOS and were associated with a reduced risk of PCOS. In public datasets, NUCB2 exhibited significantly increased expression in PCOS samples, while PGF and CRIM1 showed the opposite trends. These three genes were enriched in pathways related to cellular functions, metabolic processes, and the operation of the nervous system, and they were co-expressed in smooth muscle. Additionally, five cell clusters were annotated, among which fibroblasts were identified as key cells due to their highest expression of all three key genes. Further analysis revealed a bifurcation event occurring during the mid-development stage of fibroblasts, with PCOS samples displaying a higher abundance of fibroblasts. In PCOS samples, fibroblasts exhibited more extensive communication with secretory epithelial cells, indicating a more complex intercellular interaction within this condition.</p><p><strong>Conclusion: </strong>This study identified three EMT-RGs: NUCB2, PGF, and CRIM1, which were associated with a reduced risk of PCOS, with fibroblast identified as a key cell group in the disease's pathology. This provides new insights for PCOS research.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"33"},"PeriodicalIF":3.8,"publicationDate":"2025-02-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11841333/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143458454","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}
Sicong Liu, Hao Lin, Ke Zhang, Quan Zhou, Yang Shen
{"title":"Potential drug targets for ovarian cancer identified through Mendelian randomization and colocalization analysis.","authors":"Sicong Liu, Hao Lin, Ke Zhang, Quan Zhou, Yang Shen","doi":"10.1186/s13048-025-01620-7","DOIUrl":"10.1186/s13048-025-01620-7","url":null,"abstract":"<p><strong>Background: </strong>The existing drugs for ovarian cancer (OC) are unsatisfactory and thus new drug targets are urgently required. We conducted proteome-wide Mendelian randomization (MR) and colocalization analysis to pinpoint potential targets for OC.</p><p><strong>Methods: </strong>Data on protein quantitative trait loci (pQTL) for 734 plasma proteins were obtained from large genome-proteome-wide association studies. Genetic associations with OC were derived from the Ovarian Cancer Association Consortium, which included 25,509 cases and 40,941 controls. MR analysis was performed to evaluate the association between the proteins and the OC risk. Colocalization analysis was conducted to check whether the identified proteins and OC shared causal variants. In addition, the phenome-wide MR analysis was performed to clarify protein associations across the phenotype, and drug target databases were examined for target validation.</p><p><strong>Results: </strong>Genetically predicted circulating levels of 44 proteins were associated with OC risk at Benjamini-Hochberg correction. Genetically predicted 17 proteins had evidence of the increased risk of OC (CLEC11A, MFAP2, TYMP, PDIA3, IL1R1, SPINK1, PLAU, DKK2, IL6ST, DLK1, LRRC15, CDON, ANGPTL1, SEMA4D, AKR1A1, TNFAIP6, and FCGR2B); 27 proteins decreased the risk of OC(SIGLEC9, RARRES1, SPINT3, TMEM132A, HAVCR2, CNTN2, TGFBI, GSTA1, HGFAC, TREML2, GRAMD1C, ASAH2, CPNE1, CCL25, MAPKAPK2, POFUT1, PREP, NTNG1, CA10, CACNA2D3, CA8, MAN1C1, MRC2, IL10RB, RBP4, GP5 and CALCOCO2). Bayesian colocalization demonstrated that GRAMD1C, RBP4, PLAU, PDIA3, MFAP2, POFUT1, MAN1C1 and DKK2 shared the same variant with OC. The phe-MR analyses assessed the side effects of these 44 identified proteins, and the drug target database offered information on both approved and investigational indications.</p><p><strong>Conclusion: </strong>This study provides proof of a causal relationship between genetically predicted 44 proteins associated with OC risk, which could serve as promising drug targets for OC.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"32"},"PeriodicalIF":3.8,"publicationDate":"2025-02-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11837690/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143458479","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}
Zhiqi Liao, Yueping Zhou, Weili Tao, Lin Shen, Kun Qian, Hanwang Zhang
{"title":"Correlation between the follicular fluid extracellular-vesicle-derived microRNAs and signaling disturbance in the oocytes of women with polycystic ovary syndrome.","authors":"Zhiqi Liao, Yueping Zhou, Weili Tao, Lin Shen, Kun Qian, Hanwang Zhang","doi":"10.1186/s13048-025-01619-0","DOIUrl":"10.1186/s13048-025-01619-0","url":null,"abstract":"<p><p>Polycystic ovary syndrome (PCOS) is a common reproductive disorder characterized by hyperandrogenism, ovulatory dysfunction, and polycystic ovaries. The quality of oocytes in PCOS patients remains poor, leading to poor pregnancy outcomes. The molecular mechanisms underlying the poor quality of oocytes in PCOS are not fully understood. This study aimed to explore the potential functional microRNAs (miRNAs) in follicular fluid (FF)-derived extracellular vesicles (FF-EVs) and their role in oocyte developmental competence in PCOS. We analyzed DEmiRNAs in FF-EVs and DEGs in oocytes from PCOS patients and controls using GEO database. We identified 14 potential functional DEmiRNAs in FF-EVs and predicted the target genes of 14 DEmiRNAs using TargetScan. We performed conjoint analyses between the target genes of these miRNAs and DEGs in oocytes, identifying 12 DEmiRNAs whose target genes overlap with oocyte DEGs. Thus, 12 functional DEmiRNAs were the hub miRNAs. These miRNAs were predicted to target genes involved in oocyte development and signaling pathways such as PI3K/Akt, Ras, and MAPK pathways. KEGG enrichment analysis suggested that these miRNAs might impair oocyte developmental competence in PCOS by dysregulating PI3K/Akt signaling pathway. qRT-PCR validated the increase of miR-93-3p and miR-152-3p, and the decrease of miR-625-5p and miR-17-5p in FF-EVs of PCOS patients. This study highlighted the significance of FF-EVs in the pathology of PCOS and revealed the potential role of the increase of miR-93-3p and miR-152-3p, and the decrease of miR-625-5p and miR-17-5p in impairing oocyte developmental competence in PCOS. Further research is needed to elucidate the specific mechanisms by which these miRNAs affect oocyte development and to explore the potential therapeutic implications.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"31"},"PeriodicalIF":3.8,"publicationDate":"2025-02-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11837631/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143449417","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":"The impact of preoperative immunonutritional status on prognosis in ovarian cancer: a multicenter real-world study.","authors":"Xingyu Liu, Ming Li, Yingjun Zhao, Xiaofei Jiao, Yang Yu, Ruyuan Li, Shaoqing Zeng, Jianhua Chi, Guanchen Ma, Yabing Huo, Zikun Peng, Jiahao Liu, Qi Zhou, Dongling Zou, Li Wang, Qingshui Li, Jing Wang, Shuzhong Yao, Youguo Chen, Ding Ma, Ting Hu, Qinglei Gao","doi":"10.1186/s13048-025-01607-4","DOIUrl":"10.1186/s13048-025-01607-4","url":null,"abstract":"<p><strong>Background: </strong>To investigate the effect of preoperative immunonutritional status on prognosis in epithelial ovarian cancer patients.</p><p><strong>Methods: </strong>A multicenter real-world study included 922 patients with histologically confirmed epithelial ovarian cancer who received comprehensive staged surgery or debulking surgery at seven tertiary hospitals in China between 2012 and 2023. Prognostic nutritional index (PNI) and systemic immune-inflammation index (SII) were used to assess the immunonutritional status for their superior predictive power to indicate the nutritional status and the inflammatory immunity. Cox regression analyses were employed to identify variables associated with progression-free survival (PFS) and overall survival (OS).</p><p><strong>Results: </strong>In the early-stage cohort of 224 epithelial ovarian cancer patients, the optimal cut-off value for PNI was 47.47 for both PFS and OS, while the optimal cut-off value for SII values were 551.37 for PFS and 771.78 for OS. In the late-stage group of 698 patients, the optimal PNI thresholds were 47.76 for PFS and 46.00 for OS, with SII values of 720.96 for PFS and 1686.11 for OS. In multivariate analysis of early-stage patients, high PNI was an independent protective factor for PFS (hazard ratio (HR), 0.39 (95% confidence interval (CI) 0.20-0.76), P = 0.006) and OS (HR, 0.44 (95% CI 0.20-0.97), P = 0.042), respectively. High SII was significantly associated with PFS (HR, 2.43 (95% CI 1.23-4.81), P = 0.011) and marginally unfavorable for OS (HR, 2.05 (95% CI 0.96-4.39), P = 0.064). In advanced population, PNI (HR, 0.77 (95% CI 0.60-0.99), P = 0.043) and SII (HR, 1.34 (95% CI 1.01-1.78), P = 0.041) were independent prognostic factors for OS but had no impact on PFS (P = 0.185, P = 0.188, respectively).</p><p><strong>Conclusion: </strong>Poor preoperative immunonutritional status has a deleterious effect on the prognosis of patients with ovarian cancer. Intervention in patients suffering from suboptimal preoperative immunonutritional status may facilitate improved survival outcomes.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"30"},"PeriodicalIF":3.8,"publicationDate":"2025-02-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11831797/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143441135","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}
Edyta Biskup, Joanna Lopacinska-Jørgensen, Claus Høgdall, Estrid V Høgdall
{"title":"Potential use of DNA methylation in cervical swabs for early ovarian cancer diagnosis.","authors":"Edyta Biskup, Joanna Lopacinska-Jørgensen, Claus Høgdall, Estrid V Høgdall","doi":"10.1186/s13048-025-01609-2","DOIUrl":"10.1186/s13048-025-01609-2","url":null,"abstract":"<p><strong>Introduction: </strong>Early diagnosis of ovarian cancer, using cost-effective and non-invasive methods remains an unmet medical need, largely due to unspecific symptoms of the disease.</p><p><strong>Objective: </strong>Our goal was to identify differentially methylated CpG loci between cervical swabs obtained from patients diagnosed with benign ovarian disease and with malignant pelvic mass.</p><p><strong>Methodology: </strong>Using Infinium EPICv2 array, we interrogated methylation profiles of 77 cervical swabs. The study cohort was then divided into a training and testing set to develop a diagnostic signature. We applied several strategies to pinpoint CpG sites able to differentiate cervical swabs obtained from ovarian cancer patients and patients with benign ovarian disease.</p><p><strong>Results and conclusions: </strong>None of the statistical methods applied identified CpG loci capable of diagnosing ovarian cancer with sufficient specificity and sensitivity. We conclude that methylation differences observed do not adequately distinguish between benign and malignant ovarian disease. The variations attributable to clinical conditions are likely obscured by the differences in cell composition, which is the primary source of sample heterogeneity. Therefore, we suggest that diagnostic tools should not rely on local methylation profile of the cervix but rather focus on detecting cancer-specific sequences transferred from the tumor site and present in cervical swabs. Ovarian cancer is difficult to detect early, and we aimed to explore whether DNA methylation in cervical swabs could serve as a diagnostic tool. However, our study found that methylation patterns in these samples do not reliably distinguish between benign and malignant conditions, likely due to variations in cell composition. We recommend future research focus on detecting tumor-specific DNA sequences in cervical swabs instead.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"29"},"PeriodicalIF":3.8,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11830180/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143425628","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":"Tumour-derived exosomal miR-205 promotes ovarian cancer cell progression through M2 macrophage polarization via the PI3K/Akt/mTOR pathway.","authors":"Liuqing He, Quan Chen, Xiaoying Wu","doi":"10.1186/s13048-025-01616-3","DOIUrl":"10.1186/s13048-025-01616-3","url":null,"abstract":"<p><strong>Background: </strong>Tumour-associated macrophages (TAMs) are the most abundant immune cells in the tumour environment and are considered similar to M2 macrophages, which facilitate cancer progression. Exosomes, as important mediators of the cross-talk between tumour cells and tumour-associated macrophages, can facilitate the development and metastasis of ovarian cancer by mediating M2 macrophage polarization. However, the exact mechanisms underlying the communication between ovarian cancer (OC) cells and tumour-associated macrophages during OC progression remain unclear.</p><p><strong>Results: </strong>Here, we demonstrated that high expression of miR-205 was associated with M2 macrophage infiltration, which affected the prognosis of OC patients. Importantly, tumour-derived miR-205 could be transported from OC cells to macrophages via exosomes and promote cancer cell invasion and metastasis by inducing M2-like macrophage polarization. Animal experiments further confirmed that exosomal miR-205-induced M2 macrophages accelerated OC progression in vivo. Mechanistically, miR-205 downregulated PTEN, activating the PI3K/AKT/mTOR signalling pathway, which is critical for M2 polarization.</p><p><strong>Conclusions: </strong>These results reveal that exosomal miR-205 plays a pivotal role in macrophage polarization within the OC microenvironment, highlighting its potential as a therapeutic target for OC treatment. This study not only enhances our understanding of the interactions between tumour and immune cells but also opens new avenues for targeted therapies against exosomal miR-205 in ovarian cancer.</p>","PeriodicalId":16610,"journal":{"name":"Journal of Ovarian Research","volume":"18 1","pages":"28"},"PeriodicalIF":3.8,"publicationDate":"2025-02-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11829414/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143425630","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}