1
|
Kenealy BP, Lochner JE. Cancer Screening in Women. Prim Care 2025; 52:233-248. [PMID: 40412903 DOI: 10.1016/j.pop.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Cancer is the second leading cause of death in US women. Lung cancer has the highest mortality rate in the United States, followed by breast cancer, colorectal cancer, and pancreatic cancer. The incidence of new cancer cases per year in the United States is highest for breast cancer, followed by lung cancer, colorectal cancer, and uterine cancer. Cancer incidence and mortality rates are higher for many under-resourced communities. Cancers with effective screening programs include lung, breast, colorectal and cervical cancers. Clinicians should offer screening for these cancers to all women who meet criteria.
Collapse
Affiliation(s)
- Brian P Kenealy
- Department of Family Medicine and Community Health, University of Wisconsin- Madison, 610 North Whitney Way Suite 200, Madison, WI 53705, USA.
| | - Jennifer E Lochner
- Department of Family Medicine and Community Health, University of Wisconsin- Madison, 610 North Whitney Way Suite 200, Madison, WI 53705, USA
| |
Collapse
|
2
|
Yin J, Ma Y, Fu H, Fan Y, Xiang D, Ding L, Huang J. Spartin Promotes Smurf1-Mediated Ubiquitination Modification of YWHAZ to Inhibit Cisplatin Resistance in Ovarian Cancer. FASEB J 2025; 39:e70658. [PMID: 40386996 DOI: 10.1096/fj.202401164r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 04/18/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
Cisplatin (DDP) remains the commonly used chemotherapeutic drug for ovarian cancer (OV); however, DDP resistance poses a great challenge to the outcomes of patients. This work investigated the biological function and mechanism of Spartin in DDP resistance of OV. The growth and apoptosis of DDP-resistant OV cells were assessed by CCK-8, colony formation, and flow cytometry, respectively. Autolysosome fusion was observed by immunofluorescent staining of LC3 and LAMP2. The interaction between E3 ligase Smurf1 and YWHAZ or Spartin protein, and the ubiquitination level of YWHAZ were determined by Co-IP assay. Expression levels of autophagy or apoptosis-related markers were measured by RT-qPCR, western blotting, and immunohistochemistry. DDP resistance was assessed by xenograft tumor experiments in vivo. We found that Spartin expression was lower, while YWHAZ expression was higher in DDP-resistant OV samples and cells. Lower expression of Spartin indicated a poorer survival rate of OV patients. In addition, overexpression of Spartin sensitized OV cells to DDP and repressed autophagy. Moreover, Spartin bound to Smurf1 to promote Smurf1-mediated ubiquitination and degradation of YWHAZ, restraining autophagy and DDP resistance. Overexpression of YWHAZ counteracted the effects of Spartin against DDP resistance by promoting autophagy. In conclusion, Spartin-induced Smurf1-mediated ubiquitination modification of YWHAZ to inactivate autophagy, thereby increasing the sensitivity of OV cells to DDP. Our findings suggest that Spartin-combined therapy might act as an effective approach to fight against DDP resistance in OV.
Collapse
Affiliation(s)
- Jun Yin
- Department of Pharmaceutics, The Affiliated Shunde Hospital of Jinan University, Foshan, Guangdong Province, P.R. China
- Department of Gynecology, Affiliated Tumor Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, P.R. China
- Department of Pharmaceutics, Chongqing University Jiangjin Hospital, Chongqing, P.R. China
| | - Yan Ma
- Department of Gynecology, Affiliated Tumor Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, P.R. China
| | - Hong Fu
- Department of Pediatrics, Chongqing University Jiangjin Hospital, Chongqing, P.R. China
| | - Ying Fan
- Department of Pharmaceutics, Chongqing University Jiangjin Hospital, Chongqing, P.R. China
| | - Debing Xiang
- Oncology, Chongqing University Jiangjin Hospital, Chongqing, P.R. China
| | - Ling Ding
- Department of Pharmaceutics, Chongqing University Jiangjin Hospital, Chongqing, P.R. China
| | - Jing Huang
- Department of Pediatrics, Chongqing University Jiangjin Hospital, Chongqing, P.R. China
| |
Collapse
|
3
|
Sharma A, Hanisha, Rana A, Sharma I. Molecular Crosstalk by miR-449a and miR-34b in endometrial and ovarian cancer cells in vitro. Gene 2025; 947:149337. [PMID: 39965746 DOI: 10.1016/j.gene.2025.149337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/08/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
AIMS Endometrial and ovarian cancers, the sixth and eighth most prevalent cancers in women globally, account for nearly 8% of all new female cancer cases annually. MicroRNAs (miRNAs) have emerged as a promising field in cancer treatment, offering new avenues for targeted therapies and diagnostic tools. Recent miRNA-based cancer research has uncovered various miRNAs commonly dysregulated in cancer and which possess tumor-suppressive functions. These miRNAs influence genes crucial for cellular differentiation, proliferation, apoptosis, and metabolism. MAIN METHODS In the present study, the researchers investigated the effect of dysregulation of two such miRNAs, miR-449a and miR-34b, on the oncogenes involved in the progression of endometrial and ovarian cancer using the respective RL95-2 and SKOV3 cell lines. The transcriptional gene expression analysis was done by Real-Time Quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR). KEY FINDINGS It was found that the overexpression of miR-449a and miR-34b downregulated HIF-1α, VEGF, c-Myc, COX-2, and TNF-α while upregulating TP53 in both cancer types. Conversely, inhibiting these miRNAs increased the levels of HIF-1α, VEGF, c-Myc, COX-2, and TNF-α, and decreased TP53. However, co-transfection with both mimic and inhibitor had varying effects. SIGNIFICANCE The study demonstrated that these miRNAs could influence critical processes such as angiogenesis, proliferation, inflammation, tumorigenesis, and apoptosis in cancer cells, highlighting their potential as therapeutic targets. However, the varied effects observed with the co-transfection of mimics and inhibitors suggest a complex interplay that requires further investigation.
Collapse
Affiliation(s)
- Anuradha Sharma
- Department of Zoology, Panjab University, Chandigarh 160014, INDIA.
| | - Hanisha
- Department of Zoology, Panjab University, Chandigarh 160014, INDIA.
| | - Akanksha Rana
- Department of Zoology, Panjab University, Chandigarh 160014, INDIA.
| | - Indu Sharma
- Department of Zoology, Panjab University, Chandigarh 160014, INDIA.
| |
Collapse
|
4
|
Zhou M, Pi J, Zhao Y. Integrative Multi-Omics Analysis Reveals Molecular Subtypes of Ovarian Cancer and Constructs Prognostic Models. J Immunother 2025:00002371-990000000-00136. [PMID: 40200832 DOI: 10.1097/cji.0000000000000557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/18/2025] [Indexed: 04/10/2025]
Abstract
SUMMARY Ovarian cancer (OV) remains the most lethal gynecological malignancy. The aim of this study was to identify molecular subtypes of OV through integrative multi-omics analysis and construct machine learning-based prognostic models for predicting the efficacy of immunotherapy. In here, the mutation, copy number variation, RNA sequencing expression profiles, and clinical information were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Multi-omics data were stratified using the MOVICS package, identifying different molecular subtypes. Our analysis identified 2 molecular subtypes (CS1 and CS2) with significant survival differences. Transcriptional regulatory network analysis revealed differential activation of transcription factors such as FOXA1 and GATA3 in CS1, whereas AR and ESR2 were enriched in CS2. A robust prognostic signature comprising 5 key genes was developed through the integration of 10 machine learning algorithms, demonstrating high predictive power across data sets. Immune cell infiltration analysis revealed that anti-tumor immune cells were more abundant in low-risk groups, whereas pro-tumor immune cells predominated in high-risk groups. Furthermore, low-risk patients exhibited better immunotherapy responses and higher tumor mutational burden (TMB). In conclusion, our findings underscore the potential of multi-omics integration in unveiling novel OV subtypes and constructing predictive models that inform personalized treatment strategies. Future research should focus on validating these findings in larger cohorts to enhance OV management through targeted therapeutic approaches.
Collapse
Affiliation(s)
- Min Zhou
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | | | | |
Collapse
|
5
|
Lan H, Liu H, Hou H, Zhang C, Zhu J, Zhou N, Zhang X. Combination of anlotinib with immunotherapy enhanced both anti-angiogenesis and immune response in high-grade serous ovarian cancer. Front Immunol 2025; 16:1539616. [PMID: 40260248 PMCID: PMC12009696 DOI: 10.3389/fimmu.2025.1539616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Abstract
Background High-grade serous ovarian cancer (HGSOC) poses significant treatment challenges due to frequent recurrence and resistance to conventional therapies. Combination of anlotinib with immunotherapy have showed promise in various cancers, but its impact on HGSOC remains to be fully elucidated. Methods A retrospective analysis was performed on 36 HGSOC patients treated with anlotinib-based therapies, including both monotherapy and combination treatment with anti-PD-L1/anti-PD-1 antibody (aPD-L1/aPD-1). Peripheral blood mononuclear cell-derived patient-derived xenograft (PBMC-PDX) model was established from drug-resistant recurrent HGSOC patient-derived tumor cells, and single-cell RNA sequencing (scRNA-seq) was conducted to dissect the TME following treatment with anlotinib, anlotinib + aPD-L1 and anlotinib + aPD-1. Results Clinical analysis revealed a disease control rate (DCR) of 71.43% for anlotinib monotherapy, which improved to 100% when combined with aPD-L1/aPD-1. In PBMC-PDX models, treatment evaluation showed that anlotinib decreased tumor volume, an effect further enhanced by its combination with aPD-L1. scRNA-seq analysis demonstrated that anlotinib reduced the proportions of myofibroblastic cancer-associated fibroblasts and ESM1+ endothelial cells, resulting in decreased angiogenesis. The combination of anlotinib and aPD-L1 further amplified these effects, promoting CD8+ T cell infiltration and reversing T cell exhaustion, whereas anlotinib + aPD-1 showed limited efficacy in this regard. Additionally, anlotinib + immunotherapy induced a shift toward M1 polarization of myeloid cells, enhanced anti-tumor activity, and inhibited immune escape. Cell-cell communication analysis revealed reduced APP-CD74 signaling and increased CD99-CD99 signaling, which might contribute to immune activation. Conclusion The combination of anlotinib and aPD-L1 effectively modulates the HGSOC tumor microenvironment by inhibiting angiogenesis, enhancing immune infiltration, and reversing T cell exhaustion.
Collapse
Affiliation(s)
- Hongwei Lan
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Hui Liu
- Department of Clinical Laboratory, Qingdao Women’s and Children’s Hospital, Qingdao, Shandong, China
| | - Helei Hou
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chuantao Zhang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jingjuan Zhu
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Na Zhou
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaochun Zhang
- Precision Medicine Center of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
6
|
Huang X, Li X, Shan W, Dou Y, Yu Q, Chen Y, Wang Z, Zhang H, Wang Y, Lu X, Peng W, Xia B. Integrating Single-Cell and Bulk RNA Sequencing Data to Explore Sphingolipid Metabolism Molecular Signatures in Ovarian Cancer Prognosis: an Original Study. Int J Med Sci 2025; 22:1958-1977. [PMID: 40225866 PMCID: PMC11983299 DOI: 10.7150/ijms.107391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/05/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Ovarian cancer (OC) is the deadliest malignant tumor in the female reproductive system. Sphingolipid metabolism (SM) is crucial for cellular function and has been linked to OC progression. Dysregulation of sphingolipid pathways contributes to tumor growth, chemoresistance, and metastasis in OC. Currently, investigations into the relationship between sphingolipid-related genes (SRGs) and OC prognosis in their initial stages. Our study aimed to develop a novel molecular subtyping based on SRGs and construct a signature to predict the prognosis of patients with OC, immune cell infiltration characteristics, and chemotherapy sensitivity. Methods: Bulk and single-cell RNA-sequencing data of OC was analyzed primarily from the TCGA and GEO databases. The gene set related to the sphingolipid pathway (hsa00600) was selected from the SM pathway, and the enrichment of SRGs was analyzed in the annotated single-cell sequencing data. The Scanpy function was used to score the gene features of each cell and further identify differentially expressed genes. By intersecting with the genes most closely related to SM activity identified through Weighted Gene Co-expression Network Analysis (WGCNA) based on bulk RNA sequencing data, and after performing univariate COX, multivariate COX and LASSO regression, three SRGs were identified. Subsequently, the SRGs-related prognostic signature was constructed. The analysis was further extended to clinical feature correlation, GSEA, tumor microenvironment (TME) analysis and chemotherapy sensitivity analysis. Finally, the expression and function of the key gene GBP5 in the model were validated through in vitro experiments. Results: Compared to other sites, SRG scores were highest in ascites, and among different cell types, SRG scores were highest in T cells. By integrating scRNA-seq and bulk RNA-seq analysis, three SRGs (C5AR1, GBP5, and MARCHF3) were ultimately selected to develop a prognostic model for SRGs. In this model, patients with higher risk scores had shorter overall survival, which was validated in the testing cohort. Immune infiltration analysis revealed that the risk score was negatively correlated with the abundance of CD8+ T cell infiltration and positively correlated with the abundance of M2 macrophage infiltration. Chemotherapy sensitivity analysis showed that the high-risk group exhibited increased resistance to Oxaliplatin, Gemcitabine, and Sorafenib. In vitro, we demonstrated that knockdown of the protective gene GBP5 in HEYA8 and SKOV3 cells enhanced cell viability, proliferation, and invasiveness, reduced apoptosis, and increased IC50 values for chemotherapy drugs. Conclusion: Our model effectively identifies high-risk patients and provides a reference for prognosis prediction using SRG signature. Moreover, hub gene GBP5 acts as a tumor inhibitory factor and regulates the chemosensitivity of oxaliplatin, gemcitabine, and sorafenib in OC.
Collapse
Affiliation(s)
- Xu Huang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Xiaoyu Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Wulin Shan
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
| | - Yingyu Dou
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Qiongli Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Yao Chen
- Bengbu Medical University, Bengbu, Anhui, 233000, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Zengying Wang
- Bengbu Medical University, Bengbu, Anhui, 233000, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Haomin Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Yumeng Wang
- Bengbu Medical University, Bengbu, Anhui, 233000, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Xiaofei Lu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Wenju Peng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| | - Bairong Xia
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230031, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui, 230031, China
| |
Collapse
|
7
|
Li H, Fan T, Qin M, Chen S, Zhou W, Wu P, Yuan Y, Tang X, Zeng T, Fang J, Yi T, Zhang J, Zou J, Li Y. ESM1 suppresses LncRNA GAS5/miR-23a-3p/PTEN axis to promote the cisplatin-chemotherapy resistance of ovarian cancer cells via activating the PI3K/AKT pathway. Discov Oncol 2025; 16:327. [PMID: 40089962 PMCID: PMC11911279 DOI: 10.1007/s12672-025-02113-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/10/2025] [Indexed: 03/18/2025] Open
Abstract
BACKGROUND Cisplatin chemotherapy is an important treatment for advanced ovarian cancer (OC). However, the development of cisplatin resistance greatly limits the survival time of OC patients. Endothelial cell-specific molecule 1 (ESM1) has been found to be an important proto-oncogene promoting OC, but its mediating OC cisplatin resistance remains unknown. METHODS We used quantitative polymerase chain reaction (qPCR) to measure transcription levels of ESM1, Growth arrest specific transcript 5 (GAS5), miR-23a-3p, and Phosphatase And Tensin Homolog (PTEN). A double luciferase reporter gene assay confirmed the direct binding of GAS5 to miR-23a-3p and miR-23a-3p to PTEN mRNA. The effects of ESM1, GAS5, miR-23a-3p, and PTEN on OC cisplatin resistance were tested with an Half Maximal Inhibitory Concentration (IC50) assay. Flow cytometry was used to assess the effects of ESM1, GAS5, and miR-23a-3p on cisplatin-induced OC apoptosis. Changes in apoptosis-related proteins and PI3K/AKT-related proteins were analyzed with western blot (WB). RESULTS ESM1 inhibits the levels of GAS5 and PTEN but increases miR-23a-3p. ESM1 and miR-23a-3p promote OC cisplatin resistance. GAS5 and miR-23a-3p promote cisplatin sensitivity for OC cells. Moreover, the main molecular mechanism is the ESM1/GAS5/miR-23a-3p/PTEN/PI3K/Akt signaling axis. CONCLUSION ESM1 promotes OC cisplatin resistance by activating the Phosphoinositide-3-Kinase (PI3K)/AKT Serine/Threonine Kinase (Akt) signaling pathway through the GAS5/miR-23a-3p/PTEN signaling axis. This suggests that prescriptive ESM1 regulates key downstream molecular mechanisms via non-coding RNA and can be used before neoadjuvant chemotherapy in OC is initiated.
Collapse
Affiliation(s)
- Hui Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Tingyu Fan
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Mei Qin
- Department of Gynecology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Shuanghua Chen
- Hunan Traditional Chinese Medicine College, Zhuzhou, Hunan, China
- Department of Infectious Disease, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Wenchao Zhou
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Peiting Wu
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Yuwei Yuan
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Tian Zeng
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China
| | - Jiawen Fang
- Nursing Class of 2022, School of Nursing, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Ting Yi
- Trauma Center, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, Hunan, China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China.
| | - Juan Zou
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China.
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Hengyang Medical School, Cancer Research Institute, University of South China, Hengyang, Hunan, China.
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China.
| |
Collapse
|
8
|
Harvanik P, Šemeláková M, Solárová Z, Solár P. Novel factors of cisplatin resistance in epithelial ovarian tumours. Adv Med Sci 2025; 70:94-102. [PMID: 39880191 DOI: 10.1016/j.advms.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/11/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025]
Abstract
Ovarian tumours are these days one of the biggest oncogynecological problems. In addition to surgery, the treatment of ovarian cancer includes also chemotherapy in which platinum preparations are one of the most used chemotherapeutic drugs. The principle of antineoplastic effects of cisplatin (cis-diamminedichloroplatinum(II), CDDP) is its binding to the DNA and the formation of adducts. While DNA adducts induce the process of apoptosis, or inhibit the process of DNA replication, which prevents further division of tumour cells, various molecular mechanisms can reverse this process. On the other hand, with increasing scientific knowledge, it is becoming clearer that chemotherapy resistance is a very complex process. In this regard, factors and the amount of their expression may regulate the effect of resistance to chemotherapy. This review focuses on new molecular mechanisms and factors such as mitochondrial dynamics, epithelial-mesenchymal transition (EMT), cluster of differentiation, exosomes and others, that could be involved in the emergence of CDDP resistance.
Collapse
Affiliation(s)
- Pavol Harvanik
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Martina Šemeláková
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Zuzana Solárová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic
| | - Peter Solár
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, Košice, Slovak Republic.
| |
Collapse
|
9
|
Chattaraj A, Mishra V, Mishra Y. Carbon Nanotubes in the Diagnosis and Treatment of Ovarian Cancer. Indian J Microbiol 2025; 65:538-553. [PMID: 40371046 PMCID: PMC12069779 DOI: 10.1007/s12088-024-01367-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/28/2024] [Indexed: 05/16/2025] Open
Abstract
One of the most serious gynecological diseases in the world is ovarian cancer (OC). These days, the majority of patients are identified at an advanced stage (III or IV), with subpar diagnosis resulting in a return of the illness. Conventional medicines fail as a result of issues with early illness identification and treatment processing, including issues with dosage delivery, side effects, and treatment resistance. The carbon nanotube (CNT)-based drug delivery systems for specific OC therapy are highlighted in this review. These systems have several advantages against free drugs, including nontoxicity, biological compatibility, high biodegradability, increased therapeutic impact, and non-inflammatory effects. Crucially, functionalized CNTs with particular ligands like cancer antigen (CA125), Human epididymis protein 4 (HE4), Mucin 1, and folic acid (FA) allow for selective targeting of OC and ultimately increase therapeutic potential in comparison to their nonfunctionalized counterparts. This review focused on the potential applications of CNTs in the detection and treatment of OC, as well as their present status and future clinical developments. Graphical Abstract
Collapse
Affiliation(s)
- Aditi Chattaraj
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411 India
| | - Yachana Mishra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411 India
| |
Collapse
|
10
|
Ren F, Pang X, Jin F, Luan N, Guo H, Zhu L. Integration of scRNA-seq and bulk RNA-seq to reveal the association and potential molecular mechanisms of metabolic reprogramming regulated by lactylation and chemotherapy resistance in ovarian cancer. Front Immunol 2025; 16:1513806. [PMID: 40093000 PMCID: PMC11907005 DOI: 10.3389/fimmu.2025.1513806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Objective Ovarian cancer (OC) ranks among the foremost causes of mortality in gynecological malignancies, with chemoresistance being the primary factor contributing to unfavorable prognosis. This work seeks to clarify the mechanisms of resistance-related lactylation in OC, intending to offer novel theoretical foundations and therapy strategies for addressing chemoresistance. Methods Through the combined analysis of bulk RNA-seq and single-cell RNA-seq data, we initially found lactylation genes linked to chemoresistance. Subsequently, we employed differential expression analysis, survival analysis, enrichment analysis, and other methodologies to further investigate the roles and molecular mechanisms of these genes in tumor resistance. Ultimately, we investigated the differential expression of these genes in resistant and non-resistant tissues and cells via experimentation. Results We found two candidate genes associated with lactylation chemoresistance, ALDH1A1 and S100A4. Analysis of single-cell data indicated that tumor cells represent the primary cell subpopulation relevant to resistance studies. Subpopulation analysis indicated that several tumor cell subtypes were markedly linked to resistance, with elevated expression levels of ALDH1A1 and S100A4 in the resistant subpopulation, notably correlating with various immunological and metabolic pathways. Analysis of metabolic pathways indicated that oxidative phosphorylation and glycolysis activity was elevated in the resistant subpopulation, and lactic acid buildup was associated with chemoresistance. The investigation of the marker gene protein-protein interaction network in the resistant subgroup elucidated the intricate interactions among these genes. The expression levels of ALDH1A1 and S100A4 in the OC tissues of the platinum-resistant cohort were markedly elevated compared to the sensitive cohort, with a considerable rise in S100A4 expression observed in resistant OC cells, demonstrating co-localization with lactylation. Conclusion This work elucidates the significant function of lactylation in OC chemoresistance and identifies ALDH1A1 and S100A4 as possible genes associated with drug resistance. These findings enhance our comprehension of the mechanisms behind chemoresistance in OC and offer critical insights for the formulation of novel therapeutic options.
Collapse
Affiliation(s)
| | | | | | | | | | - Liancheng Zhu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Trétarre B, Satgé D. Ovarian Cancer in Women with Intellectual Disability: Current Data. Cancers (Basel) 2025; 17:805. [PMID: 40075653 PMCID: PMC11898487 DOI: 10.3390/cancers17050805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/14/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
OBJECTIVE We evaluate ovarian cancer (OC) in women with intellectual disability (ID). METHODS We reviewed the literature and added personal observations. The literature search included data from epidemiological studies on cancer incidence and mortality, institutional experiences, and case reports. We also used data from the Hérault Tumor Registry (HTR) in southern France. RESULTS A total of 72 articles met the inclusion criteria, which included 41 cases of OC. The review yielded 29 (74%) germ cell tumors, mainly in girls and young women, and only 4 (10%) ovarian carcinomas, all in adult women. In contrast, the HTR contained six cases of OC and one borderline tumor in adult women with ID aged > 45 years, but no cancer in children and adolescents with ID. These OC cases in adults were discovered at an advanced stage. We found that symptoms revealing OC in women with ID do not differ from those in the general population. However, diagnosis is more complicated in women with ID because they do not communicate easily and may express pain and unease in an unusual way, often through behavioral changes. CONCLUSION OC could be as frequent in women with ID as in the general population and discovered at a late stage. The literature review indicates that girls and adolescents with ID develop mainly germ cell OC, and few carcinomas have been reported in women with ID. In contrast, the HTR was similar to the general population, with carcinomas in women with ID and no OC in children with ID.
Collapse
Affiliation(s)
- Brigitte Trétarre
- Registre des Tumeurs de l’Hérault, 298 Rue des Apothicaires, 34090 Montpellier, France
- Oncodéfi, Parc Euromédecine, 209 Avenue des Apothicaires, 34090 Montpellier, France;
- Centre d’Epidémiologie et de Recherche en Santé des Populations INSERM U1295, Toulouse III University, 31000 Toulouse, France
| | - Daniel Satgé
- Oncodéfi, Parc Euromédecine, 209 Avenue des Apothicaires, 34090 Montpellier, France;
- UMR 1302 Institut Desbrest d’Epidémiologie et de Santé Publique INSERM, Université de Montpellier, 340093 Montpellier, France
| |
Collapse
|
12
|
Kuang M, Liu Y, Chen H, Chen G, Gao T, You K. Big data analysis and machine learning of the role of cuproptosis-related long non-coding RNAs (CuLncs) in the prognosis and immune landscape of ovarian cancer. Front Immunol 2025; 16:1555782. [PMID: 40070821 PMCID: PMC11893572 DOI: 10.3389/fimmu.2025.1555782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Background Ovarian cancer (OC) is a severe malignant tumor with a significant threat to women's health, characterized by a high mortality rate and poor prognosis despite conventional treatments such as cytoreductive surgery and platinum-based chemotherapy. Cuproptosis, a novel form of cell death triggered by copper ion accumulation, has shown potential in cancer therapy, particularly through the involvement of CuLncs. This study aims to identify risk signatures associated with CuLncs in OC, construct a prognostic model, and explore potential therapeutic drugs and the impact of CuLncs on OC cell behavior. Methods We analyzed ovarian cancer data (TCGA-OV) from the TCGA database, including transcriptomic and clinical data from 376 patients. Using Pearson correlation and LASSO regression, we identified 8 prognostic CuLncs to construct a risk signature model. Patients were categorized into high- and low-risk groups based on their risk scores. We performed survival analysis, model validation, drug sensitivity analysis, and in vitro experiments to assess the model's performance and the functional impact of key CuLncs on OC cell proliferation, invasion, and migration. Results The prognostic model demonstrated significant predictive power, with an area under the curve (AUC) of 0.702 for 1-year, 0.640 for 3-year, and 0.618 for 5-year survival, outperforming clinical pathological features such as stage and grade. High-risk OC patients exhibited higher Tumor Immune Dysfunction and Exclusion (TIDE) scores, indicating stronger immune evasion ability. Drugs such as JQ12, PD-0325901, and sorafenib showed reduced IC50 values in the high-risk group, suggesting potential therapeutic benefits. In vitro experiments revealed that knockdown of LINC01956, a key CuLnc in the risk signature, significantly inhibited the proliferation, invasion, and migration of OC cells (P<0.05). Conclusion Our study identified a prognostic risk model based on CuLncs and explored their potential as therapeutic targets in OC. The findings highlight the importance of CuLncs in OC prognosis and immune response, providing new insights for future research and clinical applications.
Collapse
Affiliation(s)
- Mingqin Kuang
- Gynecology and Oncology Department of Ganzhou Cancer Hospital, Ganzhou, Jiangxi, China
| | - Yueyang Liu
- Department of Gynecology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hongxi Chen
- Department of Gynecology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Guandi Chen
- Department of Gynecology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Tian Gao
- Department of Gynecology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Keli You
- Department of Gynecology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
13
|
Wu H, Zhang J, Wang Q, Li Z, Li L, Xie Y. Metformin combined with CB-839 specifically inhibits KRAS-mutant ovarian cancer. Sci Rep 2025; 15:6072. [PMID: 39972191 PMCID: PMC11840008 DOI: 10.1038/s41598-025-90963-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/17/2025] [Indexed: 02/21/2025] Open
Abstract
KRAS mutations can cause metabolic reprogramming in ovarian cancer, leading to an increased metastatic capacity. This study investigated the metabolic reprogramming changes induced by KRAS mutations in ovarian cancer and the mechanism of action of metformin combined with a glutaminase 1 inhibitor (CB-839). KRAS-mutant ovarian cancer accounted for 14% of ovarian cancers. The expression of glucose metabolism-related (PFKFB3, HK2, GLUT1, and PDK2) and glutamine metabolism-related enzymes (GLS1 and ASCT2) was elevated in KRAS-mutant ovarian cancer cells compared with that in wild-type cells. KRAS-mutant cells had a higher aerobic oxidative capacity than did wild-type cells. Metformin inhibited proliferation, the expression of glucose metabolism-related enzymes, and the aerobic oxidative capacity of KRAS-mutant cells compared with those of control cells. Furthermore, it enhanced the expression of glutamine metabolism-related enzymes in KRAS-mutant cells. Metformin combined with CB-839 inhibited the proliferation and aerobic oxidation of KRAS-mutant cells to a greater extent than that observed in wild-type cells. Additionally, the inhibitory effects of metformin and CB-839 in the KRAS-mutant ovarian cancer NOD-SCID mouse model were significantly stronger than those in the drug-alone group. KRAS mutations lead to enhanced glucose and glutamine metabolism in ovarian cancer cells, which was inhibited by metformin combined with CB-839.
Collapse
Affiliation(s)
- Han Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jialin Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qiujie Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zijiao Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Linlin Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ya Xie
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
14
|
Alsaab HO, Almutairy B, Almobarki AO, Mughaedh MAA, Alzahrani MS. Exosome's role in ovarian disease pathogenesis and therapy; Focus on ovarian cancer and failure. J Reprod Immunol 2025; 167:104403. [PMID: 39662240 DOI: 10.1016/j.jri.2024.104403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/16/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024]
Abstract
In the eukaryotic system, exosomes are categorized as unique extracellular vesicles with dimensions ranging from 30 to 150 nm. These vesicles contain a variety of endogenous molecules, such as proteins, DNA, mRNA, microRNA, and circular RNA. They are essential for a wide range of metabolic events and have the potential to be used as therapeutic or diagnostic targets for a number of diseases, including ovarian diseases. By inducing changes in the surrounding environment, the donor exosomes transfer their contents to the receiving cells, so demonstrating the biological implications of major interactions between cells. Mesenchymal stem cells (MSCs) have produced exosomes have shown promise as a treatment for premature organ failure (POF or POI). Furthermore, exosomal transport has many complexities, and contributes to the pathophysiology of ovarian cancer by affecting cell growth, migration, metastastsis and etc. Owing to these facts, in this paper, we present the progress developed in the understanding of exosomes as a viable therapeutic avenue and indisputable prognostic targets in ovarian disorders.
Collapse
Affiliation(s)
- Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia.
| | - Bandar Almutairy
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia.
| | | | | | - Mohammad S Alzahrani
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif , Saudi Arabia
| |
Collapse
|
15
|
Bedia JS, Jacobs IJ, Ryan A, Gentry-Maharaj A, Burnell M, Singh N, Manchanda R, Kalsi JK, Dawnay A, Fallowfield L, McGuire AJ, Campbell S, Parmar MKB, Menon U, Skates SJ. Estimating the ovarian cancer CA-125 preclinical detectable phase, in-vivo tumour doubling time, and window for detection in early stage: an exploratory analysis of UKCTOCS. EBioMedicine 2025; 112:105554. [PMID: 39808947 PMCID: PMC11782890 DOI: 10.1016/j.ebiom.2024.105554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 12/20/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The ovarian cancer (OC) preclinical detectable phase (PCDP), defined as the interval during which cancer is detectable prior to clinical diagnosis, remains poorly characterised. We report exploratory analyses from the United Kingdom Collaborative Trial of Ovarian Cancer Screening (UKCTOCS). METHODS In UKCTOCS between Apr-2001 and Sep-2005, 101,314 postmenopausal women were randomised to no screening (NS) and 50,625 to annual multimodal screening (MMS) (until Dec-2011) using serum CA-125 interpreted by the Risk of Ovarian Cancer Algorithm (ROCA). All provided a baseline blood sample. Women with invasive epithelial OC diagnosed between randomisation and trial censorship (Dec-2014) in the MMS and NS arms with two or more CA-125 measurements, including one within two years of diagnosis were included. OC-free women (2:1 to cases) from the MMS arm provided information on baseline CA-125 distribution. CA-125 measurements were obtained from MMS results, secondary analysis of baseline samples, and medical records. PCDP duration and in-vivo tumour doubling time were estimated using the change-point model underlying ROCA. Early-stage (Stage I and II) PCDP was estimated from a Bayesian model for the probability of early stage given a CA-125 measurement. FINDINGS Of 541 women (2371 CA-125 measurements) with high-grade serous cancer (HGSC), 93% (504/541) secreted CA-125 into the circulation. Median CA-125 PCDP duration for clinically-diagnosed HGSC was 15.2 (IQR 13.1-16.9, 95% IPR 9.6-21.8) months, of which 11.9 (IQR 10.5-13.1, 95% IPR 7.5-16.5) months was in early stage. The median HGSC in-vivo tumour doubling time for cancers secreting CA-125 was 2.9 (IQR 2.3-3.7, 95% IPR 1.5-7.6) months. INTERPRETATION We report a comprehensive characterisation of the OC CA-125 PCDP. The 12-month window for early-stage detection and short tumour doubling time of HGSC provide a benchmark for researchers evaluating novel screening approaches including need to reduce diagnostic workup interval. Equally the findings provide urgent impetus for clinicians to reduce intervals from presentation to treatment onset. FUNDING NCI Early Detection Research Network, Concord (MA) Detect Ovarian Cancer Early Fund, MRC Clinical Trials Unit at UCL Core Funding.
Collapse
Affiliation(s)
- Jacob S Bedia
- MGH Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA
| | - Ian J Jacobs
- Department of Women's Cancer, Elizabeth Garrett Anderson Institute for Women's Health, UCL, London, UK
| | - Andy Ryan
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Aleksandra Gentry-Maharaj
- Department of Women's Cancer, Elizabeth Garrett Anderson Institute for Women's Health, UCL, London, UK; MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Matthew Burnell
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Naveena Singh
- Department of Cellular Pathology, Barts Health NHS Trust, London, UK
| | - Ranjit Manchanda
- Centre for Cancer Screening, Prevention & Early Diagnosis, Wolfson Institute of Population Health, Queen Mary University of London, London, UK; Department of Health Services Research, Faculty of Public Health & Policy, London School of Hygiene & Tropical Medicine, London, UK; Department of Gynaecological Oncology, Barts Health NHS Trust, London, UK; MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Jatinderpal K Kalsi
- AGE Research Unit, School of Public Health, Imperial College London, London, UK
| | - Anne Dawnay
- Department of Clinical Biochemistry, Barts Health NHS Trust, London, UK
| | - Lesley Fallowfield
- Sussex Health Outcomes Research and Education in Cancer (SHORE-C), Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | | | | | - Mahesh K B Parmar
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Usha Menon
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, UCL, London, UK
| | - Steven J Skates
- MGH Biostatistics Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Xiao X, Long F, Yu S, Wu W, Nie D, Ren X, Li W, Wang X, Yu L, Wang P, Wang G. Col1A1 as a new decoder of clinical features and immune microenvironment in ovarian cancer. Front Immunol 2025; 15:1496090. [PMID: 39845977 PMCID: PMC11750837 DOI: 10.3389/fimmu.2024.1496090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/12/2024] [Indexed: 01/24/2025] Open
Abstract
Backgrounds Collagen type I alpha 1 chain (COL1A1) is a key protein encoding fibrillar collagen, playing a crucial role in the tumor microenvironment (TME) due to its complex functions and close association with tumor invasiveness. This has made COL1A1 a focal point in cancer biology research. However, studies investigating the relationship between COL1A1 expression levels and clinical characteristics of ovarian cancer (OC) remain limited. Methods This study integrated resources from publicly available online databases and immunohistochemistry (IHC) techniques to analyze and validate COL1A1 expression in OC tissues, and evaluated its potential association with clinical features in OC patients. The prognostic value of COL1A1 was assessed using Kaplan-Meier (KM) survival curve analysis. The TIMER and TISIDB databases to explore the potential relationship between COL1A1 expression and immune microenvironment in OC tissues. The LinkedOmics and INPUT2 databases were used to analyze differential gene expression in OC, This was followed by enrichment analysis using the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) annotations to identify and predict potential signaling pathways associated with COL1A1. Results Our study demonstrated that COL1A1 expression was significantly elevated in OC tissues compared to normal ovarian tissues. This elevated expression was closely associated with tumor metastasis, poor prognosis, and advanced pathological stages in OC patients. Moreover, COL1A1 expression showed a significant correlation with immune cell infiltration and the expression of immune-related genes within the TME.Further analyses revealed that COL1A1 and its co-expressed genes were primarily enriched in key signaling pathways involved in OC invasion, metastasis, and angiogenesis, indicating its potential role in driving OC progression. Conclusions Our study found that upregulation of COL1A1 expression is significantly associated with lymph node metastasis of OC and can affect the immune microenvironment. Based on this, COL1A1 could serve as a promising biomarker for OC prognosis and provide a new perspective for the development of potential immunotherapies for patients with OC.
Collapse
Affiliation(s)
- Xiao Xiao
- Department of Gynecology, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Fangyi Long
- Laboratory Medicine Center, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Shaolan Yu
- Laboratory Medicine Center, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Wengjuan Wu
- Department of Gynecology, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Dayan Nie
- Laboratory Medicine Center, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xiaoyan Ren
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Wen Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xujuan Wang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Ling Yu
- Laboratory Medicine Center, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Pinghan Wang
- Laboratory Medicine Center, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Gang Wang
- Department of Gynecology, Sichuan Provincial Women’s and Children’s Hospital, The Affiliated Women’s and Children’s Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Lukey A, Sowamber R, Huntsman D, Pearce CL, Howard AF, Meza R, Law MR, Phung MT, Hanley GE. Evaluating Ovarian Cancer Risk-Reducing Salpingectomy Acceptance: A Survey. CANCER RESEARCH COMMUNICATIONS 2025; 5:187-194. [PMID: 39785678 PMCID: PMC11780486 DOI: 10.1158/2767-9764.crc-24-0566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
SIGNIFICANCE This study found that many participants were willing to consider RRS to prevent ovarian cancer. Further research on RRS should be undertaken to understand how this can be best used for ovarian cancer prevention.
Collapse
Affiliation(s)
- Alexandra Lukey
- Faculty of Medicine, School of Population and Public Health, University of British Columbia, Vancouver, Canada
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Ramlogan Sowamber
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- BC Cancer Research Institute, Vancouver, Canada
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - David Huntsman
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- BC Cancer Research Institute, Vancouver, Canada
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Celeste Leigh Pearce
- Faculty of Applied Sciences, School of Nursing, University of British Columbia, Vancouver, Canada
| | - A. Fuchsia Howard
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan
| | - Rafael Meza
- Faculty of Medicine, School of Population and Public Health, University of British Columbia, Vancouver, Canada
- BC Cancer Research Institute, Vancouver, Canada
| | - Michael R. Law
- Faculty of Medicine, School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Minh Tung Phung
- Department of Population Health Sciences, University of Wisconsin–Madison, Madison, Wisconsin
| | - Gillian E. Hanley
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of British Columbia, Vancouver, Canada
- BC Cancer Research Institute, Vancouver, Canada
| |
Collapse
|
18
|
de Melo IG, Tavares V, Savva-Bordalo J, Rei M, Liz-Pimenta J, Pereira D, Medeiros R. Endothelial Dysfunction Markers in Ovarian Cancer: VTE Risk and Tumour Prognostic Outcomes. Life (Basel) 2024; 14:1630. [PMID: 39768338 PMCID: PMC11678387 DOI: 10.3390/life14121630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Ovarian cancer (OC) presents daunting lethality rates worldwide, with frequent late-stage diagnosis and chemoresistance, highlighting the need for improved prognostic approaches. Venous thromboembolism (VTE), a major cancer mortality factor, is partially driven by endothelial dysfunction (ED). ED's pro-inflammatory state fosters tumour progression, suggesting a VTE-independent link between ED and cancer. Given this triad's interplay, ED markers may influence OC behaviour and patients' prognosis. Thus, the impact of ED-related genes and single-nucleotide polymorphisms (SNPs) on OC-related VTE and patient thrombogenesis-independent prognosis was investigated. NOS3 upregulation was linked to lower VTE incidence (χ2, p = 0.013), while SELP upregulation was associated with shorter overall survival (log-rank test, p = 0.048). Dismissing patients with VTE before OC diagnosis, SELP rs6136 T allele carriers presented lower progression-free survival (log-rank test, p = 0.038). Nevertheless, due to the SNP minor allele underrepresentation, further investigation is required. Taken together, ED markers seem to exhibit roles that depend on the clinical context, such as tumour-related thrombogenesis or cancer prognosis. Validation with larger cohorts and more in-depth functional studies are needed for data clarification and potential therapeutic strategies exploitation to tackle cancer progression and thrombosis in OC patients.
Collapse
Affiliation(s)
- Inês Guerra de Melo
- Molecular Oncology and Viral Pathology Group, Research Centre of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
| | - Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Centre of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Joana Savva-Bordalo
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Mariana Rei
- Department of Gynaecology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal;
| | - Joana Liz-Pimenta
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
- Department of Medical Oncology, Centro Hospitalar de Trás-os-Montes e Alto Douro (CHTMAD), 5000-508 Vila Real, Portugal
| | - Deolinda Pereira
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Centre of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
19
|
Koyanagi T, Saga Y, Takahashi Y, Tamura K, Takahashi S, Taneichi A, Takei Y, Mizukami H, Fujiwara H. Forced Vasohibin-1 Expression Increases Paclitaxel Sensitivity of Ovarian Cancer by Inhibiting Microtubule Activity. Cancer Rep (Hoboken) 2024; 7:e70100. [PMID: 39710372 DOI: 10.1002/cnr2.70100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/26/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Vasohibin-1 (VASH1), an angiogenic inhibitor, exhibits tubulin carboxypeptidase activity, which is involved in microtubule functions. Paclitaxel, the core chemotherapeutic agent for ovarian cancer chemotherapy, has a point of action on microtubules and may interact with VASH1. AIMS To examine the influence of VASH1 on intracellular tubulin detyrosination status, cyclin B1 expression, and paclitaxel chemosensitivity using VASH1-overexpressing ovarian cancer cell lines. METHODS AND RESULTS Gene-transfected human ovarian cancer cell lines were subjected to western blot analysis. Western blot analysis of VASH1-overexpressing ovarian cancer cells revealed upregulated expression of detyrosinated tubulin and cyclin B1 compared with control cells. By WST-1 assay, paclitaxel chemosensitivity of VASH1-overexpressing ovarian cancer cells was markedly enhanced compared with that of control cells, whereas there was no significant difference in chemosensitivity to cisplatin. The forced expression of VASH1 enhanced tubulin carboxypeptidase activity and increased cyclin B1 expression, resulting in augmented paclitaxel chemosensitivity in ovarian cancer cells. CONCLUSION Ovarian cancer treatment strategies targeting VASH1 can potentiate the effects of conventional chemotherapy by inhibiting angiogenesis and regulating microtubule activity.
Collapse
Grants
- 17K11294 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 19K18702 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 20K09627 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 22K09551 Ministry of Education, Culture, Sports, Science and Technology of Japan
Collapse
Affiliation(s)
- Takahiro Koyanagi
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yasushi Saga
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yoshifumi Takahashi
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Kohei Tamura
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Suzuyo Takahashi
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Akiyo Taneichi
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yuji Takei
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Hiroyuki Fujiwara
- Department of Obstetrics and Gynecology, School of Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
20
|
Li S, Lei N, Chen M, Guo R, Han L, Qiu L, Wu F, Jiang S, Tong N, Wang K, Li Y, Chang L. Exploration of organoids in ovarian cancer: From basic research to clinical translation. Transl Oncol 2024; 50:102130. [PMID: 39303357 PMCID: PMC11437877 DOI: 10.1016/j.tranon.2024.102130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024] Open
Abstract
Ovarian cancer is a highly heterogeneous tumor with a poor prognosis. The lack of reliable and efficient research models that can accurately mimic heterogeneity has impeded in-depth investigations and hindered the clinical translation of research findings in ovarian cancer. Organoid models have emerged as a promising in vitro approach, demonstrating remarkable fidelity to the histological, molecular, genomic, and transcriptomic features of their tissues of origin. In recent years, organoids have contributed to advancing our understanding of ovarian cancer initiation, metastasis, and drug resistance mechanisms, as well as facilitating clinical screening of effective therapeutic agents. The establishment of high-throughput organoid culture systems, coupled with cutting-edge technologies such as organ-on-a-chip, genetic engineering, and 3D printing, has tremendous potential for accelerating ovarian cancer research translation. In this review, we present a comprehensive overview of the latest exploration of organoids in basic ovarian cancer research and clinical translation. Furthermore, we discuss the prospects and challenges associated with the use of organoids and related novel technologies in the context of ovarian cancer. This review provides insights into the application of organoids in ovarian cancer.
Collapse
Affiliation(s)
- Siyu Li
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Ningjing Lei
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengyu Chen
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Ruixia Guo
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Liping Han
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Luojie Qiu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Fengling Wu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Shan Jiang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Ningyao Tong
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Kunmei Wang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| | - Yong Li
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia; Cancer Care Centre, St. George Hospital, Kogarah, NSW 2217, Australia.
| | - Lei Chang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China.
| |
Collapse
|
21
|
Braga EA, Burdennyy AM, Uroshlev LA, Zaichenko DM, Filippova EA, Lukina SS, Pronina IV, Astafeva IR, Fridman MV, Kazubskaya TP, Loginov VI, Dmitriev AA, Moskovtsev AA, Kushlinskii NE. Ten Hypermethylated lncRNA Genes Are Specifically Involved in the Initiation, Progression, and Lymphatic and Peritoneal Metastasis of Epithelial Ovarian Cancer. Int J Mol Sci 2024; 25:11843. [PMID: 39519394 PMCID: PMC11547154 DOI: 10.3390/ijms252111843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Our work aimed to evaluate and differentiate the role of ten lncRNA genes (GAS5, HAND2-AS1, KCNK15-AS1, MAGI2-AS3, MEG3, SEMA3B-AS1, SNHG6, SSTR5-AS1, ZEB1-AS1, and ZNF667-AS1) in the development and progression of epithelial ovarian cancer (EOC). A representative set of clinical samples was used: 140 primary tumors from patients without and with metastases and 59 peritoneal metastases. Using MS-qPCR, we demonstrated an increase in methylation levels of all ten lncRNA genes in tumors compared to normal tissues (p < 0.001). Using RT-qPCR, we showed downregulation and an inverse relationship between methylation and expression levels for ten lncRNAs (rs < -0.5). We further identified lncRNA genes that were specifically hypermethylated in tumors from patients with metastases to lymph nodes (HAND2-AS1), peritoneum (KCNK15-AS1, MEG3, and SEMA3B-AS1), and greater omentum (MEG3, SEMA3B-AS1, and ZNF667-AS1). The same four lncRNA genes involved in peritoneal spread were associated with clinical stage and tumor extent (p < 0.001). Interestingly, we found a reversion from increase to decrease in the hypermethylation level of five metastasis-related lncRNA genes (MEG3, SEMA3B-AS1, SSTR5-AS1, ZEB1-AS1, and ZNF667-AS1) in 59 peritoneal metastases. This reversion may be associated with partial epithelial-mesenchymal transition (EMT) in metastatic cells, as indicated by a decrease in the level of the EMT marker, CDH1 mRNA (p < 0.01). Furthermore, novel mRNA targets and regulated miRNAs were predicted for a number of the studied lncRNAs using the NCBI GEO datasets and analyzed by RT-qPCR and transfection of SKOV3 and OVCAR3 cells. In addition, hypermethylation of SEMA3B-AS1, SSTR5-AS1, and ZNF667-AS1 genes was proposed as a marker for overall survival in patients with EOC.
Collapse
Affiliation(s)
- Eleonora A. Braga
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (A.M.B.); (D.M.Z.); (E.A.F.); (S.S.L.); (I.V.P.); (I.R.A.); (V.I.L.); (A.A.M.)
| | - Alexey M. Burdennyy
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (A.M.B.); (D.M.Z.); (E.A.F.); (S.S.L.); (I.V.P.); (I.R.A.); (V.I.L.); (A.A.M.)
| | - Leonid A. Uroshlev
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia; (L.A.U.); (M.V.F.)
| | - Danila M. Zaichenko
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (A.M.B.); (D.M.Z.); (E.A.F.); (S.S.L.); (I.V.P.); (I.R.A.); (V.I.L.); (A.A.M.)
| | - Elena A. Filippova
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (A.M.B.); (D.M.Z.); (E.A.F.); (S.S.L.); (I.V.P.); (I.R.A.); (V.I.L.); (A.A.M.)
| | - Svetlana S. Lukina
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (A.M.B.); (D.M.Z.); (E.A.F.); (S.S.L.); (I.V.P.); (I.R.A.); (V.I.L.); (A.A.M.)
| | - Irina V. Pronina
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (A.M.B.); (D.M.Z.); (E.A.F.); (S.S.L.); (I.V.P.); (I.R.A.); (V.I.L.); (A.A.M.)
| | - Iana R. Astafeva
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (A.M.B.); (D.M.Z.); (E.A.F.); (S.S.L.); (I.V.P.); (I.R.A.); (V.I.L.); (A.A.M.)
| | - Marina V. Fridman
- Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia; (L.A.U.); (M.V.F.)
| | - Tatiana P. Kazubskaya
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (T.P.K.); (N.E.K.)
| | - Vitaly I. Loginov
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (A.M.B.); (D.M.Z.); (E.A.F.); (S.S.L.); (I.V.P.); (I.R.A.); (V.I.L.); (A.A.M.)
| | - Alexey A. Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia;
| | - Aleksey A. Moskovtsev
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; (A.M.B.); (D.M.Z.); (E.A.F.); (S.S.L.); (I.V.P.); (I.R.A.); (V.I.L.); (A.A.M.)
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (T.P.K.); (N.E.K.)
- Russian Medical Academy of Continuing Professional Education, 125993 Moscow, Russia
| | - Nikolay E. Kushlinskii
- N.N. Blokhin National Medical Research Center of Oncology, 115478 Moscow, Russia; (T.P.K.); (N.E.K.)
| |
Collapse
|
22
|
Braga EA, Filippova EA, Uroshlev LA, Lukina SS, Pronina IV, Kazubskaya TP, Kushlinskiy DN, Loginov VI, Fridman MV, Burdennyy AM, Kushlinskii NE. LncRNA Genes of the SNHG Family: Co-methylation and Common Functions in Ovarian Cancer. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:2051-2068. [PMID: 39647832 DOI: 10.1134/s0006297924110178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/01/2024] [Accepted: 09/03/2024] [Indexed: 12/10/2024]
Abstract
Genes of the small nucleolar RNA host gene (SNHG) family may participate in oncogenesis through the regulatory functions of encoded long non-coding RNAs (lncRNAs) and by influencing formation of small nucleolar RNAs and ribosome biogenesis. The aim of this work was to evaluate changes in the methylation levels and extent of co-methylation of the SNHG family lncRNA genes (SNHG1, GAS5/SNHG2, SNHG6, SNHG12, SNHG17) in clinical samples of ovarian cancer (OC) as an indication for the similarity of their roles in oncogenesis. Analysis of a representative set of 122 OC samples by quantitative methylation-specific PCR showed a statistically significant (p < 0.01-0.0001) increase in the methylation level of all five studied lncRNA genes. There was also a correlation between the increased methylation levels of GAS5, SNHG6, and SNHG12 and OC progression (clinical stage, tumor size, and metastasis), indicating possible functional significance of hypermethylation of these genes. For four genes (SNHG1, GAS5, SNHG6, and SNHG12), a statistically significant pairwise positive correlation of methylation levels (co-methylation) was observed (rs > 0.35; p ≤ 0.001), which was in agreement with the GEPIA 2.0 data (426 OC samples) showing co-expression of these genes (rs > 0.5; p < 0.001). The correlation between the expression levels of GAS5 and SNHG6 was confirmed by RT-qPCR (rs = 0.46; p = 0.007). Bioinformatics analysis predicted miRNAs common for the SNHG1, GAS5, SNHG6, and SNHG12 lncRNA and potentially capable of interacting with one or more of these lncRNAs via competing endogenous RNA mechanism, as well as mRNAs, whose expression might be affected by the studied lncRNAs. We also investigated a possible involvement of genes for these mRNAs in oncogenesis-related processes, such as RNA processing and splicing and epithelial-mesenchymal transition. As a result of this work, four SNHG family lncRNAs with coregulation and joint putative biological functions in the pathogenesis of OC were identified.
Collapse
Affiliation(s)
- Eleonora A Braga
- Research Institute of General Pathology and Pathophysiology, Moscow, 125315, Russia.
| | - Elena A Filippova
- Research Institute of General Pathology and Pathophysiology, Moscow, 125315, Russia
| | - Leonid A Uroshlev
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Svetlana S Lukina
- Research Institute of General Pathology and Pathophysiology, Moscow, 125315, Russia
| | - Irina V Pronina
- Research Institute of General Pathology and Pathophysiology, Moscow, 125315, Russia
| | - Tatyana P Kazubskaya
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115522, Russia
| | - Dmitry N Kushlinskiy
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115522, Russia
| | - Vitaliy I Loginov
- Research Institute of General Pathology and Pathophysiology, Moscow, 125315, Russia
| | - Marina V Fridman
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, 119991, Russia
| | - Alexey M Burdennyy
- Research Institute of General Pathology and Pathophysiology, Moscow, 125315, Russia.
| | - Nikolay E Kushlinskii
- Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, Moscow, 115522, Russia
| |
Collapse
|
23
|
Zhu B, Gu H, Mao Z, Beeraka NM, Zhao X, Anand MP, Zheng Y, Zhao R, Li S, Manogaran P, Fan R, Nikolenko VN, Wen H, Basappa B, Liu J. Global burden of gynaecological cancers in 2022 and projections to 2050. J Glob Health 2024; 14:04155. [PMID: 39148469 PMCID: PMC11327849 DOI: 10.7189/jogh.14.04155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024] Open
Abstract
Background The incidence and mortality of gynaecological cancers can significantly impact women's quality of life and increase the health care burden for organisations globally. The objective of this study was to evaluate global inequalities in the incidence and mortality of gynaecological cancers in 2022, based on The Global Cancer Observatory (GLOBOCAN) 2022 estimates. The future burden of gynaecological cancers (GCs) in 2050 was also projected. Methods Data regarding to the total cases and deaths related to gynaecological cancer, as well as cases and deaths pertaining to different subtypes of GCs, gathered from the GLOBOCAN database for the year 2022. Predictions for the number of cases and deaths in the year 2050 were derived from global demographic projections, categorised by world region and Human Development Index (HDI). Results In 2022, there were 1 473 427 new cases of GCs and 680 372 deaths. The incidence of gynecological cancer reached 30.3 per 100 000, and the mortality rate hit 13.2 per 100 000. The age-standardised incidence of GCs in Eastern Africa is higher than 50 per 100 000, whereas the age-standardised incidence in Northern Africa is 17.1 per 100 000. The highest mortality rates were found in East Africa (ASMR (age-standardised mortality rates) of 35.3 per 100 000) and the lowest in Australia and New Zealand (ASMR of 8.1 per 100 000). These are related to the endemic areas of HIV and HPV. Very High HDI countries had the highest incidence of GCs, with ASIR (age-standardised incidence rates) of 34.8 per 100 000, and low HDI countries had the second highest incidence rate, with an ASIR of 33.0 per 100 000. Eswatini had the highest incidence and mortality (105.4 per 100 000; 71.1 per 100 000) and Yemen the lowest (5.8 per 100 000; 4.4 per 100 000). If the current trends in morbidity and mortality are maintained, number of new cases and deaths from female reproductive tract tumours is projected to increase over the next two decades. Conclusions In 2022, gynaecological cancers accounted for 1 473 427 new cases and 680 372 deaths globally, with significant regional disparities in incidence and mortality rates. The highest rates were observed in Eastern Africa and countries with very high and low HDI, with Eswatini recording the most severe statistics. If current trends continue, the number of new cases and deaths from gynaecological cancers is expected to rise over the next two decades, highlighting the urgent need for effective interventions.
Collapse
Affiliation(s)
- Binhua Zhu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hao Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhihan Mao
- Henan Medical College, Zhengzhou University, Zhengzhou, China
| | - Narasimha M Beeraka
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, India
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Xiang Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mahesh Padukudru Anand
- Department of Pulmonary Medicine, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| | - Yufei Zheng
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruiwen Zhao
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Siting Li
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Prasath Manogaran
- Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
- Department of Clinical and Translational Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| | - Ruitai Fan
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Vladimir N Nikolenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Haixiao Wen
- Department of Gynecologic Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Mysore, Karnataka, India
| | - Junqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
Alblihy A. From desert flora to cancer therapy: systematic exploration of multi-pathway mechanisms using network pharmacology and molecular modeling approaches. Front Pharmacol 2024; 15:1345415. [PMID: 38666020 PMCID: PMC11043532 DOI: 10.3389/fphar.2024.1345415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Ovarian cancer, often labeled a "silent killer," remains one of the most compelling and challenging areas of cancer research. In 2019 alone, a staggering 222,240 new cases of ovarian cancer were reported, with nearly 14,170 lives tragically lost to this relentless disease. The absence of effective diagnostic methods, increased resistance to chemotherapy, and the heterogeneous nature of ovarian cancer collectively contribute to the unfavorable prognosis observed in the majority of cases. Thus, there is a pressing need to explore therapeutic interventions that offer superior efficacy and safety, thereby enhancing the survival prospects for ovarian cancer patients. Recognizing this potential, our research synergizes bioinformatics with a network pharmacology approach to investigate the underlying molecular interactions of Saudi Arabian flora (Onopordum heteracanthum, Acacia ehrenbergiana, Osteospermum vaillantii, Cyperus rotundus, Carissa carandas, Carissa spinarum, and Camellia sinensis) in ovarian cancer treatment. At first, phytoconstituents of indigenous flora and their associated gene targets, particularly those pertinent to ovarian cancer, were obtained from open-access databases. Later, the shared targets of plants and diseases were compared to identify common targets. A protein-protein interaction (PPI) network of predicted targets was then constructed for the identification of key genes having the highest degree of connectivity among networks. Following that, a compound-target protein-pathway network was constructed, which uncovered that, namely, hispidulin, stigmasterol, ascorbic acid, octopamine, cyperene, kaempferol, pungenin, citric acid, d-tartaric acid, beta-sitosterol, (-)-epicatechin gallate, and (+)-catechin demonstrably influence cell proliferation and growth by impacting the AKT1 and VEGFA proteins. Molecular docking, complemented by a 20-ns molecular dynamic (MD) simulation, was used, and the binding affinity of the compound was further validated. Molecular docking, complemented by a 20-ns MD simulation, confirmed the binding affinity of these compounds. Specifically, for AKT1, ascorbic acid showed a docking score of -11.1227 kcal/mol, interacting with residues Ser A:240, Leu A:239, Arg A:243, Arg C:2, and Glu A:341. For VEGFA, hispidulin exhibited a docking score of -17.3714 kcal/mol, interacting with Asn A:158, Val A:190, Gln B:160, Ser A:179, and Ser B:176. To sum up, both a theoretical and empirical framework were established by this study, directing more comprehensive research and laying out a roadmap for the potential utilization of active compounds in the formulation of anti-cancer treatments.
Collapse
Affiliation(s)
- Adel Alblihy
- Medical Center, King Fahad Security College (KFSC), Riyadh, Saudi Arabia
- Department of Criminal Justice and Forensic Sciences, King Fahad Security Collage, Riyadh, Saudi Arabia
| |
Collapse
|