Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Eur J Med Res ; 29(1): 231, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38609993

ABSTRACT

BACKGROUND: High-grade serous ovarian carcinoma (HGSOC) is the most aggressive and prevalent subtype of ovarian cancer and accounts for a significant portion of ovarian cancer-related deaths worldwide. Despite advancements in cancer treatment, the overall survival rate for HGSOC patients remains low, thus highlighting the urgent need for a deeper understanding of the molecular mechanisms driving tumorigenesis and for identifying potential therapeutic targets. Whole-exome sequencing (WES) has emerged as a powerful tool for identifying somatic mutations and alterations across the entire exome, thus providing valuable insights into the genetic drivers and molecular pathways underlying cancer development and progression. METHODS: Via the analysis of whole-exome sequencing results of tumor samples from 90 ovarian cancer patients, we compared the mutational landscape of ovarian cancer patients with that of TCGA patients to identify similarities and differences. The sequencing data were subjected to bioinformatics analysis to explore tumor driver genes and their functional roles. Furthermore, we conducted basic medical experiments to validate the results obtained from the bioinformatics analysis. RESULTS: Whole-exome sequencing revealed the mutational profile of HGSOC, including BRCA1, BRCA2 and TP53 mutations. AP3S1 emerged as the most weighted tumor driver gene. Further analysis of AP3S1 mutations and expression demonstrated their associations with patient survival and the tumor immune response. AP3S1 knockdown experiments in ovarian cancer cells demonstrated its regulatory role in tumor cell migration and invasion through the TGF-ß/SMAD pathway. CONCLUSION: This comprehensive analysis of somatic mutations in HGSOC provides insight into potential therapeutic targets and molecular pathways for targeted interventions. AP3S1 was identified as being a key player in tumor immunity and prognosis, thus providing new perspectives for personalized treatment strategies. The findings of this study contribute to the understanding of HGSOC pathogenesis and provide a foundation for improved outcomes in patients with this aggressive disease.


Subject(s)
Ovarian Neoplasms , Humans , Female , Exome Sequencing , Ovarian Neoplasms/genetics , Carcinogenesis , Computational Biology
2.
Life Sci ; 320: 121512, 2023 May 01.
Article in English | MEDLINE | ID: mdl-36858312

ABSTRACT

Endometrial cancer (EC) is a malignant tumor with a high incidence in women, and the survival rate of high-risk patients decreases significantly after disease progression. The regulatory role of long non-coding RNAs (LncRNAs) in tumors has been widely appreciated, but there have been few studies in EC. To investigate the effect of HOXB-AS3 in EC, we used bioinformatics tools for prediction and collected clinical samples to detect the expression of HOXB-AS3. Colony formation assay, MTT assay, flow cytometry and apoptosis assay, and transwell assay were used to verify the role of HOXB-AS3 in EC. HOXB-AS3 was upregulated in EC, promoted the proliferation and invasive ability of EC cells, and inhibited apoptosis. In addition, the ROC curve illustrated its diagnostic value. We explored experiments via lentiviral transduction, FISH, Oil Red O staining, TC and FFA content detection, RNA-pulldown, RIP, and other mechanisms to reveal that HOXB-AS3 can bind to PTBP1 and co-regulate the expression of SREBP1, thereby regulating lipid metabolism in EC cells. To the best of our knowledge, this is the first study on HOXB-AS3 in disorders of lipid metabolism in EC. In addition, we believe HOXB-AS3 has the potential to be a neoplastic marker or a therapeutic target.


Subject(s)
Carcinoma, Endometrioid , Endometrial Neoplasms , MicroRNAs , RNA, Long Noncoding , Female , Humans , Carcinoma, Endometrioid/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Endometrial Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Lipid Metabolism , MicroRNAs/genetics , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
3.
Mol Oncol ; 16(3): 813-829, 2022 02.
Article in English | MEDLINE | ID: mdl-33751805

ABSTRACT

Long noncoding RNAs (lncRNAs) have a profound effect on biological processes in various malignancies. However, few studies have investigated their functions and specific mechanisms in endometrial cancer. In this study, we focused on the role and mechanism of lncRNA-ZXF1 in endometrial cancer. Bioinformatics and in vitro and in vivo experiments were used to explore the expression and function of lncRNA-ZXF1. We found that lncRNA-ZXF1 altered the migration and invasion of endometrioid endometrial cancer (EEC) cells. Furthermore, our results suggest that lncRNA-ZXF1 regulates EEC cell proliferation. This regulation may be achieved by the lncRNA-ZXF1-mediated alteration in the expression of P21 through two mechanisms. One is that lncRNA-ZXF1 functions as a molecular sponge of miR-378a-3p to regulate PCDHA3 expression and then modulate the expression of P21. The other is that lncRNA-ZXF1 inhibits CDC20-mediated degradation of ubiquitination by directly binding to P21. To the best of our knowledge, this study is the first to explore lncRNA-ZXF1 functioning as a tumor-suppressing lncRNA in EEC. LncRNA-ZXF1 may become therapeutic, diagnostic, and prognostic indicator in the future.


Subject(s)
Endometrial Neoplasms , MicroRNAs , RNA, Long Noncoding , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Endometrial Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Ubiquitination/genetics
4.
Biochem Biophys Res Commun ; 548: 148-154, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33640608

ABSTRACT

Endocrine therapy is a promising treatment for endometrial cancer (EC) that preserves fertility, however, progesterone-resistance is currently the major challenges. The Cancer Genome Atlas (TCGA) database analysis showed that CNR1 was closely have a negative correlation with overall survival (OS) and relapse-free survival (RFS) in endometrial cancer. To explore the role of CNR1 in progesterone resistance and possible molecular regulation mechanism, we established stable progesterone-resistant cell lines (IshikawaPR) via progesterone tolerance of ordinary cancer cells (Ishikawa). The difference of CNR1 level in two cell lines was assessed by MTT, RT-PCR, Western blot, immunofluorescence. Then, lentiviruses constructed CNR1-knockdown with GV248 as the tool vector were used to transfect IshikwaPR cells, and the changes of biological behavior and progesterone sensitivity was verified respectively through plate cloning experiment, EdU assay, flow cytometry cycle analysis, transwell, Scratch test, etc. We founded after CNR1 was knocked down, the proliferative activity and ability to migrate of IshikawaPR cells decreased, progesterone-response sensitivity could be improved. Moreover, knockdown of CNR1 can also down-regulate ERK and NFκ B expression and activation. Furthermore, subcutaneous xenograft in nude mice was tested similarly in vivo. The above datas suggest that targeting CNR1 may reverse the progesterone resistance in endometrial cancer and may coordinate the role of ERK pathway activation.


Subject(s)
Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Endometrium/abnormalities , MAP Kinase Signaling System , Receptor, Cannabinoid, CB1/metabolism , Uterine Diseases/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Endometrial Neoplasms/genetics , Endometrium/metabolism , Endometrium/pathology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , MAP Kinase Signaling System/drug effects , Medroxyprogesterone/pharmacology , Mice, Inbred BALB C , Mice, Nude , Receptor, Cannabinoid, CB1/genetics , Up-Regulation/drug effects , Up-Regulation/genetics , Uterine Diseases/genetics , Uterine Diseases/pathology
5.
Cancer Med ; 8(9): 4380-4388, 2019 08.
Article in English | MEDLINE | ID: mdl-31215145

ABSTRACT

Progestin resistance limits the effectiveness of progestin therapy in endometrial carcinoma for patients who desire to preserve fertility. To investigate the molecular mechanism of progestin resistance in endometrial carcinoma, we performed microarray analysis among Ishikawa and progestin resistant cell IshikawaPR cells. We found that epithelial to mesenchymal transition (EMT) was involved in progestin resistance and dachshund family transcription factor 1 (DACH1) is positively correlated with progesterone receptor (PGR). Knockdown of DACH1 in Ishikawa cell promoted proliferation, metastasis ability, and resistance to progestin. Conversely, overexpression of DACH1 in IshikawaPR cell rendered more sensitive to progestin treatment. Xenograft model assay also had similar results. In addition, our data showed that DACH1 overexpression inhibited EMT and decreased c-Jun, Notch1 and Hes1expression. Our study demonstrated for the first time that EMT is involved in progestin resistance of EC. The response to progestin could be reserved by DACH1 suppressed EMT through Notch1 pathway via c-Jun.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Endometrial Neoplasms/drug therapy , Eye Proteins/genetics , Signal Transduction/drug effects , Transcription Factors/genetics , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Knockdown Techniques , Humans , Mice , Progestins/administration & dosage , Progestins/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Receptor, Notch1/metabolism , Receptors, Progesterone/genetics , Xenograft Model Antitumor Assays
6.
J Transl Med ; 17(1): 58, 2019 02 27.
Article in English | MEDLINE | ID: mdl-30813939

ABSTRACT

BACKGROUND: Progesterone resistance is a problem in endometrial carcinoma, and its underlying molecular mechanisms remain poorly understood. The aim of this study was to elucidate the molecular mechanisms of progesterone resistance and to identify the key genes and pathways mediating progesterone resistance in endometrial cancer using bioinformatics analysis. METHODS: We developed a stable MPA (medroxyprogesterone acetate)-resistant endometrial cancer cell subline named IshikawaPR. Microarray analysis was used to identify differentially expressed genes (DEGs) from triplicate samples of Ishikawa and IshikawaPR cells. PANTHER, DAVID and Metascape were used to perform gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, and cBioPortal for progesterone receptor (PGR) coexpression analysis. GEO microarray (GSE17025) was utilized for validation. The protein-protein interaction network (PPI) and modular analyses were performed using Metascape and Cytoscape. Further validation were performed by real-time polymerase chain reaction (RT-PCR). RESULTS: In total, 821 DEGs were found and further analyzed by GO, KEGG pathway enrichment and PPI analyses. We found that lipid metabolism, immune system and inflammation, extracellular environment-related processes and pathways accounted for a significant portion of the enriched terms. PGR coexpression analysis revealed 7 PGR coexpressed genes (ANO1, SOX17, CGNL1, DACH1, RUNDC3B, SH3YL1 and CRISPLD1) that were also dramatically changed in IshikawaPR cells. Kaplan-Meier survival statistics revealed clinical significance for 4 out of 7 target genes. Furthermore, 8 hub genes and 4 molecular complex detections (MCODEs) were identified. CONCLUSIONS: Using microarray and bioinformatics analyses, we identified DEGs and determined a comprehensive gene network of progesterone resistance. We offered several possible mechanisms of progesterone resistance and identified therapeutic and prognostic targets of progesterone resistance in endometrial cancer.


Subject(s)
Computational Biology/methods , Endometrial Neoplasms/genetics , Endometrium/abnormalities , Uterine Diseases/genetics , Cell Line, Tumor , Down-Regulation/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Ontology , Gene Regulatory Networks , Humans , Kaplan-Meier Estimate , Protein Interaction Maps/genetics , Receptors, Progesterone/metabolism , Reproducibility of Results , Signal Transduction/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...