8 results on '"Cho U"'
Search Results
2. Mitochondrial fission enhances IL-6-induced metastatic potential in ovarian cancer via ERK1/2 activation.
- Author
-
Lee J, Han Y, Kim S, Jo H, Wang W, Cho U, Kim SI, Kim B, and Song YS
- Subjects
- Humans, Female, Cell Line, Tumor, Neoplasm Metastasis, Mitochondria metabolism, Receptors, Interleukin-6 metabolism, Cell Movement drug effects, Mitochondrial Dynamics drug effects, Ovarian Neoplasms pathology, Ovarian Neoplasms metabolism, Interleukin-6 metabolism, Dynamins metabolism, MAP Kinase Signaling System drug effects
- Abstract
Ovarian cancer is a lethal gynecologic cancer mostly diagnosed in an advanced stage with an accumulation of ascites. Interleukin-6 (IL-6), a pro-inflammatory cytokine is highly elevated in malignant ascites and plays a pleiotropic role in cancer progression. Mitochondria are dynamic organelles that undergo fission and fusion in response to external stimuli and dysregulation in their dynamics has been implicated in cancer progression and metastasis. Here, we investigate the effect of IL-6 on mitochondrial dynamics in ovarian cancer cells (OVCs) and its impact on metastatic potential. Treatment with IL-6 on ovarian cancer cell lines (SKOV3 and PA-1) led to an elevation in the metastatic potential of OVCs. Interestingly, a positive association was observed between dynamin-related protein 1 (Drp1), a regulator of mitochondrial fission, and IL-6R in metastatic ovarian cancer tissues. Additionally, IL-6 treatment on OVCs was linked to the activation of Drp1, with a notable increase in the ratio of the inhibitory form p-Drp1(S637) to the active form p-Drp1(S616), indicating enhanced mitochondrial fission. Moreover, IL-6 treatment triggered the activation of ERK1/2, and inhibiting ERK1/2 mitigated IL-6-induced mitochondrial fission. Suppressing mitochondrial fission through siRNA transfection and a pharmacological inhibitor reduced the IL-6-induced migration and invasion of OVCs. This was further supported by 3D invasion assays using patient-derived spheroids. Altogether, our study suggests the role of mitochondrial fission in the metastatic potential of OVCs induced by IL-6. The inhibition of mitochondrial fission could be a potential therapeutic approach to suppress the metastasis of ovarian cancer., (© 2024 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.)
- Published
- 2024
- Full Text
- View/download PDF
3. Stearoyl-CoA desaturase 1 inhibition induces ER stress-mediated apoptosis in ovarian cancer cells.
- Author
-
Lee J, Jang S, Im J, Han Y, Kim S, Jo H, Wang W, Cho U, Kim SI, Seol A, Kim B, and Song YS
- Subjects
- Female, Humans, Apoptosis, Endoplasmic Reticulum Stress, Carcinoma, Ovarian Epithelial, Lipids, Stearoyl-CoA Desaturase metabolism, Ovarian Neoplasms
- Abstract
Ovarian cancer is a leading cause of death among gynecologic tumors, often detected at advanced stages. Metabolic reprogramming and increased lipid biosynthesis are key factors driving cancer cell growth. Stearoyl-CoA desaturase 1 (SCD1) is a crucial enzyme involved in de novo lipid synthesis, producing mono-unsaturated fatty acids (MUFAs). Here, we aimed to investigate the expression and significance of SCD1 in epithelial ovarian cancer (EOC). Comparative analysis of normal ovarian surface epithelial (NOSE) tissues and cell lines revealed elevated SCD1 expression in EOC tissues and cells. Inhibition of SCD1 significantly reduced the proliferation of EOC cells and patient-derived organoids and induced apoptotic cell death. Interestingly, SCD1 inhibition did not affect the viability of non-cancer cells, indicating selective cytotoxicity against EOC cells. SCD1 inhibition on EOC cells induced endoplasmic reticulum (ER) stress by activating the unfolded protein response (UPR) sensors and resulted in apoptosis. The addition of exogenous oleic acid, a product of SCD1, rescued EOC cells from ER stress-mediated apoptosis induced by SCD1 inhibition, underscoring the importance of lipid desaturation for cancer cell survival. Taken together, our findings suggest that the inhibition of SCD1 is a promising biomarker as well as a novel therapeutic target for ovarian cancer by regulating ER stress and inducing cancer cell apoptosis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
4. Prediction of homologous recombination deficiency from Oncomine Comprehensive Assay Plus correlating with SOPHiA DDM HRD Solution.
- Author
-
Kang J, Na K, Kang H, Cho U, Kwon SY, Hwang S, and Lee A
- Subjects
- Female, Humans, Allelic Imbalance, Poly(ADP-ribose) Polymerases genetics, Genomic Instability, Homologous Recombination, Ovarian Neoplasms genetics, Ovarian Neoplasms pathology
- Abstract
Objective: Poly(ADP-ribose) polymerase (PARP) inhibitors are used for targeted therapy for ovarian cancer with homologous recombination deficiency (HRD). In this study, we aimed to develop a homologous recombination deficiency prediction model to predict the genomic integrity (GI) index of the SOPHiA DDM HRD Solution from the Oncomine Comprehensive Assay (OCA) Plus. We also tried to a find cut-off value of the genomic instability metric (GIM) of the OCA Plus that correlates with the GI index of the SOPHiA DDM HRD Solution., Methods: We included 87 cases with high-grade ovarian serous carcinoma from five tertiary referral hospitals in Republic of Korea. We developed an HRD prediction model to predict the GI index of the SOPHiA DDM HRD Solution. As predictor variables in the model, we used the HRD score, which included percent loss of heterozygosity (%LOH), percent telomeric allelic imbalance (%TAI), percent large-scale state transitions (%LST), and the genomic instability metric (GIM). To build the model, we employed a penalized logistic regression technique., Results: The final model equation is -21.77 + 0.200 × GIM + 0.102 × %LOH + 0.037 × %TAI + 0.261 × %LST. To improve the performance of the prediction model, we added a borderline result category to the GI results. The accuracy of our HRD status prediction model was 0.958 for the test set. The accuracy of HRD status using GIM with a cut-off value of 16 was 0.911., Conclusion: The Oncomine Comprehensive Assay Plus provides a reliable biomarker for homologous recombination deficiency., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2024 Kang et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
5. Computational modeling of malignant ascites reveals CCL5-SDC4 interaction in the immune microenvironment of ovarian cancer.
- Author
-
Kim S, Han Y, Kim SI, Lee J, Jo H, Wang W, Cho U, Park WY, Rando TA, Dhanasekaran DN, and Song YS
- Subjects
- Ascites metabolism, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Cell Line, Tumor, Cell Polarity, Disease Progression, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Killer Cells, Natural metabolism, Macrophages metabolism, Middle Aged, Models, Theoretical, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Prognosis, Sequence Analysis, RNA, Single-Cell Analysis methods, Survival Analysis, T-Lymphocytes metabolism, Tumor Microenvironment, Ascites pathology, Chemokine CCL5 genetics, Chemokine CCL5 metabolism, Ovarian Neoplasms mortality, Syndecan-4 genetics, Syndecan-4 metabolism
- Abstract
Fluid accumulation in the abdominal cavity is commonly found in advanced-stage ovarian cancer patients, which creates a specialized tumor microenvironment for cancer progression. Using single-cell RNA sequencing (scRNA-seq) of ascites cells from five patients with ovarian cancer, we identified seven cell types, including heterogeneous macrophages and ovarian cancer cells. We resolved a distinct polarization state of macrophages by MacSpectrum analysis and observed subtype-specific enrichment of pathways associated with their functions. The communication between immune and cancer cells was predicted through a putative ligand-receptor pair analysis using NicheNet. We found that CCL5, a chemotactic ligand, is enriched in immune cells (T cells and NK cells) and mediates ovarian cancer cell survival in the ascites, possibly through SDC4. Moreover, SDC4 expression correlated with poor overall survival in ovarian cancer patients. Our study highlights the potential role of T cells and NK cells in long-term survival patients with ovarian cancer, indicating SDC4 as a potential prognostic marker in ovarian cancer patients., (© 2021 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
6. Mitochondrial fission causes cisplatin resistance under hypoxic conditions via ROS in ovarian cancer cells.
- Author
-
Han Y, Kim B, Cho U, Park IS, Kim SI, Dhanasekaran DN, Tsang BK, and Song YS
- Subjects
- Antineoplastic Agents pharmacology, Apoptosis, Cell Proliferation, Female, Humans, Mitochondria drug effects, Mitochondria metabolism, Mitochondrial Proteins genetics, Mitochondrial Proteins metabolism, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Quinazolinones pharmacology, Reactive Oxygen Species metabolism, Signal Transduction, Tumor Cells, Cultured, Tumor Microenvironment, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Cisplatin pharmacology, Drug Resistance, Neoplasm drug effects, Hypoxia physiopathology, Mitochondria pathology, Mitochondrial Dynamics, Ovarian Neoplasms pathology
- Abstract
Mitochondria undergo fission and fusion continually for survival through the course of cellular adaption processes in response to changes in the surrounding environment. Dysregulated mitochondrial dynamics has been reported in various diseases including cancer. Under hypoxic conditions (<1% O
2 ), the relationship between mitochondrial dynamics and sensitivity to cisplatin (CDDP) was examined in ovarian cancer cells. We found that hypoxia promoted mitochondrial fission and CDDP resistance in ovarian cancer cells. Hypoxia-induced reactive oxygen species (ROS) caused an increase in mitochondrial fission, a response abolished by free radical scavenging with N-acetylcysteine (NAC) and Trolox. Also, treatment of hydrogen peroxide (H2 O2 ) decreased inhibitory p-Drp1 (Ser637) content and increased mitochondrial fission. Suppression of mitochondrial fission enhanced the CDDP sensitivity of hypoxic ovarian cancer cells. Lastly, in tumor spheroids from malignant ascites or tissues of patients with advanced-stage ovarian cancer, pretreatment with Mdivi-1 increased the CDDP sensitivity. Taken together, our results implicate that hypoxia-induced ROS trigger mitochondrial fission and CDDP resistance through downregulation of p-Drp1 (Ser637) and Mfn1 in ovarian cancer cells. Inhibition of Drp1 by Mdivi-1 treatment or si-Drp1 transfection increased CDDP sensitivity of ovarian cancer cells under hypoxia. Therefore, mitochondrial dynamics of cancer cells adapting to the hypoxic tumor microenvironment could be a potential target for anticancer therapy.- Published
- 2019
- Full Text
- View/download PDF
7. Development of Web-Based Nomograms to Predict Treatment Response and Prognosis of Epithelial Ovarian Cancer.
- Author
-
Kim SI, Song M, Hwangbo S, Lee S, Cho U, Kim JH, Lee M, Kim HS, Chung HH, Suh DS, Park T, and Song YS
- Subjects
- Adult, Aged, Cytoreduction Surgical Procedures, Disease-Free Survival, Female, Humans, Internet, Middle Aged, Platinum therapeutic use, Prognosis, ROC Curve, Retrospective Studies, Survival Rate, Tertiary Care Centers, Treatment Outcome, Carcinoma, Ovarian Epithelial pathology, Carcinoma, Ovarian Epithelial therapy, Nomograms, Ovarian Neoplasms pathology, Ovarian Neoplasms therapy
- Abstract
Purpose: Discovery of models predicting the exact prognosis of epithelial ovarian cancer (EOC) is necessary as the first step of implementation of individualized treatment. This study aimed to develop nomograms predicting treatment response and prognosis in EOC., Materials and Methods: We comprehensively reviewed medical records of 866 patients diagnosed with and treated for EOC at two tertiary institutional hospitals between 2007 and 2016. Patients' clinico-pathologic characteristics, details of primary treatment, intra-operative surgical findings, and survival outcomes were collected. To construct predictive nomograms for platinum sensitivity, 3-year progression-free survival (PFS), and 5-year overall survival (OS), we performed stepwise variable selection by measuring the area under the receiver operating characteristic curve (AUC) with leave-one-out cross-validation. For model validation, 10-fold cross-validation was applied., Results: The median length of observation was 42.4 months (interquartile range, 25.7 to 69.9 months), during which 441 patients (50.9%) experienced disease recurrence. The median value of PFS was 32.6 months and 3-year PFS rate was 47.8% while 5-year OS rate was 68.4%. The AUCs of the newly developed nomograms predicting platinum sensitivity, 3-year PFS, and 5-year OS were 0.758, 0.841, and 0.805, respectively. We also developed predictive nomograms confined to the patients who underwent primary debulking surgery. The AUCs for platinum sensitivity, 3-year PFS, and 5-year OS were 0.713, 0.839, and 0.803, respectively., Conclusion: We successfully developed nomograms predicting treatment response and prognosis of patients with EOC. These nomograms are expected to be useful in clinical practice and designing clinical trials.
- Published
- 2019
- Full Text
- View/download PDF
8. Pro-inflammatory M1 macrophage enhances metastatic potential of ovarian cancer cells through NF-κB activation.
- Author
-
Cho U, Kim B, Kim S, Han Y, and Song YS
- Subjects
- Cell Line, Tumor, Cell Movement genetics, Disease Progression, Female, Gene Expression Regulation genetics, Humans, Inflammation genetics, Inflammation metabolism, Macrophages metabolism, Neoplasm Invasiveness genetics, Neoplasm Invasiveness pathology, Obesity complications, Obesity metabolism, Obesity pathology, Ovarian Neoplasms genetics, Signal Transduction, Transcription, Genetic genetics, Tumor Microenvironment genetics, Tumor Necrosis Factor-alpha metabolism, Inflammation pathology, Macrophages pathology, NF-kappa B metabolism, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology
- Abstract
Obesity is a serious health problem and critically related to poor prognosis in cancer, presumably through induction of chronic inflammation. The major culprit for cancer progression in obesity is presumed to be macrophages. Accumulation of macrophages in adipose tissue due to obesity induced chronic inflammation has been observed. However, obesity-induced macrophage accumulation related to ovarian cancer progression remains unclear. So, the role of macrophage in cancer progression is needed to be further defined for therapeutic intervention. Here we determined the effect of macrophage type 1 (M1 macrophage) on ovarian cancer cells in relation to the metastasis. Ovarian cancer cell lines (PA-1, SKOV3) and monocyte-derived macrophages were used in this study. Treatment with M1 macrophage conditioned media on ovarian cancer cells increased the metastatic potential, such as migration and invasion capabilities. Interestingly, upon treatment with M1 macrophage conditioned media, nuclear translocation of NF-κB, p60, and p50, from the cytosol was enhanced together with increased transcriptional activity of the NF-κB. Pre-treatment with TPCK (NF-κB inhibitor) and NF-κB siRNA on ovarian cancer cells suppressed M1 macrophage-induced metastatic potential. Furthermore, Treatment of TNF-α on ovarian cancer cells showed NF-κB activation. Co-treatment with TNF-α inhibitor, etanercept, and M1 macrophage conditioned media on ovarian cancer cell lines reversed M1 macrophage conditioned media induced NF-κB activation. Taken together, TNF-α released from M1 macrophage increased metastatic potential in ovarian cancer cells through the activation of NF-κB signaling pathway. These results provide a new insight into the critical role of M1 macrophage in the tumor microenvironment in ovarian cancer., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.