Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunopharmacol ; 137: 112401, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38878485

ABSTRACT

OBJECTIVE: This study aimed to investigate the role of JMJD2A in radiotherapy tolerance of esophageal squamous cell carcinoma (ESCC). METHODS: The levels of H3K9me3 modification were analyzed in anti-PD-1 therapy non-responder or responder patients, and the expression differences of H3K9me3-related modifying enzymes were assessed in TCGA-ESCC and ICGC cohorts. Subsequently, JMJD2A was knocked down in ESCC cells using CRISPR-Cas9 or lentivirus-mediated shRNA, and changes in malignant behavior of ESCC cells were observed. RNA-seq, ATAC-seq, and ChIP-seq analyses were then conducted to investigate the genes and downstream signaling pathways regulated by JMJD2A, and functional validation experiments were performed to analyze the role of downstream regulated genes and pathways in ESCC malignant behavior and immune evasion. RESULTS: JMJD2A was significantly overexpressed in ESCC and anti-PD-1 therapy non-responders. Knockdown or deletion of JMJD2A significantly promoted the malignant behavior and immune evasion of ESCC. JMJD2A facilitated the structural changes in chromatin and promoted the binding of SMARCA4 to super-enhancers, thereby inducing the expression of GPX4. This resulted in the inhibition of radiation-induced DNA damage and cell ferroptosis, ultimately promoting the malignant behavior and immune evasion of ESCC cells. CONCLUSION: JMJD2A plays an indispensable role in the malignant behavior and immune evasion of ESCC. It regulates the binding of SMARCA4 to super-enhancers and affects the chromatin's epigenetic landscape, thereby promoting the expression of GPX4 and attenuating iron-mediated cell death caused by radiotherapy. Consequently, it triggers the malignant behavior and immune evasion of ESCC cells.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Neoplastic , Jumonji Domain-Containing Histone Demethylases , Humans , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Cell Line, Tumor , Radiation Tolerance/genetics , Immune Evasion , Tumor Escape , Ferroptosis/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Histones/metabolism , DNA Damage , DNA Helicases/genetics , DNA Helicases/metabolism
2.
Environ Toxicol ; 39(2): 592-611, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37493251

ABSTRACT

Esophageal cancer (EC) is a common digestive malignancy that ranks sixth in cancer deaths, with a 5-year survival rate of 15%-25%. As a result, reliable prognostic biomarkers are required to accurately predict the prognosis of EC. T-cell exhaustion (TEX) is associated with poorer prognosis and immune infiltration in EC. In this study, nine risk genes were finally screened to constitute the prognostic model using least absolute shrinkage and selection operator analysis. Patients were divided into two groups based on the expression of the TEX-related genes: high-risk group and low-risk group. The expression of TEX-related genes differed significantly between the two groups. The findings revealed that the risk model developed was highly related to the clinical prognosis and amount of immune cell infiltration in EC patients. It was also significantly correlated with the therapeutic sensitivity of multiple chemotherapeutic agents in EC patients. Subsequently, we successfully constructed drug-resistant cell lines KYSE480/CDDP-R and KYSE180/CDDP-R to verify the correlation between PD-1 and drug resistance in EC. Then, we examined the mRNA and protein expression levels of PD-1 in parental and drug-resistant cells using qPCR and WB. It was found that the expression level of PD-1 was significantly increased in the plasma red of drug-resistant cells. Next, we knocked down PD-1 in drug-resistant cells and found that the resistance of EC cells to CDDP was significantly reduced. And the proportion of apoptotic cells in cells treated with 6 µM CDDP for 24 h was significantly in increase. The TEX-based risk model achieved good prediction results for prognosis prediction in EC patients. And it was also significantly associated with the level of immune cell infiltration and drug therapy sensitivity of EC patients. Additionally, the downregulation of PD-1 may be associated with increased drug sensitivity in EC and enhanced T-cell infiltration. The high-risk group had lower TIDE scores, indicating that the high-risk group benefits more after receiving immunotherapy. Thus, the TEX-based risk model can be used as a novel tumor prognostic biomarker.


Subject(s)
Esophageal Neoplasms , Tumor Microenvironment , Humans , Prognosis , Programmed Cell Death 1 Receptor , T-Cell Exhaustion , Esophageal Neoplasms/genetics , Esophageal Neoplasms/therapy , Immunotherapy , Algorithms
4.
Cancer Med ; 12(2): 2089-2103, 2023 01.
Article in English | MEDLINE | ID: mdl-35771026

ABSTRACT

Esophageal cancer (EC) is an aggressive malignancy that accounts for numerous cancer-related deaths worldwide. The multimodal combination therapy approach can be potentially used to treat EC effectively. However, distinct biomarker of significant specificity are still needed to develop individualized treatment strategies and provide accurate prognostic predictions. Therefore, we aimed to investigate the associated genes subtypes identified were, IFN-γDominant, Inflammatory, Lymphocyte Depleted, etc. and construct a risk model based on these genes to predict the overall survival (OS) of patients suffering from EC. Three immune subtypes were defined in the Cancer Genome Atlas (TCGA) cohort with different tumor microenvironment (TME) and clinical outcomes based on radio-differentiated immune genes. Subsequently, a risk model of immune characteristics included the immune cell infiltration levels and pathway activity was developed based on the genomic changes between the subtypes. In the TCGA dataset, as well as in subgroup analysis with different stages, gender, age, and pathological type, a high-risk score was identified as an adverse factor for OS using the method of the univariate Cox regression analysis and tROC analysis. Furthermore, it was observed that the high-risk group was characterized by depleted immunophenotype, active cell metabolism, and a high tumor mutation burden (TMB). The low-risk group was characterized by high TME abundance and active immune function. Differences in the biological genotypes may account for the differences in the prognosis and treatment response. Extensive research was carried out, and the results revealed that the low-risk group exhibited a significant level of therapeutic advantage in the field of immunotherapy. A risk model was developed based on the immune characteristics. It can be used to optimize risk stratification for patients suffering from EC. The results can potentially help provide new perspectives on treatment methods.


Subject(s)
Esophageal Neoplasms , Humans , Prognosis , Esophageal Neoplasms/genetics , Esophageal Neoplasms/therapy , Computational Biology , Genomics , Immunotherapy , Tumor Microenvironment/genetics
5.
Biochem Cell Biol ; 99(6): 707-716, 2021 12.
Article in English | MEDLINE | ID: mdl-34723651

ABSTRACT

This study aimed to explore the effect of ultrasound-stimulated microbubbles (USMBs) on tumor radiosensitivity in esophageal carcinoma (EC). The human EC cell line KYSE-510 and human umbilical vein endothelial cells (HUVECs) were exposed to radiation alone or in combination with USMBs. CCK-8, colony formation, and EdU assays were used to determine cell viability and proliferation. Cell apoptosis was assessed using flow cytometry. Cell migration and invasion were examined by wound healing and transwell assays. Western blotting showed that the protein levels were associated with apoptosis, epithelial-mesenchymal transition (EMT), and angiogenesis. An endothelial tube-forming assay was used to detect the angiogenic activity of HUVECs. Xenograft experiments were used to examine the effect of USMBs on EC radiosensitivity in vivo. The expression of Ki-67 in tumors was detected using immunohistochemistry. USMBs enhanced the suppressive effect of radiation on proliferation, migration, invasion, and EMT, and promoted radiation-induced apoptosis in EC cells in vitro. Angiogenesis in EC was suppressed by radiation and further inhibited by the combination of radiation and USMBs. In vivo experiments revealed that USMBs increased the radiosensitivity of ECs to tumor growth. Collectively, USMBs enhanced the effects of radiotherapy in esophageal carcinoma.


Subject(s)
Carcinoma , Esophageal Neoplasms , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/radiotherapy , Human Umbilical Vein Endothelial Cells , Humans , Microbubbles
6.
Bioengineered ; 12(1): 3000-3013, 2021 12.
Article in English | MEDLINE | ID: mdl-34180353

ABSTRACT

Ultrasound (US) is reported to improve the delivery efficiency of drugs loading onto large nanoparticles due to the sonoporation effect. Microbubbles (MBs) can be used as contrast agents of US expanding and contracting under low-amplitude US pressure waves. Ultrasound-stimulated microbubbles (USMBs) therapy is a promising option for the treatment of various cancers as a radiosensitizer. However, its role in esophageal squamous cell carcinoma (ESCC) remains unknown. In our study, human ESCC cell lines (KYSE-410, KYSE-1140) were treated with radiation solely, US alone, or radiation in combination with US or USMBs. The migration and invasion abilities of ESCC cells were examined by wound healing and Transwell assays. ESCC cell apoptosis was assessed using flow cytometry analysis and TUNEL assays. The levels of proteins associated with cell apoptosis and angiogenesis were measured by western blot analysis. A tube formation assay was performed to detect the ESCC cell angiogenesis. We found that USMBs at high levels most effectively most efficiently enhanced the effect of radiation, and significant changes in the viability (48%-51%), proliferation (1%), migration (63%-71%), invasion (52%) and cell apoptosis (31%-50%) of ESCC cells were observed compared with the control group in vitro. The ESCC angiogenesis was inhibited by US or radiation treatment and further inhibited by a combination of radiation and US or USMBs. USMBs at high levels most effectively enhanced the inhibitory effect of radiotherapy on ESCC cell apoptosis. Overall, USMBs enhanced the radiosensitivity of esophageal squamous cell carcinoma cells.Graphical abstractUSMBs treatment enhanced the anti-tumor effect of radiation on ESCC cell proliferation, migration, invasion, angiogenesis and apoptosis in vitro.1USMBs enhance the radiation-induced inhibition on ESCC cell growth2USMBs promote the radiation effect on ESCC cell apoptosis3USMBs enhance radiation-caused suppression on ESCC cell migration and invasion4USMBs enhance the suppression of radiation on ESCC angiogenesis[Figure: see text].


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Microbubbles , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Humans , Radiation-Sensitizing Agents/chemistry , Ultrasonic Waves
7.
Onco Targets Ther ; 13: 3319-3324, 2020.
Article in English | MEDLINE | ID: mdl-32368090

ABSTRACT

BACKGROUND: Penile squamous cell carcinoma (PSCC) bears poor prognosis due to its rarity and limited treatment options, especially after failure of standard treatments. Effective therapeutic options were desperately needed. CASE PRESENTATION: We report a recurrent metastatic PSCC patient with positive programmed death ligand 1 (PD-L1) expression (≥10%) and tumor mutation burden (TMB) of 8.87 (Muts/Mb) who obtained significant response to immunotherapy, with progression-free survival (PFS) exceeding 10 months. CONCLUSION: This is the first case presenting remarkable response to immunotherapy in a Chinese PSCC patient. The remarkable response might be associated with PD-L1 expression, indicating that PD-L1 expression could be a promising biomarker for immunotherapy in PSCC. TMB ranking may also contribute to patient selection. However, large clinical trials are needed to validate these notions.

8.
Endocr Relat Cancer ; 17(1): 39-50, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19776289

ABSTRACT

Glucocorticoids (GCs) are widely used as co-medication in the therapy of solid malignant tumors to relieve some of the side effects of chemotherapeutic drugs. However, recent studies have shown that GCs could render cancer cells more resistant to cytotoxic drug-induced apoptosis, but the mechanism is largely unknown. In the present study, we found that the treatment of human ovarian cancer cell lines HO-8910 and SKOV3 with synthetic GCs dexamethasone (Dex) significantly increased their adhesion to extracellular matrix (ECM) and their resistance to apoptosis induced by cytotoxic drugs cisplatin and paclitaxel. Dex also increased the protein levels of adhesion molecules integrins beta1, alpha 4, and alpha 5 in HO-8910 cells. The neutralizing antibody against integrin beta1 prevented Dex-induced adhesion and significantly abrogated the protective effect of Dex toward cytotoxic agents. We further found that transforming growth factor-beta1 (TGF-beta1) alone not only increased cell adhesion and cell survival of HO-8910 cells in the presence of cisplatin, but also had synergistic pro-adhesion and pro-survival effects with Dex. Moreover, TGF-beta1-neutralizing antibody that could block TGF-beta1-induced cell adhesion and apoptosis resistance markedly abrogated the synergistic pro-adhesion and pro-survival effects of Dex and TGF-beta1. Finally, we further demonstrated that Dex could up-regulate the expression of TGF-beta receptor type II and enhance the responsiveness of cells to TGF-beta1. In conclusion, our results indicate that increased adhesion to ECM through the enhancement of integrin beta1 signaling and TGF-beta1 signaling plays an important role in chemoresistance induced by GCs in ovarian cancer cells.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma/pathology , Cell Adhesion/drug effects , Cisplatin/pharmacology , Dexamethasone/pharmacology , Drug Resistance, Neoplasm/drug effects , Extracellular Matrix/metabolism , Integrin beta1/physiology , Ovarian Neoplasms/pathology , Paclitaxel/pharmacology , Transforming Growth Factor beta1/physiology , Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Carcinoma/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Integrin alpha4beta1/biosynthesis , Integrin alpha4beta1/genetics , Integrin alpha5beta1/biosynthesis , Integrin alpha5beta1/genetics , Integrin beta1/immunology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Neoplasm Proteins/physiology , Ovarian Neoplasms/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/immunology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/drug effects
9.
J Steroid Biochem Mol Biol ; 101(4-5): 179-87, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17046241

ABSTRACT

Although there is ample evidence that glucocorticoids (GCs) have an antiproliferative effect on many cell types, the molecular mechanism remains elusive. We reported in our previous study that Dex treatment led to cell growth arrest in a human ovarian cancer cell HO-8910. RhoB, as a member of Rho GTPases, have been implicated to be a negative regulator of cell proliferation. In this study, we provided novel evidence that Dex induced the expressions of small GTPase RhoB mRNA and protein, but not RhoA and RhoC mRNA in a dose- and time-dependent fashion via glucocorticoid receptor (GR). Over-expression of RhoB increased while inhibition of RhoB expression by RNA interference reversed Dex-induced growth arrest, indicating that RhoB signaling is involved in Dex-induced proliferation inhibition. We also presented the novel observation that over-expression or activation of RhoB signaling elevated the basal transcriptional activity of the transcription factor NF-kappaB in HO-8910 cells. Furthermore, elevating RhoB signaling enhanced the inhibitory effect of Dex on NF-kappaB activity, while attenuating RhoB signaling almost abrogated Dex suppression of NF-kappaB signaling, indicating that RhoB pathway is involved in the regulation of NF-kappaB activity and is essential for Dex transcriptional repression on NF-kappaB signaling in HO-8910 cells.


Subject(s)
Dexamethasone/pharmacology , NF-kappa B/genetics , Transcription, Genetic , Up-Regulation , rhoB GTP-Binding Protein/metabolism , Cell Line, Tumor , Cell Proliferation , Glucocorticoids/pharmacology , Humans , NF-kappa B/physiology , Promoter Regions, Genetic , Receptors, Glucocorticoid/physiology , Signal Transduction , Transfection , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , rhoB GTP-Binding Protein/genetics , rhoC GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL