Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Sci Rep ; 13(1): 8673, 2023 05 29.
Article in English | MEDLINE | ID: mdl-37248363

ABSTRACT

Radiotherapy benefits patients with advanced esophageal squamous cell carcinoma (ESCC) in terms of symptom relief and long-term survival. In contrast, a substantial proportion of ESCC patients have not benefited from radiotherapy. This study aimed to establish and validate an artificial neural network-based radiomics model for the pretreatment prediction of the radiotherapy response of advanced ESCC by using integrated data combined with feasible baseline characteristics of computed tomography. A total of 248 patients with advanced ESCC who underwent baseline CT and received radiotherapy were enrolled in this study and were analyzed by two types of radiomics models, machine learning and deep learning. As a result, the Att. Resnet50 pretrained network model indicated superior performance, with AUCs of 0.876, 0.802 and 0.732 in the training, internal validation, and external validation cohorts, respectively. Similarly, our Att. Resnet50 pretrained network model showed excellent calibration and significant clinical benefit according to the C index and decision curve analysis. Herein, a novel pretreatment radiomics model was established based on deep learning methods and could be used for radiotherapy response prediction in advanced ESCC patients, thus providing reliable evidence for therapeutic decision-making.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Radiation Oncology , Humans , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/radiotherapy , Esophageal Squamous Cell Carcinoma/diagnostic imaging , Esophageal Squamous Cell Carcinoma/radiotherapy , Area Under Curve , Neural Networks, Computer , Retrospective Studies
2.
J Transl Med ; 20(1): 626, 2022 12 28.
Article in English | MEDLINE | ID: mdl-36578050

ABSTRACT

BACKGROUND: Radiotherapy is one of the main treatments for esophageal squamous cell carcinoma (ESCC), but its efficacy is limited by radioresistance. MicroRNAs play a crucial role in posttranscriptional regulation, which is linked to the cancer response to radiation. METHODS: We successfully established a radioresistant cell line model by using fractionated irradiation. qRT-PCR was adopted to detect the expression of miR-4443 in human normal esophageal cell lines, tumor cells, and radioresistant cells. Next, CCK-8, colony formation, apoptosis, and cell cycle assays were used to assess the biological effect of miR-4443. Weighted gene coexpression network analysis (WGCNA) was performed to identify potential radiosensitivity-related genes. Additionally, we predicted the probable targets of the miRNA using bioinformatic methods and confirmed them using Western blot. RESULTS: miR-4443 was significantly upregulated in radioresistant ESCC cells. Enhancement of miR-4443 further decreased the radiosensitivity of ESCC cells, while inhibition of miR-4443 increased the radiosensitivity of ESCC cells. Notably, miR-4443 modulated radiosensitivity by influencing DNA damage repair, apoptosis, and G2 cycle arrest. By using WGCNA and experimental validation, we identified PTPRJ as a key target for miRNA-4443 to regulate radiosensitivity. The effects of miR-4443 overexpression or inhibition could be reversed by increasing or decreasing PTPRJ expression. CONCLUSION: In this study, miR-4443 is found to promote radiotherapy resistance in ESCC cells by regulating PTPRJ expression, which provides a new perspective and clue to alleviate radioresistance.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , MicroRNAs , Humans , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/radiotherapy , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/radiotherapy , Esophageal Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Apoptosis/genetics , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/radiation effects , Gene Expression Regulation, Neoplastic , Radiation Tolerance/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics
3.
Vaccines (Basel) ; 10(10)2022 Oct 15.
Article in English | MEDLINE | ID: mdl-36298586

ABSTRACT

BACKGROUND: Esophageal cancer (ESCA) is a major cause of cancer-related mortality worldwide. Altered fatty acid metabolism is a hallmark of cancer. However, studies on the roles of fatty acid metabolism-related genes (FRGs) in ESCA remain limited. METHOD: We identified differentially expressed FRGs (DE-FRGs). Then, the DE-FRGs prognostic model was constructed and validated using a comprehensive analysis. Moreover, the correlation between the risk model and clinical characteristics was investigated. A nomogram for predicting survival was established and evaluated. Subsequently, the difference in tumor microenvironment (TME) was compared between two risk groups. The sensitivity of key DE-FRGs to chemotherapeutic interventions and their correlation with immune cells were investigated. Finally, DEGs between two risk groups were measured and the prognostic value of key DE-FRGs in ESCA was confirmed in other databases. RESULTS: A prognostic model was constructed based on seven selected DEG-FRGs. TNM staging and CD8+ T cells were significantly correlated with high-risk groups. Low-risk groups exhibited more infiltrated M0 macrophages, an activation of type II interferon (IFN-γ) responses, and were found to be more suitable for immunotherapy. Seven key DE-FRGs with prognostic value were found to be considerably influenced by different chemotherapy drugs. CONCLUSION: A prognostic model based on seven DE-FRGs may efficiently predict patient prognosis and immunotherapy response, helping to develop individualized treatment strategies in ESCA.

4.
Sci Rep ; 12(1): 1960, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35121801

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is the main subtype of esophageal cancer. Since autophagy-related genes (ARGs) play a key role in the pathogenesis of many tumors, including ESCC, the purpose of this study is to establish an autophagy-related prognostic risk signature based on ARGs expression profile, and to provide a new method for improving prediction of clinical outcomes. We obtained the expression profiles of ESCC from public data (GSE53625) and extracted the portion of ARGs. Differential expression analysis and enrichment analysis were performed to confirm abnormal autophagy-related biological functions. Univariate and multivariate Cox regression analyses were performed on RNA microarray data (GSE53625) to construct a prognostic risk signature associated with autophagy. The performance of the model was evaluated by receiver operating characteristic (ROC) analysis, survival analysis and Brier score. The model was subjected to bootstrap internal validation. The potential molecular mechanism of gene signature was explored by gene set enrichment analysis (GSEA). Spearman correlation coefficient examined the correlation between risk score and immune status and ferroptosis. The expression levels of genes and proteins were validated by qRT-PCR and immunohistochemistry in ESCC cell lines and ESCC tissues. We constructed and validated an autophagy-related prognostic risk signature in 179 patients with ESCC. The long-term survival of patients in high-risk group was lower than that in low-risk group (log-rank, P value < 0.001). ROC analysis and Brier score confirmed the reliability of the signature. GSEA results showed significant enrichment of cancer- and autophagy-related signaling pathways in the high-risk ESCC patients and immunoregulatory signaling pathways in the low-risk ESCC patients. Correlation analysis showed that the risk signature can effectively predict the effect of immunotherapy. About 33.97% (71/209) ferroptosis-related genes were significantly correlated with risk scores. Finally, the results of qRT-PCR and immunohistochemistry experiments were consistent with bioinformatics analysis. In brief, we constructed a novel autophagy-related gene signature (VIM, UFM1, TSC2, SRC, MEFV, CTTN, CFTR and CDKN1A), which could improve the prediction of clinical outcomes in patients with ESCC.


Subject(s)
Autophagy-Related Proteins/genetics , Autophagy/genetics , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Gene Expression Profiling , Transcriptome , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/therapy , Humans , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests , Prognosis , Risk Assessment , Risk Factors , Time Factors
5.
Int J Gen Med ; 14: 8325-8339, 2021.
Article in English | MEDLINE | ID: mdl-34815705

ABSTRACT

BACKGROUND: Considering the significance of autophagy and long non-coding RNAs (lncRNAs) in the biology of esophageal squamous cell carcinoma (ESCC), the present study aimed to identify a new autophagy-related lncRNA signature to forecast the clinical outcomes of ESCC patients and to guide individualized treatment. METHODS: The expression profiles were obtained from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) database. We extracted autophagy-related genes from the Human Autophagy Database and identified autophagy-related lncRNAs through Spearman correlation analysis. Univariate, least absolute shrinkage and selection operator and multivariate Cox regression analyses were performed on GSE53625 to construct an autophagy-related lncRNAs prognostic signature. The model was subjected to bootstrap internal validation, and the expression levels of lncRNAs were verified by TCGA database. The potential molecular mechanism of the model was explored by gene set enrichment analysis (GSEA). Spearman correlation coefficient examined the correlation between risk score and ferroptosis-associated genes as well as the response to immunotherapy and chemotherapy. RESULTS: We identified and validated an autophagy-related lncRNAs prognostic signature in 179 patients with ESCC. The prognosis of patients in the low-risk group was significantly better than that in the high-risk group (p-value <0.001). The reliability of the model was verified by Brier score and ROC. GSEA results showed significant enrichment of cancer- and autophagy-related signaling pathways in the high-risk group and metabolism-related pathways in the low-risk group. Correlation analysis indicated that the model can effectively forecast the effect of immunotherapy and chemotherapy. About 35.41% (74/209) ferroptosis-related genes were significantly correlated with risk scores. CONCLUSION: In brief, we constructed a novel autophagy-related lncRNAs signature (LINC02024, LINC01711, LINC01419, LCAL1, FENDRR, ADAMTS9-AS1, AC025244.1, AC015908.6 and AC011997.1), which could improve the prediction of clinical outcomes and guide individualized treatment of ESCC patients.

6.
Int J Gen Med ; 14: 5845-5862, 2021.
Article in English | MEDLINE | ID: mdl-34566425

ABSTRACT

PURPOSE: Ferroptosis and long non-coding RNA (lncRNA) expression signatures have been associated with the clinical progression and immune-contexture of different solid tumors. The study aimed to identify a prognostic signature of ferroptosis-related lncRNAs (falncRNAs) to forecast the immune scenery and immunotherapy response in esophageal cancer (EC). PATIENTS AND METHODS: Gene expression profiles of EC were extracted from The Cancer Genome Atlas (TCGA) database, and ferroptosis-related genes were downloaded from the FerrDb database, which identified differentially expressed falncRNAs (DEfalncRNAs) via differential analysis. DEfalncRNA pairs associated with prognosis were identified by constructing a matrix, univariate and least absolute shrinkage and selection operator (LASSO) analysis. The prognostic signature was constructed by multivariate analysis. We appraised the forecasting capability of prognostic signature in survival, clinicopathological features, immune landscape, efficacy of immunotherapy, and drug sensitivity. The potential molecular mechanism of signature was investigated by gene set enrichment analysis (GSEA). RESULTS: We obtained 18 DEfalncRNA pairs to define a novel prognostic signature that was determined on a discovery cohort of 158 tumor samples and 11 adjacent normal tissues from TCGA and internally validated, with the definition of high- vs low-risk groups based on 3 years overall survival. We demonstrated that the high- vs low-risk groups differed for clinical parameters and computationally predicted drug sensitivity and tumor immune contexture, with the high-risk group having worse survival, more aggressive disease (node involvement, metastasis), reduced drug sensitivity, higher tumor mutation load, and gene signatures of infiltration of pro-tumoral immune cell subsets. The GSEA results revealed that ferroptosis and immunoregulatory pathways were significantly enriched in the high-risk group. CONCLUSION: The prognostic signature based on falncRNAs has the potential to forecast the survival, immune scenery, efficacy of immunotherapy, and drug sensitivity of EC, which is helpful for clinical prediction and individualized treatment.

7.
Cancer Manag Res ; 13: 4841-4852, 2021.
Article in English | MEDLINE | ID: mdl-34188537

ABSTRACT

PURPOSE: Resistance to radiotherapy results in a high treatment failure rate for locally advanced esophageal squamous cell carcinoma (ESCC). Ubiquitin-like with plant homeodomain and ring-finger domains 1 (UHRF1), is associated with poor prognosis in ESCC. The present study aims to characterize the effect of UHRF1 silencing on the radiosensitivity of ESCC and its potential mechanism. METHODS: Both in vitro and in vivo experiments were conducted to observe the effects of UHRF1 silencing on the radiosensitivity of ESCC. The effects of UHRF1 silencing on the apoptosis of ESCC cells were assessed by flow cytometry. The expression of apoptosis-related factors (caspase-3 and Bcl-2), PI3K/Akt/mTOR signaling pathway-related factors (PTEN, p-Akt and Akt, p-mTOR and mTOR), and DNMT1 were measured via Western blot, and the status of PTEN methylation was detected by methylation-specific PCR. Immunohistochemistry was used to detect the expressions of PTEN, p-AKT, and p-mTOR in xenograft tumor tissues. RESULTS: In vitro and in vivo experiments showed that UHRF1 knock-down inhibited ESCC cell growth and enhanced their radiosensitivity. shUHRF1 combined with radiation significantly increased ESCC cell apoptosis. Meanwhile, it activated the expression of caspase-3 and inhibited the expression of Bcl-2. shUHRF1 inhibited the expression of DNMT1 and reduced the methylation of PTEN, and then upregulated the expression of PTEN to inhibit the PI3K/Akt/mTOR signaling pathway. On the contrary, the PI3K/Akt/mTOR signaling pathway can be activated by upregulation of UHRF1. CONCLUSION: Our findings provide a theoretical basis for UHRF1 as a target to improve the radiosensitivity of ESCC.

8.
Bosn J Basic Med Sci ; 21(6): 760-772, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-33823125

ABSTRACT

This study aimed to build up nomogram models to evaluate overall survival (OS) and cancer-specific survival (CSS) in early-onset esophageal cancer (EOEC). Patients diagnosed with esophageal cancer (EC) from 2004 to 2015 were extracted from the Surveillance Epidemiology and End Results (SEER) database. Clinicopathological characteristics of younger versus older patients were compared, and survival analysis was performed in both groups. Independent related factors influencing the prognosis of EOEC were identified by univariate and multivariate Cox analysis, which were incorporated to construct a nomogram. The predictive capability of the nomogram was estimated by the concordance index (C-index), calibration plot, receiver operating characteristic (ROC) curve, and decision curve analysis (DCA). A total of 534 younger and 17,243 older patients were available from the SEER database. Younger patients were randomly segmented into a training set (n = 266) and a validation set (n = 268). In terms of the training set, the C-index of the OS nomogram was 0.740 (95% CI: 0.707-0.773), and that of the CSS nomogram was 0.752 (95% CI: 0.719-0.785). In view of the validation set, the C-index of OS and CSS were 0.706 (95% CI: 0.671-0.741) and 0.723 (95% CI: 0.690-0.756), respectively. Calibration curves demonstrated the consistent degree of fit between actual and predicted values in nomogram models. From the perspective of DCA, the nomogram models were more beneficial than the tumor-node-metastasis (TNM) and the SEER stage for EOEC. In brief, the nomogram model can be considered as an individualized quantitative tool to predict the prognosis of EOEC patients to assist clinicians in making treatment decisions.


Subject(s)
Esophageal Neoplasms/pathology , Nomograms , Adult , Age Factors , Aged , Aged, 80 and over , China/epidemiology , Esophageal Neoplasms/epidemiology , Female , Humans , Male , Middle Aged , Neoplasm Staging , Prognosis , Risk Factors , SEER Program , United States/epidemiology
9.
Cancer Cell Int ; 21(1): 165, 2021 Mar 12.
Article in English | MEDLINE | ID: mdl-33712005

ABSTRACT

BACKGROUND: Radiotherapy is a major treatment for esophageal squamous cell carcinoma (ESCC). However, HPV infection related radioresistance caused poor prognosis of ESCC. The function of SOCS6, which has been shown to be a tumor suppressor in several cancers, has not been fully investigated up till now. In this manuscript, we aim to further investigate the role of SOCS6 in regulating ESCC radioresistance. METHODS: Fifty-seven ESCC patients were enrolled for survival analysis. SOCS6 was stably overexpressed in HPV+ ESCC and ESCC cells, and cells were treated with radiation and then subjected to colony formation assays. Expression of DNA damage repair regulating proteins were examined by Western blotting. Cell growth, cell migration and cisplatin sensitivity were then analyzed. Sphere formation assays and flow cytometry were used to investigate changes in cancer stem cell (CSC) properties. Immunofluorescent staining and confocal microscopy were used to locate SOCS6 and c-Kit. Ubiquitylation level of c-Kit were analyzed after immunoprecipitation. Then, coimmunoprecipitation (CoIP) of SOCS6 and c-Kit were performed. In vivo, xenograft animal models were treated with radiation to examine the radiosensitivity. RESULTS: SOCS6 is correlated with better prognosis in ESCC patients. Radioresistance is impaired by SOCS6 upregulation, which inhibited cell growth, migration and increased sensitivity to cisplatin. SOCS6 significantly decreased the population of CSCs expressing the surface biomarker CD271 or CD24low/CD44high and their ability of sphere formation. SOCS6 and c-Kit were collocated in the cytoplasm. Blotting of ubiquitin and CoIP experiments indicated that the mechanism was related to ubiquitylation and degradation of the receptor c-Kit. Xenograft tumor mouse model showed that SOCS6 inhibited tumor growth and promoted radiosensitivity in vivo. CONCLUSIONS: Our findings suggest that SOCS6 can promote the radiosensitivity of HPV+ ESCC and ESCC cells and reduce their stemness via ubiquitylation and degradation of c-Kit. Thus, SOCS6 is a potential target for overcoming radioresistance of ESCC.

10.
Clin Chim Acta ; 512: 100-105, 2021 Jan.
Article in English | MEDLINE | ID: mdl-31465770

ABSTRACT

BACKGROUND: Ovarian epithelial cancer is the leading cause of deaths associated with gynecologic malignancies. Genistein represents a major type of phytoestrogens widely found in foods and herbal medicines. Although multiple epidemiological studies indicated that the consumption of genistein or other isoflavones is associated with a decreased ovarian cancer risk, the cellular effects and underlying mechanisms are not fully understood. This study focuses on the effect of genistein on the proliferation and cell cycle regulation of ovarian cancer cells. METHODS: Ovarian cancer OVCAR-5 cells were treated with genistein in an estrogen-free condition. Cell counting and MTS assays were performed to determine the cell proliferation alterations. Real-time PCR and Western blotting were conducted to examine the expression changes in key cell cycle regulators. RESULTS: Genistein significantly promoted the proliferation and the viability of OVCAR-5 cells. Upon genistein treatment, cellular mRNA and protein expression levels of PCNA, Cyclin D1 and CDK4 were increased, but those of p21 and p27 were decreased. CONCLUSION: In contrary to results of many previous studies, we observed that genistein was able to upregulate the proliferation and G1-S transition of ovarian cancer OVCAR-5 cells. The discrepancy could be caused by diverged experimental conditions and/or different ER expression patterns of cell lines. The findings may provide basic information for in-depth analysis on the role(s) and mechanisms by which genistein confers its effect on ovarian cancer progression.


Subject(s)
Cyclin D1 , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , Cell Line, Tumor , Cell Proliferation , Cyclin D1/genetics , Cyclin-Dependent Kinase 4 , Female , Genistein/pharmacology , Humans , Ovarian Neoplasms/drug therapy
11.
Future Oncol ; 15(2): 121-132, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30232909

ABSTRACT

AIM: We aimed to investigate the effect of current treatment based on stage and histology type, which were important factors for treating esophageal cancer. METHODS: Log-rank test, COX and nomograms were used for survival analysis. DCA, C-index and calibration curves were used for validation. RESULTS: A total of 3224 patients were recruited. As for cT2-T4aM0 patients, chemotherapy and radiation prolonged overall survival (OS) for esophageal squamous cell carcinoma (ESCC) and chemotherapy improved OS for esophageal adenocarcinoma (EAC). Meanwhile, neoadjuvant radiotherapy had longer OS than adjuvant radiotherapy for ESCC. As for T4b patients, radiation and chemotherapy correlated with better OS for ESCC and chemotherapy prolonged OS for EAC. CONCLUSION: Neoadjuvant radiotherapy might be optimal for cT2-T4aM0 ESCC. Radiation was recommended for T4b ESCC while chemotherapy was recommended for T4b EAC.


Subject(s)
Adenocarcinoma/therapy , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/therapy , Models, Biological , Nomograms , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Chemoradiotherapy, Adjuvant/methods , Cohort Studies , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Esophagectomy , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoadjuvant Therapy/methods , Neoplasm Staging , SEER Program/statistics & numerical data , Survival Analysis , Survival Rate , Treatment Outcome
12.
Biomed Pharmacother ; 108: 316-324, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30227324

ABSTRACT

BACKGROUND: The prognosis of esophageal squamous cell carcinoma (ESCC) is relatively poor due to the absence of efficient treatment. In this manuscript, we have investigated the specific roles and molecular mechanisms of LINC00657 to order to identify novel therapeutic targets for ESCC. METHOD: The LINC00657 expression in ESCC tissues and cell lines were evaluated by quantitative real-time PCR. The expression of LINC00657 in ESCC cells was regulated by lentivirus transfection. Online bioinformatics analysis tools were used to predict the potential targets of LINC00657 and miR-615-3p. TCGA database was used to analyze the prognosis of ESCC patients. Transwell, wound healing assay and MTT were performed to investigate the ESCC cells' biological functions. JunB expression was evaluated by Western blot. RESULT: LINC00657 was moderately increased in ESCC both in vivo and in vitro and up regulated by irradiation. LINC00657 knockdown could inhibit the migration and proliferation of ESCC cells. And downregulation of LINC00657 significantly enhanced the radio-sensitivity. Moreover, LINC00657 could act as a ceRNA to increase the expression of JunB by binding to miR-615-3p. Meanwhile, overexpression of miR-615-3p resulted in anti-tumor effects and led to the down-regulation of JunB. Survival analysis from TCGA indicated that ESCC patients with higher JunB expression had significant poorer prognosis. CONCLUSION: LINC00657 might be involved in regulating ESCC's response to radiation; and it functioned as an oncogene in ESCC by targeting miR-615-3p and JunB, providing novel potential therapeutic targets.


Subject(s)
Carcinogenesis/genetics , Esophageal Squamous Cell Carcinoma/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Transcription Factors/metabolism , Base Sequence , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Radiation Tolerance/genetics , Transcription Factors/genetics
13.
Biomed Pharmacother ; 102: 567-574, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29597090

ABSTRACT

Radiotherapy is one of the common treatments for esophageal squamous cell carcinoma (ESCC). Yet, local recurrence led by radioresistance is still not solved. Lobaplatin (LBP) is known to have powerful clinical anti-tumor activities in various tumors, but its effect in radiotherapy is rarely studied. Here we report that LBP is a promising radiosensitizer for ESCC. We treated ESCC cells with LBP and radiation, both separately and in combination. Untreated cells were set as control groups. We found that LBP inhibited ESCC cell growth and enhanced their radiosensitivity. LBP also impeded the tumor growth in vivo. LBP combined with radiation significantly increased ESCC cell apoptosis. Meanwhile, LBP obviously decreased the expression of cancer stem cells biomarker CD271 both in vitro and in vivo. The molecular mechanism was related to the downregulation of Bcl-2/Bax ratio, PI3K and p-AKT (Ser473) expression. Taken together, our findings indicated that LBP could enhance the radiosensitivity of ESCC cells by increasing radiation-induced apoptosis, attenuating cancer stemness and inhibiting PI3K/AKT pathway. These results provide a foundation for the combined therapy of radiation and LBP for ESCC in clinical practice.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/enzymology , Cyclobutanes/therapeutic use , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/enzymology , Neoplastic Stem Cells/pathology , Organoplatinum Compounds/therapeutic use , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Radiation Tolerance , Animals , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclobutanes/pharmacology , Down-Regulation/drug effects , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins , Organoplatinum Compounds/pharmacology , Radiation Tolerance/drug effects , Receptors, Nerve Growth Factor , Signal Transduction/drug effects , Up-Regulation/drug effects , bcl-2-Associated X Protein/metabolism
14.
Biomed Pharmacother ; 101: 406-413, 2018 May.
Article in English | MEDLINE | ID: mdl-29501762

ABSTRACT

Gastric cancer incidence is relatively higher in China than that in developed countries; however, molecular mechanisms considering the initiation and progression of gastric cancer are still unclear. For decades, numerous microRNAs have been found to regulate a wide range of biological functions in gastric cancer. However, the oncogenic function of miR-615-3p in gastric cancer has not been reported to date. With the help of gene and microRNA chips in 10 patients, we were able to screen differential expressed genes and microRNAs compared with normal gastric tissues. After that, online bioinformatics analysis tools were used to predict microRNAs' potential targets. As a result, miR-615-3p and its potential target, CELF2, were selected for further experiments. QRT-PCR and western blot results indicated the aberrant high expression of miR-615-3p and low expression of CELF2 in gastric cancer both in vivo and in vitro. Moreover, miR-615-3p expression correlated to T and M stage. Up regulation of miR-615-3p inhibited the apoptosis, promoted proliferation and migration and led to the down-regulation of CELF2. Meanwhile, down-regulation of miR-615-3p resulted in anti-tumor effects. Immunochemistry staining of CELF2 showed its association with T, N and M stage. In addition, overexpression of CELF2 could reverse miR-615-3p's oncogenic functions stated before. These findings indicate that miR-615-3p promotes gastric cancer proliferation and migration by suppressing CELF2 expression for the first time, providing clues for future clinical practices.


Subject(s)
Apoptosis/genetics , CELF Proteins/genetics , Cell Movement/genetics , Cell Proliferation/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Stomach Neoplasms/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Stomach Neoplasms/pathology , Up-Regulation/genetics
15.
Oncol Rep ; 39(4): 1601-1608, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29393484

ABSTRACT

Radiation therapy is one of the most important treatments for unresectable and locally advanced esophageal squamous cell carcinoma (ESCC), however, the response to radiotherapy is sometimes limited by the development of radioresistance. Sinomenine hydrochloride (SH) has anticancer activity, but its effect on the radiosensitivity of ESCC is unclear. We determined the effect of SH on the radiosensitivity of ESCC cells and elucidated its potential radiosensitization mechanisms in vitro and in vivo. ESCC cells were subjected to SH and radiation, both separately and in combination. Untreated cells served as controls. The CCK­8 assay was used to evaluate cell proliferation, and the clonogenic assay to estimate radiosensitization. Flow cytometry was used to investigate cell cycle phases and cell apoptosis. Bcl­2, Bax, cyclin B1, CDK1, Ku86, Ku70, and Rad51 expression was evaluated using western blotting. In vivo, tumor xenografts were created using BALB/c nude mice. Tumor­growth inhibition was recorded, and Ki­67 and Bax expression in the tumor tissues was assessed using immunohistochemistry. SH inhibited ESCC cell growth and markedly increased their radiosensitivity by inducing G2/M phase arrest. SH combined with radiation therapy significantly increased ESCC cell apoptosis. The molecular mechanism by which SH enhanced radiosensitivity of ESCC cells was related to Bcl­2, cyclin B1, CDK1, Ku86, Ku70, and Rad51 downregulation and Bax protein expression upregulation. SH combined with radiation considerably delayed the growth of tumor xenografts in vivo. Immunohistochemical analysis showed that in the SH combined with radiation group, the expression of Bax was significantly higher while that of Ki­67 was lower than the expressions in the control groups. Taken together, our findings showed that SH could improve the sensitivity of radiation in ESCC cells by inducing G2/M phase arrest, promoting radiation­induced apoptosis and inhibiting DSB­repair pathways. SH appears to be a prospective radiosensitizer for improving the efficacy of radiotherapy for ESCC.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/radiotherapy , Morphinans/administration & dosage , Radiation Tolerance/drug effects , Animals , Apoptosis/drug effects , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasm Proteins/genetics , Xenograft Model Antitumor Assays
16.
Mol Carcinog ; 56(12): 2610-2619, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28731203

ABSTRACT

PTEN pseudogene (PTENP1) has a tumor suppressive role in multiple cancers. However, its involvement in esophageal squamous cell carcinoma (ESCC) remains largely unknown. In this study, we set out to identify the role of PTENP1 in the development of ESCC. Gene Expression Omnibus database was employed to investigate the expression of PTENP1 in ESCC. sRNA target Database (StarBase v2.0) was used to query the downstream of PTENP1. Next, both in vitro and in vivo experiments were employed to explore the function. Cell proliferation was evaluated by CCK-8, soft agar, and colony formation assays. Expression of relative genes was assessed by quantitative real-time PCR (qRT-PCR) and Western blotting. 3'UTR luciferase assay was used to confirm the miRNA binding. The clinical significance of PTENP1 was further validated by immunohistochemistry (IHC) and correlation with clinicopathological indicators in additional samples (n = 93). We found expression of PTENP1 in ESCC was lower than that in the corresponding adjacent normal tissues (n = 17). Overexpression of PTENP1 in Eca109 and TE-1 cells resulted in inhibited proliferation and altered expression of SOCS6-p-STAT3-HIF-1α pathway both in vitro and in vivo. Subsequent IHC reported a similar trend in human ESCC samples. 3'UTR luciferase assay demonstrated that PTENP1 3'UTR decoyed miR-17-5p from binding to SOCS6. Moreover, PTENP1 expression was correlated with clinicopathological indicators to varying degrees, including histological grade, TNM stage, infiltration depth, lymph node metastasis, and overall survival. Taken together, these results suggested an anti-oncogenic role of PTENP1. Meanwhile, PTENP1 may also serve as a candidate of prognostic indicator for ESCC patients.


Subject(s)
Carcinoma, Squamous Cell/genetics , Esophageal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , PTEN Phosphohydrolase/genetics , Pseudogenes , Suppressor of Cytokine Signaling Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , HEK293 Cells , Humans , Kaplan-Meier Estimate , Male , Mice, Nude , MicroRNAs/genetics , Middle Aged , PTEN Phosphohydrolase/metabolism , RNA Interference , Suppressor of Cytokine Signaling Proteins/metabolism , Transplantation, Heterologous
17.
Oncotarget ; 7(35): 57050-57065, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27489353

ABSTRACT

High-risk human papillomavirus (HPV), especially HPV16, correlates with cancerogenesis of human esophageal squamous cell carcinoma (ESCC) and we have reported that HPV16 related with a poor prognosis of ESCC patients in China. We aim to investigate the potential role and mechanism of HPV16 in ESCC development and progress. Our following researches demonstrated that ESCC cells which were stably transfected by HPV16 E6-E7 lentiviral vector showed a remarkable cancer stem-like cells (CSCs) phenotype, such as: migration, invasion, spherogenesis, high expression of CSCs marker in ESCC---p75NTR, chemoresistance, radioresistance, anti-apoptosis ability in vitro and cancerogenesis in vivo. HPV16 E6-E7 induced PI3K/Akt signaling pathway activation and this affect could be effectively inhibited by LY294002, a specific PI3K inhibitor. It was also indicated that the inhibition of PI3K/Akt signaling pathway by PI3K and Akt siRNA reverse the effect which induced by HPV16 E6-E7 in ESCC cells. Taken together, the present study demonstrates that HPV16 E6-E7 promotes CSCs phenotype in ESCC cells through the activation of PI3K/Akt signaling pathway. Targeting the PI3K/Akt signaling pathway in HPV16 positive tissues is an available therapeutic for ESCC patients.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Neoplastic Stem Cells/cytology , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/metabolism , Animals , Carcinoma, Squamous Cell/virology , Cell Cycle , Cell Movement , Cell Survival , Chromones/chemistry , Enzyme Activation , Esophageal Neoplasms/virology , Esophageal Squamous Cell Carcinoma , Female , Humans , Lentivirus/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Morpholines/chemistry , Neoplasm Invasiveness , Neoplasm Transplantation , Nerve Tissue Proteins/genetics , Phenotype , Receptors, Nerve Growth Factor/genetics , Signal Transduction
18.
Onco Targets Ther ; 8: 573-81, 2015.
Article in English | MEDLINE | ID: mdl-25784817

ABSTRACT

BACKGROUND: Previous studies indicate that human papillomavirus 16 (HPV16) infection plays a pivotal role in the etiology of esophageal squamous cell carcinoma (ESCC). We aim to detect the influence of HPV16 infection on ESCC patient prognosis. PATIENTS AND METHODS: Immunohistochemical staining for HPV16 E6 oncoprotein, the low-affinity p75 neurotrophin receptor (p75NTR), and phosphatidylinositol 3-kinase (PI3K) was performed on 103 archived surgical specimens from patients with ESCC and 54 control samples from patients with benign esophageal tumor or inflammatory lesions. All patients were from the Shaan Xi Province, People's Republic of China. RESULTS: HPV16 E6 expression was significantly higher in the ESCC group (P<0.05). HPV16 E6 expression was significantly higher in men than in women (P<0.05). p75NTR expression was higher in those aged >56 years (P<0.05). PI3K expression was higher in those with a more advanced histopathological grade (P<0.05). There was a positive correlation between HPV16 E6 and p75NTR expression (r=0.547, P<0.001) and between p75NTR and PI3K expression (r=0.364, P<0.001). In 100 evaluable patients, the 5-year overall survival (OS) rate was 11%. In patients with ESCC, HPV16 E6 and PI3K expression were negatively correlated with the 3-year OS (P<0.05), 5-year OS (P<0.05), and progression-free survival (P<0.05). CONCLUSION: HPV16 infection likely contributes to the etiology of ESCC patients in Shaan Xi, People's Republic of China. HPV16 infection status and PI3K expression levels could be useful for predicting prognosis in patients with ESCC.

SELECTION OF CITATIONS
SEARCH DETAIL
...