151
|
Yuan J, Jiang Q, Gong T, Fan D, Zhang J, Chen F, Zhu X, Wang X, Qiao Y, Chen H, Liu Z, Su J. Loss of grand histone H3 lysine 27 trimethylation domains mediated transcriptional activation in esophageal squamous cell carcinoma. NPJ Genom Med 2021; 6:65. [PMID: 34381055 PMCID: PMC8358006 DOI: 10.1038/s41525-021-00232-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 07/23/2021] [Indexed: 01/23/2023] Open
Abstract
Trimethylation of histone H3 lysine 27 trimethylation (H3K27me3) may be recruited by repressive Polycomb complexes to mediate gene silencing, which is critical for maintaining embryonic stem cell pluripotency and differentiation. However, the roles of aberrant H3K27me3 patterns in tumorigenesis are not fully understood. Here, we discovered that grand silencer domains (breadth > 50 kb) for H3K27me3 were significantly associated with epithelial cell differentiation and exhibited high gene essentiality and conservation in human esophageal epithelial cells. These grand H3K27me3 domains exhibited high modification signals involved in gene silencing, and preferentially occupied the entirety of topologically associating domains and interact with each other. We found that widespread loss of the grand H3K27me3 domains in of esophageal squamous cell carcinomas (ESCCs) were enriched in genes involved in epithelium and endothelium differentiation, which were significantly associated with overexpression with increase of active modifications of H3K4me3, H3K4me1, and H3K27ac marks, as well as DNA hypermethylation in the gene bodies. A total of 208 activated genes with loss of grand H3K27me3 domains in ESCC were identified, where the higher expression and mutation of T-box transcription factor 20 (TBX20) were associated with worse patients’ outcomes. Our results showed that knockdown of TBX20 may have led to a striking defect in esophageal cancer cell growth and carcinogenesis-related pathway, including cell cycle and homologous recombination. Together, our results reveal that loss of grand H3K27me3 domains represent a catalog of remarkable activating regulators involved in carcinogenesis.
Collapse
Affiliation(s)
- Jian Yuan
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Qi Jiang
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | | | - Dandan Fan
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Ji Zhang
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Fukun Chen
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Xiaolin Zhu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinyu Wang
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Yunbo Qiao
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, China
| | | | | | - Jianzhong Su
- School of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China. .,Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China. .,Oujiang Laboratory, Wenzhou, China.
| |
Collapse
|
152
|
Gao Y, Zhu Y, Xu X, Wang F, Shen W, Leng X, Zhao J, Liu B, Wang Y, Liu P. Surface PEGylated Cancer Cell Membrane-Coated Nanoparticles for Codelivery of Curcumin and Doxorubicin for the Treatment of Multidrug Resistant Esophageal Carcinoma. Front Cell Dev Biol 2021; 9:688070. [PMID: 34386493 PMCID: PMC8353447 DOI: 10.3389/fcell.2021.688070] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/23/2021] [Indexed: 12/11/2022] Open
Abstract
Objective The emergence of multi-drug resistance (MDR) in esophageal carcinoma has severely affected the effect of chemotherapy and shortened the survival of patients. To this end, we intend to develop a biomimetic nano-targeting drug modified by cancer cell membrane, and investigate its therapeutic effect. Methods The degradable poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) co-loaded with doxorubicin (DOX) and curcumin (Cur) were prepared by solvent evaporation method. TE10 cell membrane and Distearoyl phosphatidylethanolamine-polyethylene glycol (DSPE-PEG) were then coated on the PLGA NPs by membrane extrusion to prepare the PEG-TE10@PLGA@DOX-Cur NPs (PMPNs). Size and zeta potential of the PMPNs were analyzed by lazer particle analyzer, and the morphology of PMPNs was observed by transmission electron microscope. The TE10 cell membrane protein on PMPNs was analyzed by gel electrophoresis. The DOX-resistant esophageal cancer cell model TE10/DOX was established through high-dose induction. The In vitro homologous targeting ability of PMPNs was evaluated by cell uptake assay, and the in vitro anti-tumor effect of PMPNs was assessed through CCK-8, clone formation and flow cytometry. A Balb/c mouse model of TE10/DOX xenograft was constructed to evaluate the anti-tumor effect in vivo and the bio-safety of PMPNs. Results The prepared cell membrane coated PMPNs had a regular spherical structure with an average diameter of 177 nm. PMPNs could directly target TE10 and TE10/DOX cells or TE10/DOX xenografted tumor and effectively inhibit the growth of DOX-resistant esophageal carcinoma. Besides, the PMPNs was confirmed to have high biosafety. Conclusion In this study, a targeted biomimetic nano-drug delivery system PMPNs was successfully prepared, which overcome the MDR of esophageal carcinoma by co-delivering DOX and sensitizer curcumin.
Collapse
Affiliation(s)
- Yi Gao
- Department of Gastroenterology, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Jiangyin, China
| | - Yue Zhu
- Department of Gastroenterology, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Jiangyin, China
| | - Xiaopeng Xu
- Department of Gastroenterology, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Jiangyin, China
| | - Fangjun Wang
- Department of Gastroenterology, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Jiangyin, China
| | - Weidong Shen
- Department of Gastroenterology, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Jiangyin, China
| | - Xia Leng
- Department of Gastroenterology, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Jiangyin, China
| | - Jiyi Zhao
- Department of Gastroenterology, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Jiangyin, China
| | - Bingtuan Liu
- Department of Gastroenterology, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Jiangyin, China
| | - Yangyun Wang
- State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou, China
| | - Pengfei Liu
- Department of Gastroenterology, The Affiliated Jiangyin Hospital of Xuzhou Medical University, Jiangyin, China
| |
Collapse
|
153
|
Bastami M, Mahmoodzadeh H, Saadatian Z, Daraei A, Zununi Vahed S, Mansoori Y, Nariman-Saleh-Fam Z. Perturbation of miR-146b and relevant inflammatory elements in esophageal carcinoma patients supports an immune downregulatory mechanism. Pathol Res Pract 2021; 225:153560. [PMID: 34311393 DOI: 10.1016/j.prp.2021.153560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/10/2021] [Accepted: 07/17/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Esophageal Cancer is known as one of the deadliest cancers worldwide with the squamous cell carcinoma (ESCC) being the predominant subtype. There is a growing body of evidence linking the dysregulated microRNA (miRNA) pathway of immune cells to the progression of several tumors. In a previous study, we investigated molecular alterations pertaining to miR-146a and some components of NF-kB signaling pathway and proposed a possible immune downregulatory mechanism in peripheral blood mononuclear cells (PBMCs) of ESCC patients. Here, we further scrutinized other components of this pathway by evaluating PBMC levels of miR-146b, TLR4, IL10, and TNFA. METHODS Gene expressions were quantified using RT-qPCR assays. To prevent the vulnerability of results to the expression instability of reference genes, nine additional transcripts were quantified, and stable reference genes for normalizing qPCR data were identified using the NormFinder and the geNorm algorithms. The efficiency-corrected normalized relative quantity values were used to compare gene expressions among study groups. RESULTS The PBMC expression of miR-146b and TNFA was downregulated in ESCC patients as compared to healthy subjects. While the level of TLR4 was not different among the study groups, the PBMC level of IL10 was upregulated in ESCC patients. Logistic regression analyses coupled with the ROC curve and cross-validation analysis suggested that PBMC expression may serve as potential candidate biomarker for discriminating ESCC patients from healthy subjects. CONCLUSION The present findings, in line with our previous report, propose a particular gene expression pattern in PBMCs of ESCC patients, providing evidence in support of an immune downregulatory mechanism.
Collapse
Affiliation(s)
- Milad Bastami
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | | | - Zahra Saadatian
- Department of Physiology, Faculty of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Abdolreza Daraei
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | | | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.
| | - Ziba Nariman-Saleh-Fam
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
154
|
Yang CS, Chen XL. Research on esophageal cancer: With personal perspectives from studies in China and Kenya. Int J Cancer 2021; 149:264-276. [PMID: 33270917 PMCID: PMC8141013 DOI: 10.1002/ijc.33421] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
The most common form of esophageal cancer (EC), esophageal squamous cell carcinoma (ESCC), is prevalent in many unindustrialized societies, among people with lower socioeconomic status and those who frequently use tobacco and alcohol. In some areas, ESCC mortality ranked top among all cancer. In this review, we begin with discussions of the extensive research on EC in Linxian in northern China that started 60 years ago and the recent studies in Kenya from our personal perspectives. Based on the results obtained from these studies and information from the literature, we summarize our current understanding about the risk factors for ESCC including lifestyle factors (smoking, alcohol, consumption of food and beverages at high temperature and other unhealthy habits), poor diet and nutritional insufficiencies and genetic susceptibility. Elimination or minimization of these environmental risk factors, as well as early detection and treatment of precancerous lesions, would be effective means for the prevention of ESCC. Current knowledge of molecular alterations in ESCC (gene mutations, hypermethylation and amplification or overexpression), as well as treatment of ESCC and the potential of targeted therapy, are also discussed. Finally, we propose effective approaches for the prevention of ESCC by adapting a healthy lifestyle, including a healthy diet that would also prevent other diseases. Community outreach, public education and international collaboration are important for achieving this public health goal.
Collapse
Affiliation(s)
- Chung S. Yang
- Department of Chemical BiologyErnest Mario School of Pharmacy, Rutgers, The State University of New JerseyPiscatawayNew JerseyUSA
| | - Xiaoxin Luke Chen
- Cancer Research Program, Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central UniversityDurhamNorth CarolinaUSA
| |
Collapse
|
155
|
Wang L, Wang X, Yan P, Liu Y, Jiang X. LINC00261 Suppresses Cisplatin Resistance of Esophageal Squamous Cell Carcinoma Through miR-545-3p/MT1M Axis. Front Cell Dev Biol 2021; 9:687788. [PMID: 34336838 PMCID: PMC8320661 DOI: 10.3389/fcell.2021.687788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
To improve the survival rate and cure rate of patients, it is necessary to find a new treatment scheme according to the molecular composition of (ESCC) in esophageal squamous cell carcinoma. Long non-coding RNAs (lncRNAs) regulate the progression of ESCC by various pathophysiological pathways. We explored the possible function of the lncRNA LINC00261 (LINC00261) on cisplatin (DDP) resistance of ESCC and its relative molecular mechanisms. In the study, we found that LINC00261 was downregulated in ESCC tissues, cell lines, and DDP-resistant ESCC patients. Besides, overexpression of LINC00261 not only inhibited cell proliferation, and DDP resistance but also promotes cell apoptosis. Further mechanistic research showed that LINC00261 sponged miR-545-3p which was negatively correlated with the expression of LINC00261. In addition, functional experiments revealed that upregulation of miR-766-5p promoted proliferation and enhanced DDP resistance. Subsequently, MT1M was testified to be the downstream target gene of miR-545-3p. Rescue experiments revealed that overexpression of MT1M largely restores miR-545-3p mimics-mediated function on ESCC progression. Our results demonstrate that the LINC00261 suppressed the DDP resistance of ESCC through miR-545-3p/MT1M axis.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaojun Wang
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Pengwei Yan
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Yatian Liu
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Xuesong Jiang
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
156
|
Wu X, Han R, Zhong Y, Weng N, Zhang A. Post treatment NLR is a predictor of response to immune checkpoint inhibitor therapy in patients with esophageal squamous cell carcinoma. Cancer Cell Int 2021; 21:356. [PMID: 34233686 PMCID: PMC8262036 DOI: 10.1186/s12935-021-02072-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In view of the fact that peripheral blood parameters have been reported as predictors of immunotherapy to various cancers, this study aimed to determine the predictors of response to anti-programmed death-1 (anti-PD-1) therapy in patients with esophageal squamous cell carcinoma (ESCC) from peripheral blood parameters. METHODS A retrospective analysis was conducted to investigate the predictive value of peripheral blood parameters including neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), monocyte-to-lymphocyte ratio (MLR) and systemic immune-inflammation index (SII) in the response to anti-PD-1 antibody treatment. 119 ESCC patients receiving combined treatment including anti-PD-1 antibody were enrolled in this study. RESULTS The median progression-free survival (PFS) of all ESCC patients was 3.73 months. PFS rate in ESCC patients with low NLR at 6 weeks post treatment was higher than patients with high NLR (HR = 2.097, 95% CI 0.996-4.417, P = 0.027). However, PFS rate in ESCC patients with low NLR at baseline (HR = 1.060, 95% CI 0.524-2.146, P = 0.869) or 3 weeks post treatment (HR = 1.293, 95% CI 0.628-2.663, P = 0.459) was comparable with high NLR. And no statistically different was found in PFS rate between low PLR and high PLR at baseline (HR = 0.786, 95% CI 0.389-1.589, P = 0.469), 3 weeks post treatment (HR = 0.767, 95% CI 0.379-1.552, P = 0.452) or 6 weeks post treatment (HR = 1.272, 95% CI 0.624-2.594, P = 0.488) in ESCC patients. PFS rate was also comparable between low MLR and high MLR at baseline (HR = 0.826, 95% CI 0.408-1.670, P = 0.587), 3 weeks post treatment (HR = 1.209, 95% CI 0.590-2.475, P = 0.580) or 6 weeks post treatment (HR = 1.199, 95% CI 0.586-2.454, P = 0.596). PFS rate was similar between patients with low SII and high SII at baseline (HR = 1.120, 95% CI 0.554-2.264, P = 0.749), 3 weeks post treatment (HR = 1.022, 95% CI 0.500-2.089, P = 0.951) and 6 weeks post treatment (HR = 1.759, 95% CI 0.851-3.635, P = 0.097). CONCLUSIONS NLR at 6 weeks post treatment is a predictor of the response to anti-PD-1 treatment in patients with ESCC.
Collapse
Affiliation(s)
- Xianbin Wu
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Runkun Han
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Yanping Zhong
- Department of Health Management, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Nuoqing Weng
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China.
| | - Ao Zhang
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
157
|
Pu J, Zhang T, Zhang D, He K, Chen Y, Sun X, Long W. High-Expression of Cytoplasmic Poly (A) Binding Protein 1 (PABPC1) as a Prognostic Biomarker for Early-Stage Esophageal Squamous Cell Carcinoma. Cancer Manag Res 2021; 13:5361-5372. [PMID: 34262344 PMCID: PMC8275044 DOI: 10.2147/cmar.s317631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/15/2021] [Indexed: 01/16/2023] Open
Abstract
Background and Objective Poly (A) binding protein cytoplasmic 1 (PABPC1) plays a crucial role in the regulation of RNA polyadenylation, translation initiation, and mRNA stability and may be involved in tumorigenesis. Herein, we set out to identify the prognostic value of PABPC1 expression in esophageal squamous cell carcinoma (ESCC). Methods Using quantitative real-time PCR (qRT-PCR) and immunohistochemical analysis, the present study investigated mRNA and protein expressions of PABPC1 in 231 ESCCs and their paired adjacent normal epithelial tissues. Results We observed a reduction in the average mRNA expression of PABPC1 in ESCC tissue specimen, but the mRNA expression of PABPC1 was significantly higher (P<0.001) in ESCC tissues with high PABPC1 expression and lower (P=0.033) in tissues with low PABPC1 expression. In immunohistochemical analysis, positive expression of the PABPC1 protein was identified in 179 ESCC tissue specimens (179/231, 77.5%), while the percentage of ESCC tissue specimens with high expression of PABPC1 was found to be 41.1% (95/231). PABPC1 expression was found to be significantly correlated with lymph node metastasis (LNM) (P=0.011), pathological stage (P=0.021), tumor recurrence (P<0.001), and the outcome (P<0.001) of patients with ESCC. High expression of PABPC1 was associated with poor overall survival (OS) of ESCC patients (P<0.001) among all pathological stages, particularly in the early stages (pStage-I and -II), and identified to be an independent prognostic factor for OS of patients with ESCC in multivariate analysis (HR=2.622; 95% CI, 1.68–4.129). Comparatively, the expression of Ki-67, p53, and nm23 was not associated with OS. Conclusion In this study, we discovered that PABPC1 is a prognostic biomarker and a therapeutic target for ESCC, particularly early-stage ESCC.
Collapse
Affiliation(s)
- Jiangtao Pu
- Thoracic Surgery Department of the First Affiliated Hospital, Southwest Medical University, Sichuan, People's Republic of China
| | - Tao Zhang
- Thoracic Surgery Department of the First Affiliated Hospital, Southwest Medical University, Sichuan, People's Republic of China
| | - Dengguo Zhang
- Thoracic Surgery Department of the First Affiliated Hospital, Southwest Medical University, Sichuan, People's Republic of China
| | - Kaiming He
- Thoracic Surgery Department of the First Affiliated Hospital, Southwest Medical University, Sichuan, People's Republic of China
| | - Yonghong Chen
- Laboratory of Affiliated Hospital of traditional Chinese medicine of Southwest Medical University, Sichuan, People's Republic of China
| | - Xingwang Sun
- Pathology Department of the First Affiliated Hospital, Southwest Medical University, Sichuan, People's Republic of China
| | - Wenbo Long
- Pathology Department of the First Affiliated Hospital, Southwest Medical University, Sichuan, People's Republic of China
| |
Collapse
|
158
|
Xu B, Chen H, Xu Z, Yao X, Sun X, Cheng H. CDCA2 promotes tumorigenesis and induces radioresistance in oesophageal squamous cell carcinoma cells. Mol Med Rep 2021; 24:530. [PMID: 34036376 PMCID: PMC8170267 DOI: 10.3892/mmr.2021.12169] [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: 10/11/2020] [Accepted: 04/15/2021] [Indexed: 12/24/2022] Open
Abstract
Cell division cycle‑associated 2 (CDCA2) overexpression has been demonstrated to serve a significant role in tumorigenesis in certain types of cancer. Nevertheless, its role in tumour proliferation and radioresistance in oesophageal squamous cell carcinoma (ESCC) remains to be elucidated. Thus, the present study aimed to elucidate these roles. Data were downloaded from The Cancer Genome Atlas (TCGA) to compare the gene expression profiles. The expression of CDCA2 was higher in ESCC tissues compared with normal tissues. Gene set enrichment analysis was performed based on the ESCC cohorts in TCGA database. This demonstrated that higher expression of CDCA2 was significantly associated with the expression of related components of the cell cycle phase transition and G2/M phase transition pathways. Collectively, the results revealed that CDCA2 could serve as an underlying target to regulate tumour growth and radioresistance among patients with ESCC.
Collapse
Affiliation(s)
- Bing Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hui Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zhipeng Xu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xijuan Yao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Hongyan Cheng
- Department of Synthetic Internal Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
159
|
Li Y, Zhu X, Yang M, Wang Y, Li J, Fang J, Guo W, Ma S, Guan F. YAP/TEAD4-induced KIF4A contributes to the progression and worse prognosis of esophageal squamous cell carcinoma. Mol Carcinog 2021; 60:440-454. [PMID: 34003522 DOI: 10.1002/mc.23303] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
Aberrant expression of kinesin family member 4A (KIF4A), which is associated with tumor progression, has been reported in several types of cancer. However, its expression and the underlying molecular mechanisms regulating the transcription of KIF4A in esophageal squamous cell carcinoma (ESCC) remain largely unclear. Here, we found that high KIF4A expression was positively correlated with tumor stage and poor prognosis in ESCC patients. KIF4A silencing significantly inhibited the growth and migration of ESCC cells, arrested cell cycle, and induced apoptosis. Interestingly, KIF4A expression was positively related to the expression of YAP in human ESCC tissues. YAP knockdown or disrupting YAP/TEAD4 interaction by verteporfin repressed KIF4A expression. Also, KIF4A knockdown significantly inhibited the cell growth induced by YAP overexpression. Mechanistically, YAP activated KIF4A transcriptional expression by TEAD4-mediated direct binding to KIF4A promoter. Finally, KIF4A knockdown and verteporfin treatment synergistically inhibited tumor growth in xenograft models. Together, these results indicated that KIF4A, a novel target gene of YAP/TEAD4, may be a progression and prognostic biomarker of ESCC. Targeting drugs for KIF4A combined with YAP inhibitor may be a novel therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Ya Li
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiangzhan Zhu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences , East China Normal University, Shanghai, China
| | - Minglei Yang
- Department of Orthopedic Oncology, Changzheng Hospital, The Second Military Medical University, Shanghai, China
| | - Yingying Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Jianhui Li
- Department of Pathology, Xuchang Central Hospital Affiliated to Henan University of Science and Technology, Xuchang, China
| | - Jiarui Fang
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Wenna Guo
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
160
|
Yao W, Chen Q, Li S, Jia X, Xu L, Wei L. RELT promotes the growth of esophageal squamous cell carcinoma by activating the NF-κB pathway. Cell Cycle 2021; 20:1231-1241. [PMID: 34121605 PMCID: PMC8331000 DOI: 10.1080/15384101.2021.1924451] [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: 03/02/2020] [Accepted: 04/27/2021] [Indexed: 10/21/2022] Open
Abstract
The members of the tumor necrosis factor receptor (TNFR) family have been demonstrated to play critical roles in various cancers. However, little is known about the function of the Receptor Expressed in Lymphoid Tissues (RELT) in cancers, which is a member of the TNFR family, especially in the esophageal squamous cell carcinoma (ESCC). In this study, we found that RELT expression was increased in ESCC tissues and was consequently associated with poor overall survival of ESCC patients. Moreover, RELT overexpression was found to promote cell growth, cell cycle progression, and suppressed cell apoptosis in vitro; it also decreased the expression of p27 and caspase 3, and increased the expression of survivin. In addition, RELT contributed to the tumorigenesis of ESCC in vivo. Furthermore, we suggest that RELT may function in the pathogenesis of ESCC by activating the nuclear factor κB (NF-κB) pathway. An inhibitor of NF-κB reversed the RELT-induced malignancy in the ESCC cells. Altogether, our findings identified that RELT served as an oncogene in ESCC through the NF-κB pathway, suggesting that RELT may be developed as a novel biomarker for the diagnosis and treatment of the ESCC.
Collapse
Affiliation(s)
- Wenjian Yao
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Qing Chen
- Department of Oncology, Jingjiang People’s Hospital, Jingjiang, Jiangsu, P. R. China
| | - Saisai Li
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Xiangbo Jia
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Lei Xu
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| | - Li Wei
- Department of Thoracic Surgery, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, Henan, China
| |
Collapse
|
161
|
Lu Z, Zheng S, Liu C, Wang X, Zhang G, Wang F, Wang S, Huang J, Mao S, Lei Y, Wang Z, Sun N, He J. S100A7 as a potential diagnostic and prognostic biomarker of esophageal squamous cell carcinoma promotes M2 macrophage infiltration and angiogenesis. Clin Transl Med 2021; 11:e459. [PMID: 34323409 PMCID: PMC8265170 DOI: 10.1002/ctm2.459] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Dysregulated expression of S100A7 is found in several cancers and plays an important role in tumor progression; however, its carcinogenic role in esophageal squamous carcinoma (ESCC) is still poorly understood. Here, we identified that the levels of S100A7 were remarkably upregulated in 341 tumor tissues (P < .001) and 274 serum samples (P < .001) of ESCC patients compared with normal control. It was an independent prognostic factor (P = .026). Furthermore, a new diagnostic model for ESCC based on serum S100A7, SCC, and crfra21-1 was established with area under curve (AUC) up to 0.863 (95% CI: 0.802-0.925). Mechanically, we found upregulated S100A7 could promote cell migration and proliferation through intracellular binding to JAB1 and paracrine interaction with RAGE receptors and then activates the downstream signaling pathways. In addition, exocrine S100A7 could promote M2 macrophage infiltration and polarization by up-regulating M2 macrophage associated proteins, and tumor angiogenesis by enhancing the activation of p-ErK and p-FAK pathways. Further animal experiments confirmed the role of S100A7 in promoting M2 macrophage infiltration and angiogenesis in ESCC. In conclusion, these findings highlighted the potential diagnostic and prognostic value of S100A7 in patients with ESCC. Meanwhile, our results reveal that S100A7 promotes tumor progression by activating oncogenic pathways and remodeling tumor microenvironment, which paving the way for the progress of S100A7 as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Zhiliang Lu
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Sufei Zheng
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Chengming Liu
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xinfeng Wang
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Guochao Zhang
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Feng Wang
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Sihui Wang
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jianbing Huang
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shuangshuang Mao
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yuanyuan Lei
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - ZhanYu Wang
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Nan Sun
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jie He
- Department of Thoracic SurgeryState Key Laboratory of Molecular Oncology/National Cancer Center/National Clinical Research for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
162
|
Chen X, Zhang Y, Shi Y, Niu T, Li B, Guo L, Qiao Y, Zhao J, Yuan B, Liu K. Evolution of DNA aptamers against esophageal squamous cell carcinoma using cell-SELEX. Analyst 2021; 146:4180-4187. [PMID: 34105524 DOI: 10.1039/d1an00634g] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Esophageal cancer is the ninth most common cancer and the sixth most common cause of cancer-related death worldwide, and the esophageal squamous cell carcinoma (ESCC) subtype accounts for about 90% of all cases of esophageal cancer globally. Currently, ESCC is usually diagnosed in late stages, and targeted therapy is lacking. Therefore, the development of ESCC-specific recognition molecules for an early detection and targeted treatment of ESCC is urgently needed. Aptamers are an excellent molecular recognition tool with unique advantages. In this manuscript, three aptamers (S2, S3, and S8) specific to ESCC cells were successfully screened via cell-SELEX. The experimental results displayed the high affinities of the three aptamers for target KYSE150 cells with dissociation constants in the nanomolar range. The specificity evaluation showed that S2 only bound target KYSE150 cells, but S3 and S8 were capable of targeting a series of ESCC cells. Moreover, several truncated aptamers were generated through sequence optimization. In particular, an ultrashort aptamer S3-2-3 with only 18 bases was successfully obtained; after labeling with Cy5 dyes, it was feasible for the specific imaging of ESCC tissues. Furthermore, the target types of the selected aptamers were preliminarily identified as membrane proteins, and target proteins could be captured by S3-2-3, which may be useful for biomarker discovery. Therefore, the selected aptamers hold great potential for clinical diagnosis, biomarker discovery, and the targeted therapy of ESCC.
Collapse
Affiliation(s)
- Xinhuan Chen
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China. and Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, China and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Yangyang Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Yanli Shi
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Tingting Niu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Bo Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China. and Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, China and China-US Hormel (Henan) Cancer Institute, Zhengzhou, Henan, China
| | - Linyan Guo
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Yan Qiao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China. and Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, China and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China. and Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, China and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Baoyin Yuan
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China. and Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, China and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China. and Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou, Henan, China and State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, Henan, China and China-US Hormel (Henan) Cancer Institute, Zhengzhou, Henan, China and Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou, Henan, China
| |
Collapse
|
163
|
Morshedi K, Borran S, Ebrahimi MS, Masoud Khooy MJ, Seyedi ZS, Amiri A, Abbasi-Kolli M, Fallah M, Khan H, Sahebkar A, Mirzaei H. Therapeutic effect of curcumin in gastrointestinal cancers: A comprehensive review. Phytother Res 2021; 35:4834-4897. [PMID: 34173992 DOI: 10.1002/ptr.7119] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/18/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022]
Abstract
Gastrointestinal (GI) cancers with a high global prevalence are a leading cause of morbidity and mortality. Accordingly, there is a great need to develop efficient therapeutic approaches. Curcumin, a naturally occurring agent, is a promising compound with documented safety and anticancer activities. Recent studies have demonstrated the activity of curcumin in the prevention and treatment of different cancers. According to systematic studies on curcumin use in various diseases, it can be particularly effective in GI cancers because of its high bioavailability in the gastrointestinal tract. Nevertheless, the clinical applications of curcumin are largely limited because of its low solubility and low chemical stability in water. These limitations may be addressed by the use of relevant analogues or novel delivery systems. Herein, we summarize the pharmacological effects of curcumin against GI cancers. Moreover, we highlight the application of curcumin's analogues and novel delivery systems in the treatment of GI cancers.
Collapse
Affiliation(s)
- Korosh Morshedi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sarina Borran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Zeynab Sadat Seyedi
- Department of Cell and Molecular Biology, Faculty of Chemistry, University of Kashan, Kashan, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Abbasi-Kolli
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maryam Fallah
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
164
|
Pan Q, Li B, Zhang J, Du X, Gu D. LncRNA THAP9-AS1 accelerates cell growth of esophageal squamous cell carcinoma through sponging miR-335-5p to regulate SGMS2. Pathol Res Pract 2021; 224:153526. [PMID: 34273804 DOI: 10.1016/j.prp.2021.153526] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 01/20/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is kind of common and aggressive malignant tumors with high incidence and mortality all over the world. Accumulating studies have reported that long non-coding RNAs (lncRNAs) can play a vital regulatory role in human cancers. THAP9 antisense RNA 1 (THAP9-AS1) has been identified as an oncogene in several cancers. But its role in ESCC remains to be studied. In our research, THAP9-AS1 expression in ESCC cell lines was analyzed by real-time quantitative polymerase chain reaction (RT-qPCR). Cell proliferation, migration, invasion and apoptosis as well as EMT process were analyzed by 5-Ethynyl-2'-deoxyuridine ( EdU), Transwell, Terminal deoxynucleotidyl transferase dUTP Nick-End Labeling (TUNEL) and western blot experiments. The interplay of THAP9-AS1, miR-335-5p and sphingomyelin synthase 2 (SGMS2) was analyzed by luciferase reporter assay and RNA immunoprecipitation (RIP) assay. We discovered that THAP9-AS1 was highly expressed in ESCC cell lines and that the knockdown of THAP9-AS1 inhibited proliferation, migration, and invasion as well as EMT of ECSS cells but enhanced cell apoptosis. Furthermore, miR-335-5p was proved to be sponged by THAP9-AS1 and its up-regulation could repress ESCC progression. Additionally, SGMS2 was verified to be the target gene of miR-335-5p. In rescue assay, SGMS2 overexpression could offset the suppressive role of THAP9-AS1 depletion on ESCC progression. In short, THAP9-AS1 accelerated cell growth of ESCC through sponging miR-335-5p to regulate SGMS2.
Collapse
Affiliation(s)
- Qingchun Pan
- Department of Otolaryngology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Bei Li
- Department of Otolaryngology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan, China
| | - Jin Zhang
- Department of Pathology, Suzhou Science & Technology Town Hospital, Suzhou 215153, Jiangsu, China
| | - Xiuluan Du
- Department of Pathology, Suzhou Science & Technology Town Hospital, Suzhou 215153, Jiangsu, China
| | - Donghua Gu
- Department of Pathology, Suzhou Science & Technology Town Hospital, Suzhou 215153, Jiangsu, China.
| |
Collapse
|
165
|
Identification of Prognostic Risk Model Based on DNA Methylation-Driven Genes in Esophageal Adenocarcinoma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6628391. [PMID: 34222478 PMCID: PMC8213479 DOI: 10.1155/2021/6628391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/15/2021] [Accepted: 05/08/2021] [Indexed: 01/10/2023]
Abstract
Background DNA methylation is an important part of epigenetic modification, and its abnormality is closely related to esophageal adenocarcinoma (EAC). This study was aimed at using bioinformatics analysis to identify methylation-driven genes (MDGs) in EAC patients and establish a risk model as a biological indicator of EAC prognosis. Method Downloaded EAC DNA methylation, transcriptome, and related clinical data from TCGA database. MethylMix was used to identify MDGs. R package clusterProfiler and the ConsensusPathDB online database were used to analyze the rich functions and pathways of these MDGs. The prognostic risk model was established by univariate Cox regression, Lasso regression, and multivariate Cox regression analysis. Finally each MDG in the model were carried out through the survival R package. Results A total of 273 MDGs were identified, which were enriched in transcriptional regulation and embryonic organ morphogenesis. Cox regression analysis established a risk model consisting of GPBAR1, OLFM4, FOXI2, and CASP10. In addition, further survival analysis revealed that OLFM4 and its two related sites were significantly related to the EAC patients' survival. Conclusion In summary, this study used bioinformatics methods to identify EAC MDGs and established a reliable risk prognosis model. It provided potential biomarkers for the early treatment and prognosis evaluation of EAC.
Collapse
|
166
|
Zhu ZJ, Pang Y, Jin G, Zhang HY, Wang WH, Liu JW, Tuo GX, Wu P, Yang Y, Wang ZQ, Wang K. Hypoxia induces chemoresistance of esophageal cancer cells to cisplatin through regulating the lncRNA-EMS/miR-758-3p/WTAP axis. Aging (Albany NY) 2021; 13:17155-17176. [PMID: 34081626 PMCID: PMC8312407 DOI: 10.18632/aging.203062] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/14/2021] [Indexed: 04/10/2023]
Abstract
Hypoxia contributes significantly to the development of chemoresistance of many malignancies including esophageal cancer (EC). Accumulating studies have indicated that long non-coding RNAs play important roles in chemotherapy resistance. Here, we identified a novel lncRNA-EMS/miR-758-3p/WTAP axis that was involved in hypoxia-mediated chemoresistance to cisplatin in human EC. Hypoxia induced the expressions of lncRNA EMS and WTAP, and reduced the expression of miR-758-3p in EC cell line ECA-109. In addition, the expressions of EMS and WTAP were required for the hypoxia-induced drug resistance to cisplatin in EC cells, while overexpression of miR-758-3p reversed such chemoresistance. The targeting relationships between EMS and miR-758-3p, as well as miR-758-3p and WTAP, were verified by luciferase-based reporter assays and multiple quantitative assays after gene overexpression/knockdown. Moreover, we found significant correlations between tumor expressions of these molecules. Notably, higher levels of EMS/WTAP, or lower levels of miR-758-3p in tumors predicted worse survivals of EC patients. Furthermore, in a xenograft mouse model, targeted knockdown of EMS and WTAP in ECA-109 cells markedly attenuated the resistance of tumors to cisplatin treatments. Our study uncovers a critical lncRNA-EMS/miR-758-3p/WTAP axis in regulating hypoxia-mediated drug resistance to cisplatin in EC.
Collapse
Affiliation(s)
- Zi-Jiang Zhu
- Department of Thoracic Surgery 2, Gansu Provincial People's Hospital, Lanzhou 730000, China
| | - Yao Pang
- Department of Thoracic Surgery 2, Gansu Provincial People's Hospital, Lanzhou 730000, China
| | - Gang Jin
- Department of Thoracic Surgery 2, Gansu Provincial People's Hospital, Lanzhou 730000, China
| | - Hong-Yi Zhang
- Department of Thoracic Surgery 2, Gansu Provincial People's Hospital, Lanzhou 730000, China
| | - Wen-Hao Wang
- Department of Thoracic Surgery 2, Gansu Provincial People's Hospital, Lanzhou 730000, China
| | - Jia-Wei Liu
- Department of Thoracic Surgery 2, Gansu Provincial People's Hospital, Lanzhou 730000, China
| | - Guang-Xin Tuo
- School of Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Peng Wu
- School of Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Yi Yang
- School of Clinical Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou 730000, China
| | - Ze-Quan Wang
- School of Clinical Medicine, Ningxia Medical University, Ningxia 750004, China
| | - Kui Wang
- School of Clinical Medicine, Ningxia Medical University, Ningxia 750004, China
| |
Collapse
|
167
|
Lu Y, Guan L, Xu M, Wang F. The efficacy and safety of antibodies targeting PD-1 for treatment in advanced esophageal cancer: A systematic review and meta-analysis. Transl Oncol 2021; 14:101083. [PMID: 33784583 PMCID: PMC8042180 DOI: 10.1016/j.tranon.2021.101083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/18/2021] [Accepted: 03/19/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND A novel therapy based on programmed death 1 (PD-1) inhibitors has been proved to be effective in advanced esophageal cancer. This article is a meta-analysis that aims to compare the efficacy and safety of anti-PD-1 therapy with chemotherapy in esophageal cancer. PATIENTS AND METHODS Data were collected from eligible studies searched from PubMed, Web of Science, Cochrane Library, and Embase. Pooled hazard ratio (HR) for overall survival (OS), progression-free survival (PFS), and objective response rate (ORR) was estimated to assess the efficacy of PD-1 inhibitors versus chemotherapy. The subgroup analysis was also performed to evaluate the OS benefits. The OR for the occurrence of treatment-related adverse effects was calculated to assess the safety of anti-PD-1 therapy. RESULTS A total of 4 studies were analyzed. Compared with patients with chemotherapy, patients with anti-PD-1 therapy had a significant improvement in OS (HR = 0.79, 95% CI: 0.71-0.88, and P<0.001), but no significant relationship was observed in PFS (HR = 0.96, 95% CI: 0.76-1.20, and P = 0.69) and ORR (OR = 1.92, 95% CI: 0.98-3.72, and P = 0.06). A similar result was observed in esophageal squamous cell carcinoma. The significant predictor for treatment benefit alone was histology (P = 0.009). The incidence of grade 3 - 5 treatment-related adverse effects in anti-PD-1 therapy was distinctly lower than that in chemotherapy, but there is no statistical difference in all treatment-related adverse effects. CONCLUSION Anti-PD-1 therapy significantly prolonged the OS, simultaneously lowered grade 3 - 5 treatment-related adverse effects versus chemotherapy.
Collapse
Affiliation(s)
- Yao Lu
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P R China
| | - Lulu Guan
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P R China
| | - Mengli Xu
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P R China
| | - Feng Wang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, P R China.
| |
Collapse
|
168
|
Kwak AW, Lee MJ, Lee MH, Yoon G, Cho SS, Chae JI, Shim JH. The 3-deoxysappanchalcone induces ROS-mediated apoptosis and cell cycle arrest via JNK/p38 MAPKs signaling pathway in human esophageal cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 86:153564. [PMID: 33895649 DOI: 10.1016/j.phymed.2021.153564] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 03/26/2021] [Accepted: 03/31/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND The 3-deoxysappanchalcone (3-DSC), a chemical separated from Caesalpinia sappan L, has been substantiated to display anti-inflammatory, anti-influenza, and anti-allergy activities according to previous studies. However, the underlying mechanisms of action on esophageal cancer remain unknown. PURPOSE The present research aims to survey the action mechanisms of 3-DSC in esophageal squamous cell carcinoma (ESCC) cells in vitro. METHODS Evaluation of cytotoxicity was determined by MTT tetrazolium salt assay and soft agar assay. Cell cycle distribution, apoptosis induction, reactive oxygen species (ROS) generation, mitochondrial membrane potential (MMP), and multi-caspases activity were appreciated by Muse™ Cell Analyzer. The expressions of cell cycle- and apoptosis-related proteins were presented using Western blotting. RESULTS 3-DSC blocked cell growth and colony formation ability in a concentration-dependent manner and invoked apoptosis, G2/M cell cycle arrest, ROS production, MMP depolarization, and multi-caspase activity. Furthermore, Western blotting results demonstrated that 3-DSC upregulated the expression of phospho (p)-c-jun NH2-terminal kinases (JNK), p-p38, cell cycle regulators, pro-apoptotic proteins, and endoplasmic reticulum (ER) stress-related proteins whereas downregulated the levels of anti-apoptotic proteins and cell cycle promoters. The effects of 3-DSC on ROS induction were counteracted by pretreatment with N-acetyl-L-cysteine (NAC). Also, our results indicated that p38 (SB203580) and JNK (SP600125) inhibitor slightly inhibited 3-DSC-induced apoptosis. These results showed that 3-DSC-related G2/M phase cell cycle arrest and apoptosis by JNK/p38 MAPK signaling pathway in ESCC cells were mediated by ROS. CONCLUSION ROS generation by 3-DSC in cancer cells could be an attractive strategy for apoptosis of cancer cells by inducing cell cycle arrest, ER stress, MMP loss, multi-caspase activity, and JNK/p38 MAPK pathway. Our findings suggest that 3-DSC is a promising novel therapeutic candidate for both prevention and treatment of esophageal cancer.
Collapse
Affiliation(s)
- Ah-Won Kwak
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Myeoung-Jun Lee
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Mee-Hyun Lee
- College of Korean Medicine, Dongshin University, Naju, Jeonnam 58245, Republic of Korea
| | - Goo Yoon
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Seung-Sik Cho
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Jeonnam 58554, Republic of Korea
| | - Jung-Il Chae
- Department of Dental Pharmacology, School of Dentistry, Jeonbuk National University, Jeonju 54896, Republic of Korea.
| | - Jung-Hyun Shim
- Department of Pharmacy, College of Pharmacy, Mokpo National University, Jeonnam 58554, Republic of Korea; The China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450008, PR China; Department of Biomedicine, Health & Life Convergence Sciences, BK21 Four, Biomedical and Healthcare Research Institute, Mokpo National University, Jeonnam 58554, Republic of Korea.
| |
Collapse
|
169
|
Sakai A, Nakashima Y, Miyashita Y, Ao T, Kimura Y, Shinto E, Oki E, Shimokawa M, Ueno H, Oda Y, Mori M. Histological categorisation of the desmoplastic reaction is a predictor of patient prognosis in oesophageal squamous cell carcinoma. Histopathology 2021; 79:219-226. [PMID: 33595141 DOI: 10.1111/his.14357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/12/2021] [Accepted: 02/14/2021] [Indexed: 01/06/2023]
Abstract
AIMS Histological categorisation of the desmoplastic reaction (DR) is an independent prognostic factor in colorectal cancer. However, it is unknown whether DR categorisation is predictive of oesophageal squamous cell carcinoma (OSCC) outcomes. This study aimed to evaluate the prognostic value of DR categorisation in OSCC patients. METHODS AND RESULTS Data were collected from 118 patients with OSCC who underwent a curative oesophagectomy with T2 or deeper wall invasion. The DR in each tumour was classified as mature, intermediate or immature based on the presence or absence of keloid-like collagen and myxoid stroma. We identified 49 mature DR tumours, 41 intermediate DR tumours and 28 immature DR tumours. The 5-year overall survival (OS) rate was highest in the mature DR group (42.8%), followed by the intermediate DR group (25.0%) and the immature DR group (19.9%) (P = 0.022, log-rank test; P = 0.006, log-rank trend test). The 5-year disease-specific survival (DSS) rate was also highest in the mature DR group (48.5%), followed by the intermediate DR group (30.8%) and the immature DR group (26.8%) (P = 0.031, log-rank test; P = 0.010, log-rank trend test, respectively). Multivariate analysis revealed that an immature DR was an independent poor prognostic factor of OS and DSS (P = 0.002 and P = 0.004). CONCLUSIONS DR categorisation of OSCC stroma following oesophagectomy is a useful diagnostic tool and an independent prognostic marker.
Collapse
Affiliation(s)
- Akihiro Sakai
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Anatomic Pathology, Pathological Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuichiro Nakashima
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yu Miyashita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Anatomic Pathology, Pathological Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tadakazu Ao
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Yasue Kimura
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Shinto
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
170
|
Feasibility of hepatic resection for liver metastasis of head-and-neck carcinoma or esophageal carcinoma: a multi-center experience. Surg Today 2021; 51:1932-1937. [PMID: 34032927 DOI: 10.1007/s00595-021-02305-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 03/10/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Patients with liver metastasis of head-and-neck carcinoma and esophageal carcinoma are generally not treated with hepatic resection, but there are no established standard treatment methods. We report 11 cases of hepatic resection for liver metastasis of head-and-neck carcinoma or esophageal carcinoma performed at 5 Japanese institutions. METHODS The subjects of this retrospective analysis were 11 patients who underwent hepatic resection for metastatic liver tumors, originating from head-and-neck carcinoma in 5 and from esophageal cancer in 6, between January, 2010 and March, 2020 RESULTS: There were nine men and two women (median age, 64 years; range 40-72 years). The primary disease was esophageal carcinoma in six patients and pharyngeal carcinoma in five patients. All cancers were squamous cell carcinoma. The time from the initial treatment to the diagnosis of liver metastasis was 15.3 months and the 1-year and 3-year overall survival rates after hepatic resection were 72% and 32%, respectively. The overall and disease-free survival rates after hepatic resection were significantly higher for patients who underwent hepatic resection more than 12 months after the initial treatment than for those who underwent hepatic resection within 12 months after the initial treatment (p = 0.0172 and p = 0.0120, respectively). CONCLUSIONS Liver resection may prolong the survival of patients with liver metastases controlled for more than 12 months after the initial treatment of head and neck or esophageal carcinoma.
Collapse
|
171
|
Sasaki F, Kanmura S, Oda K, Maeda H, Kabayama M, Iwaya H, Komaki Y, Arima S, Tanoue S, Hashimoto S, Fujita H, Ido A. Acetaldehyde breath test as a cancer risk marker in patients with esophageal and hypopharyngeal squamous cell carcinoma. PLoS One 2021; 16:e0251457. [PMID: 34010294 PMCID: PMC8133406 DOI: 10.1371/journal.pone.0251457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/26/2021] [Indexed: 11/21/2022] Open
Abstract
Patients with inactive acetaldehyde dehydrogenase 2 (ALDH2) are at high risk for esophageal squamous cell carcinoma (ESCC) and hypopharyngeal squamous cell carcinoma (HPSCC). The acetaldehyde breath test (ABT) may demonstrate ALDH2 gene polymorphisms. We evaluated the usefulness of the ABT in patients with ESCC and HPSCC. The squamous cell carcinoma (SCC) group consisted of 100 patients who were treated with endoscopic submucosal dissection (ESD) for ESCC or HPSCC, and the control group (HC) consisted of 275 healthy subjects. The SCC group comprised the “single subgroup” (n = 63), in which a single lesion was initially treated with ESD, and the “multiple subgroup” (n = 31), in which multiple lesions were initially treated with ESD. First, we compared the groups’ risk factors for carcinogenesis and measured the acetaldehyde-to-ethanol (A/E) ratio. Then we tested the groups’ differences in the abovementioned carcinogenic risk factors. We found that the proportion of individuals in the SCC group with inactive ALDH2 (A/E ratio ≥ 23.3) was significantly higher than that in the HC group (p = 0.035), as was the A/E ratio (p < 0.001). Also, the proportion of individuals with inactive ALDH2 in the multiple subgroup was significantly higher than that in single subgroup (p = 0.015), as was the A/E ratio (p = 0.008). In conclusion, ABT may be a potential screening tool for detecting people at risk of ESCC and HPSCC. In addition, it could be a useful tool in detecting patients at risk of multiple or double carcinomas among patients with ESCC and HPSCC. Trial registration: Trial Registration number: UMIN000040615 [https://rctportal.niph.go.jp/en/detail?trial_id=UMIN000040615], Data of Registration: 01 46 June 2020, retrospectively registered.
Collapse
Affiliation(s)
- Fumisato Sasaki
- Department of Human and Environmental Sciences, Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
- * E-mail:
| | - Shuji Kanmura
- Department of Human and Environmental Sciences, Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kohei Oda
- Department of Human and Environmental Sciences, Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hidehito Maeda
- Department of Gastroenterology and Hepatology, Izumi General Medical Center, Izumi city, Kagoshima, Japan
| | - Masayuki Kabayama
- Department of Human and Environmental Sciences, Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hiromichi Iwaya
- Department of Human and Environmental Sciences, Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuga Komaki
- Department of Human and Environmental Sciences, Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shiho Arima
- Department of Human and Environmental Sciences, Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shiroh Tanoue
- Department of Human and Environmental Sciences, Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shinichi Hashimoto
- Department of Human and Environmental Sciences, Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hiroshi Fujita
- Department of Gastroenterology and Hepatology, Izumi General Medical Center, Izumi city, Kagoshima, Japan
| | - Akio Ido
- Department of Human and Environmental Sciences, Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
172
|
Gao X, Liu Q, Chen X, Chen S, Yang J, Liu Q, Cheng Y. Screening of tumor grade-related mRNAs and lncRNAs for Esophagus Squamous Cell Carcinoma. J Clin Lab Anal 2021; 35:e23797. [PMID: 33960436 PMCID: PMC8183932 DOI: 10.1002/jcla.23797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 11/25/2022] Open
Abstract
Background The goal of our study was to screen tumor grade‐related lncRNAs and mRNAs to reveal the underlying molecular mechanism of esophagus squamous cell carcinoma (ESCC). Methods The lncRNA and mRNA sequencing data were obtained from The Cancer Genome Atlas (TCGA). Tumor grade correlation analysis of lncRNAs and mRNAs was executed, followed by the functional enrichment analysis of all tumor grade‐related mRNAs. The differentially expression mRNAs (DEmRNAs) and differentially expressed lncRNAs (DElncRNAs) were obtained. PPI network and DEmRNA‐DElncRNA interaction analysis were constructed. The functional annotation of the DEmRNAs co‐expressed with DElncRNAs was performed. The expression levels of the candidate genes were validated using qRT‐PCR. Results A total of 1864 tumor grade‐related mRNAs (846 positively related and 1018 negatively related) and 552 tumor grade‐related lncRNAs (331 positively related and 221 negatively related) were obtained. The top 10 significantly grade‐related mRNAs and lncRNAs included CA12, FABP4, DECR1, BAIAP2, IL1RAPL2, PPARD, LAD1, TSPAN10, LDOC1, ZNF853, RP11‐25G10.2, RP11‐557H15.3, RP11‐521D12.5, CHKB‐AS1, RP11‐219B4.3, CH17‐335B8.4, RP11‐99 J16‐A.2, CTB‐111H14.1, ADNP‐AS1, and JHDM1D‐AS1. SFN, IL1RAPL2, and RP11‐25G10.2 were overlapped from grade 1, grade 2, and grade 3. PPI network showed that top 10 proteins with higher degrees, including GNAI1, RAP2B, GNAZ, SHH, ADCY1, PRKAR2B, SH3GL1, GNA15, and ARRB1. A DElncRNAs‐nearby DEmRNAs network was constructed to obtain hub lncRNAs including ADAMTS9‐AS2, RP11‐210 M15.2, RP11‐13 K12.1, ZBED3‐AS1, and RP11‐25G10.2. Except for RP11‐25G10.2, ADAMTS9‐AS1, ZBED3‐AS1, SFN, ATP1A2, and GNA15 were consistent with our TCGA analysis. Conclusions Alterations of DEmRNAs and DElncRNAs may provide key insights into the molecular mechanisms of ESCC.
Collapse
Affiliation(s)
- Xin Gao
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Oncology, Dongying People's Hospital, Dongying, China
| | - Qian Liu
- Department of Oncology, Dongying People's Hospital, Dongying, China
| | - Xue Chen
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shaoping Chen
- Department of Oncology, Dongying People's Hospital, Dongying, China
| | - Jianmei Yang
- Department of Oncology, Dongying People's Hospital, Dongying, China
| | - Qiang Liu
- Department of Oncology, Dongying People's Hospital, Dongying, China
| | - Yufeng Cheng
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
173
|
Ana Choi SS, Ko JMY, Yu VZ, Ning L, Lung ML. Differentiation-related zinc finger protein 750 suppresses cell growth in esophageal squamous cell carcinoma. Oncol Lett 2021; 22:513. [PMID: 33986873 DOI: 10.3892/ol.2021.12774] [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: 11/08/2020] [Accepted: 03/12/2021] [Indexed: 11/06/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a deadly squamous cell carcinoma (SCC) of the esophagus. Development of SCCs is associated with the deregulation of the squamous cell lineage program and/or keratinocyte terminal differentiation by genomic and genetic aberrations; thus, these processes must be tightly controlled to maintain normal squamous cell development. Zinc finger protein 750 (ZNF750) is a gene involved in keratinocyte terminal differentiation and is frequently mutated and putatively silenced in ESCC, which implicates its function as a potential differentiation-related suppressor of ESCC. The present study aimed to elucidate the relationship between ZNF750 function to induce keratinocyte differentiation and tumor suppression in ESCC. The results demonstrated that chemical manipulation of esophageal keratinocyte differentiation in mouse normal esophageal epithelial organoids (mNEEO) implicated the involvement of the mouse homologue of ZNF750, Zfp750, in keratinocyte differentiation in premalignant cells. Bioinformatics analyses of data from high ZNF750-expressing ESCC tumors obtained from public databases and ZNF750-overexpressing ESCC cells compared with low ZNF750-expressing ESCC tumors and GFP-expressing ESCC cells, respectively, revealed enrichment of keratinocyte differentiation-related gene sets in these samples. Finally, the induction through to terminal differentiation of the keratinocyte by all-trans retinoic acid on parental ESCC cell lines led to the upregulation of the terminal differentiation marker Involucrin and a decrease in cell viability similar to that observed in ZNF750-overexpressing ESCC cells. The results of the present study demonstrated a functional link between the ability of ZNF750 to induce cell differentiation through to terminal differentiation and its function as a growth suppressor in ESCC. This study provides improved understanding of the role of ZNF750, a frequently mutated differentiation-related gene in ESCC, and its effects in ESCC pathogenesis.
Collapse
Affiliation(s)
- Sheyne Sta Ana Choi
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Josephine Mun-Yee Ko
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Valen Zhuoyou Yu
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Lvwen Ning
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Maria Li Lung
- Department of Clinical Oncology, The University of Hong Kong, Hong Kong, SAR, P.R. China
| |
Collapse
|
174
|
Luebeck GE, Vaughan TL, Curtius K, Hazelton WD. Modeling historic incidence trends implies early field cancerization in esophageal squamous cell carcinoma. PLoS Comput Biol 2021; 17:e1008961. [PMID: 33939693 PMCID: PMC8118544 DOI: 10.1371/journal.pcbi.1008961] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/13/2021] [Accepted: 04/13/2021] [Indexed: 12/31/2022] Open
Abstract
Patterns of cancer incidence, viewed over extended time periods, reveal important aspects of multistage carcinogenesis. Here we show how a multistage clonal expansion (MSCE) model for cancer can be harnessed to identify biological processes that shape the surprisingly dynamic and disparate incidence patterns of esophageal squamous cell carcinoma (ESCC) in the US population. While the dramatic rise in esophageal adenocarcinoma (EAC) in the US has been largely attributed to reflux related increases in the prevalence of Barrett’s esophagus (BE), the premalignant field in which most EAC are thought to arise, only scant evidence exists for field cancerization contributing to ESCC. Our analyses of incidence patterns suggest that ESCC is associated with a premalignant field that may develop very early in life. Although the risk of ESCC, which is substantially higher in Blacks than Whites, is generally assumed to be associated with late-childhood and adult exposures to carcinogens, such as from tobacco smoking, alcohol consumption and various industrial exposures, the temporal trends we identify for ESCC suggest an onset distribution of field-defects before age 10, most strongly among Blacks. These trends differ significantly in shape and strength from field-defect trends that we estimate for US Whites. Moreover, the rates of ESCC-predisposing field-defects predicted by the model for cohorts of black children are decreasing for more recent birth cohorts (for Blacks born after 1940). These results point to a potential etiologic role of factors acting early in life, perhaps related to nutritional deficiencies, in the development of ESCC and its predisposing field-defect. Such factors may explain some of the striking racial differences seen in ESCC incidence patterns over time in the US. We used a cell-level carcinogenesis model to analyze incidence patterns of esophageal squamous cell carcinoma (ESCC) in the US. We found an important role of an esophageal field-defect that is predicted to occur predominantly in childhood and predisposes to ESCC in adult life. Age-specific ESCC incidence patterns are also known to differ considerably between Blacks and Whites, and between males and females in the US, but the model consistently predicts early-childhood field-defects in all four groups. The estimated historical field-defect trends appear consistent with possible early childhood nutritional deficiencies.
Collapse
Affiliation(s)
- Georg E. Luebeck
- Public Health Sciences Division, Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- * E-mail:
| | - Thomas L. Vaughan
- Professor Emeritus, Public Health Sciences Division, Cancer Epidemiology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Kit Curtius
- Division of Biomedical Informatics, Department of Medicine, University of California, San Diego, La Jolla, California, United States of America
| | - William D. Hazelton
- Public Health Sciences Division, Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| |
Collapse
|
175
|
Marabotto E, Pellegatta G, Sheijani AD, Ziola S, Zentilin P, De Marzo MG, Giannini EG, Ghisa M, Barberio B, Scarpa M, Angriman I, Fassan M, Savarino V, Savarino E. Prevention Strategies for Esophageal Cancer-An Expert Review. Cancers (Basel) 2021; 13:2183. [PMID: 34062788 PMCID: PMC8125297 DOI: 10.3390/cancers13092183] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
In the last 30 years, we have witnessed a rapid increase in the incidence and prevalence of esophageal cancer in many countries around the word. However, despite advancements in diagnostic technologies, the early detection of this cancer is rare, and its prognosis remains poor, with only about 20% of these patients surviving for 5 years. The two major forms are the esophageal squamous cell carcinoma (ESCC), which is particularly frequent in the so-called Asian belt, and the esophageal adenocarcinoma (EAC), which prevails in Western populations. This review provides a summary of the epidemiological features and risk factors associated with these tumors. Moreover, a major focus is posed on reporting and highlighting the various preventing strategies proposed by the most important international scientific societies, particularly in high-risk populations, with the final aim of detecting these lesions as early as possible and therefore favoring their definite cure. Indeed, we have conducted analysis with attention to the current primary, secondary and tertiary prevention guidelines in both ESCC and EAC, attempting to emphasize unresolved research and clinical problems related to these topics in order to improve our diagnostic strategies and management.
Collapse
Affiliation(s)
- Elisa Marabotto
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Gaia Pellegatta
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Milan, Italy;
| | - Afscin Djahandideh Sheijani
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Sebastiano Ziola
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Patrizia Zentilin
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Maria Giulia De Marzo
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Edoardo Giovanni Giannini
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Matteo Ghisa
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (M.G.); (B.B.)
| | - Brigida Barberio
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (M.G.); (B.B.)
| | - Marco Scarpa
- Clinica Chirurgica 1, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (M.S.); (I.A.)
| | - Imerio Angriman
- Clinica Chirurgica 1, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (M.S.); (I.A.)
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy;
| | - Vincenzo Savarino
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Edoardo Savarino
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (M.G.); (B.B.)
| |
Collapse
|
176
|
Prevalence of HER2 overexpression and amplification in squamous cell carcinoma of the esophagus: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2021; 161:103339. [PMID: 33865993 DOI: 10.1016/j.critrevonc.2021.103339] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 11/22/2022] Open
Abstract
Accurate data on HER2 positivity in esophageal squamous cell carcinoma patients (ESCC) is lacking. We conducted a systematic review and meta-analysis (Single Incidence Rates; metarate package, R) to examine the prevalence of HER2 in ESCC. Data on in situ hybridization (ISH) and immunohistochemistry (IHC) were extracted to derive pooled prevalence estimates, characteristics of the studies were extracted for subgroup analysis. Eighteen studies with 1505 patients were identified. HER2 gene amplification by ISH were prevalent in 10 % (95 % CI 6.9 %-15 %). Prevalence of HER2 overexpression (IHC3+) and borderline HER2 expression (IHC2+) were 6 % (95 % CI: 3.5 %-8.7 %) and 10 % (95 % CI: 6.0 %-17 %), respectively. An estimated 8.6 % (95 % CI: 5.5 %-13 %) of ESCC were HER2 positive using initial IHC followed by reflex ISH confirmation of borderline HER2 expression. In conclusion: Estimated prevalence of HER 2 positivity in ESCC were 10 % assessed by ISH and 8.6 % assessed by initial IHC followed by ISH.
Collapse
|
177
|
Iwai N, Dohi O, Yamada S, Harusato A, Horie R, Yasuda T, Yamada N, Horii Y, Majima A, Zen K, Kimura H, Yagi N, Naito Y, Itoh Y. Prognostic risk factors associated with esophageal squamous cell carcinoma patients undergoing endoscopic submucosal dissection: a multi-center cohort study. Surg Endosc 2021; 36:2279-2289. [PMID: 33860352 DOI: 10.1007/s00464-021-08502-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/03/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Long-term outcomes of endoscopic submucosal dissection (ESD) for esophageal squamous cell carcinoma (ESCC) have not been assessed in a large, multicenter cohort. We aimed to evaluate long-term outcomes of ESD for ESCC in a real-world setting. METHODS We retrospectively recruited 659 patients who underwent ESD for ESCC at ten institutions from January 2007 to December 2015. Of these, 566 patients were analyzed and classified into three groups according to the pathologic invasion depth after ESD: epithelium/lamina propria mucosa (EP/LPM group: 454 patients), muscularis mucosa/submucosa invasion ≤ 200 μm below the inferior margin of the muscularis mucosa (MM/SM1 group: 81 patients), and submucosa invasion > 200 μm below the MM inferior margin (SM2 group: 31 patients). RESULTS The 5-year overall survival rates in the EP/LPM, MM/SM1, and SM2 groups were 92.6%, 80.0%, and 62.7%, respectively, while the 5-year disease-specific survival rates were 99.7%, 96.9%, and 88.3%, respectively. Multivariate analyses revealed that the invasion depth, Charlson Comorbidity Index (CCI), and prognostic nutritional index (PNI) were independent prognostic factors. Hazard ratios in the MM/SM1 and SM2 groups were 2.25 (95% confidence interval [CI] 1.04-4.83; P = 0.038) and 3.18 (95% CI 1.08-9.34; P = 0.036), respectively, compared to those in the EP/LPM group, while those for patients with a CCI ≥ 3 and PNI ≤ 47.75 were 3.25 (95% CI 1.79-5.89; P < 0.001) and 2.42 (95% CI 1.26-4.65; P = 0.008), respectively. CONCLUSIONS This study identified that invasion depth, presence of comorbid diseases and preoperative nutritional status are independent prognostic risk factors associated with ESCC patients undergoing ESD.
Collapse
Affiliation(s)
- Naoto Iwai
- Department of Gastroenterology and Hepatology, Fukuchiyama City Hospital, Fukuchiyama, Kyoto, Japan.,Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Osamu Dohi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Shinya Yamada
- Department of Gastroenterology and Hepatology, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Akihito Harusato
- Department of Gastroenterology, North Medical Center, Kyoto Prefectural University of Medicine, Yosanocho, Kyoto, Japan
| | - Ryusuke Horie
- Department of Gastroenterology, JCHO Kyoto Kuramaguchi Medical Center, Kyoto, Japan
| | - Takeshi Yasuda
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan.,Department of Gastroenterology, Asahi University Hospital, Gifu, Japan
| | - Nobuhisa Yamada
- Department of Gastroenterology, Matsushita Memorial Hospital, Moriguchi, Osaka, Japan
| | - Yusuke Horii
- Department of Gastroenterology, Medical Corporation Keishinkai, Kyoto Kizugawa Hospital, Joyo, Kyoto, Japan
| | - Atsushi Majima
- Department of Gastroenterology and Hepatology, Omihachiman Community Medical Center, Omihachiman, Shiga, Japan
| | - Keika Zen
- Department of Gastroenterology and Hepatology, Otsu City Hospital, Otsu, Shiga, Japan
| | - Hiroyuki Kimura
- Department of Gastroenterology and Hepatology, Japanese Red Cross Society Kyoto Daiichi Hospital, Kyoto, Japan
| | - Nobuaki Yagi
- Department of Gastroenterology, Asahi University Hospital, Gifu, Japan
| | - Yuji Naito
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
178
|
Prediction of the tumor response and survival based on computed tomography in esophageal squamous cell carcinoma after trimodality therapy. Surg Today 2021; 51:1496-1505. [PMID: 33797639 DOI: 10.1007/s00595-021-02277-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/09/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE Predicting the response to neoadjuvant chemoradiotherapy (NCRT) and the prognosis of esophageal squamous cell carcinoma (ESCC) is challenging. This study evaluated the potential of a preoperative computed tomography (CT) analysis for predicting the pathological response and survival of patients with ESCC who received trimodality therapy. METHODS A total of 119 patients with cT3 or T4 ESCC who underwent surgery following NCRT between 2007 and 2019 were assessed. The CT-based parameters were measured with enhanced CT preoperatively, prior to and during treatment. Associations between these parameters and the pathologic response, as well as the prognosis, were examined. RESULTS Pretreatment maximum CT value (p = 0.009), pretreatment mean CT value (p = 0.022), preoperative whole tumor volume (WTV, p = 0.003), preoperative largest cross section (LCS, p = 0.002), ΔLCS (pretreatment to preoperative, p = 0.004), ΔWTV (pretreatment to preoperative, p = 0.012), and Δmean CT value (pretreatment to preoperative, p = 0.012) of the primary lesion were significantly associated with a good pathological response. A Cox-regression analysis of recurrence-free survival (RFS) and overall survival (OS) showed that preoperative LCS was an independent CT-based predictor. RFS and OS were significantly higher at the optimal cut-off value of the preoperative LCS (p = 0.036 and p = 0.016, respectively). CONCLUSIONS CT-based parameters are thus considered to be valuable predictors of the tumor response and survival after trimodality therapy for ESCC.
Collapse
|
179
|
Achalasia and esophageal cancer: a large database analysis in Japan. J Gastroenterol 2021; 56:360-370. [PMID: 33538893 DOI: 10.1007/s00535-021-01763-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 01/20/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Achalasia has been reported to be associated with esophageal cancers (ECs). However, owing to the rarity of achalasia, details of achalasia-related ECs are not well investigated. METHOD The incidence of ECs in Japanese patients with achalasia and achalasia-related esophageal motility disorders (EMDs) was estimated, and risk factors for achalasia-related ECs were determined. Characteristics of ECs and treatment courses were also analyzed. RESULTS Between 2010 and 2019, 2714 Japanese patients with achalasia and achalasia-related EMDs were recorded in 7 high-volume centers; 24 patients (21 men, 3 women) developed ECs. The incidence of ECs was estimated at 0.078 and 0.28 per 100 person-years from the onset and the diagnosis of disease, respectively. Kaplan-Meier estimate suggested that, in addition to a long history of achalasia, advanced age, male sex, and regular alcohol consumption were statistically significant risk factors for EC development. A prevalence of 40 ECs (12.5% multiple lesions, and 22.7% metachronal lesions) was also noted, predominantly distributed over the thoracic esophagus. All were histologically diagnosed as squamous cell carcinoma. Superficial ECs were successfully treated with endoscopic treatment in all cases, except one. Achalasia-related Barret esophagus was extremely rare, and Barret adenocarcinoma was not detected in our cohort. CONCLUSION The high relative risk of ECs was clarified in Japanese achalasia patients, although the absolute risk remained low. Therefore, surveillance endoscopy may be recommended in limited patients with several aforementioned risk factors determined. Superficial cancer can be treated with endoscopic treatment. Multiple and metachronal ECs should be screened.
Collapse
|
180
|
Luo Q, Wu X, Zhao P, Nan Y, Chang W, Zhu X, Su D, Liu Z. OTUD1 Activates Caspase-Independent and Caspase-Dependent Apoptosis by Promoting AIF Nuclear Translocation and MCL1 Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2002874. [PMID: 33898171 PMCID: PMC8061361 DOI: 10.1002/advs.202002874] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 12/29/2020] [Indexed: 05/25/2023]
Abstract
Apoptosis-inducing factor (AIF) plays a dual role in regulating cell survival and apoptosis, acting as a prosurvival factor in mitochondria via its NADH oxidoreductase activity and activating the caspase-independent apoptotic pathway (i.e., parthanatos) after nuclear translocation. However, whether one factor conjunctively controls the separated functions of AIF is not clear. Here, it is shown that OTU deubiquitinase 1 (OTUD1) acts as a link between the two functions of AIF via deubiquitination events. Deubiquitination of AIF at K244 disrupts the normal mitochondrial structure and compromises oxidative phosphorylation, and deubiquitination of AIF at K255 enhances its DNA-binding ability to promote parthanatos. Moreover, OTUD1 stabilizes DDB1 and CUL4 associated factor 10 (DCAF10) and recruits the cullin 4A (CUL4A)-damage specific DNA binding protein 1 (DDB1) complex to promote myeloid cell leukemia sequence 1 (MCL1) degradation, thereby activating caspase-dependent apoptotic signaling. Collectively, these results reveal the central role of OTUD1 in activating both caspase-independent and caspase-dependent apoptotic signaling and propose decreased OTUD1 expression as a key event promoting chemoresistance in esophageal squamous cell carcinoma.
Collapse
Affiliation(s)
- Qingyu Luo
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Xiaowei Wu
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Pengfei Zhao
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Yabing Nan
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Wan Chang
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Xiaolin Zhu
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| | - Dan Su
- Department of PathologyZhejiang Cancer HospitalZhejiang310022China
| | - Zhihua Liu
- State Key Laboratory of Molecular OncologyNational Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100021China
| |
Collapse
|
181
|
Cao Y, Gao A, Li X, Min H, He C, Sun X, Ding WQ, Zhou J. Elevated TAB182 enhances the radioresistance of esophageal squamous cell carcinoma through G2-M checkpoint modulation. Cancer Med 2021; 10:3101-3112. [PMID: 33787085 PMCID: PMC8085956 DOI: 10.1002/cam4.3879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/01/2021] [Accepted: 03/16/2021] [Indexed: 11/09/2022] Open
Abstract
Background Radiotherapy is one of the main strategies for the treatment of esophageal squamous cell carcinoma (ESCC). However, treatment failure often occurs due to the emergence of radioresistance. In this study, we report a key regulator of radiation sensitivity, termed TAB182 that may become an ideal biomarker and therapeutic target to overcome radioresistance. Materials and Methods By applying qRT‐PCR and immunohistochemical staining, the expression of TAB182 was detected in patient tissues. We next assessed the influence of TAB182 downregulation to radiosensitivity using clonogenic survival assay and γ‐H2A.X foci analysis in TE‐1, TE‐10, and radioresistant TE‐1R cell lines after ionizing radiation. To unveil the mechanism underlying, TAB182 interacting proteins were identified by mass spectrometry following co‐immunoprecipitation. Furthermore, flow cytometry and western blot assay were applied to validate the identified proteins. Results Our results demonstrated that the expression of TAB182 is higher in cancer tissues than normal tissues and elevated expression of TAB182 correlates with poor outcomes of postoperative radiotherapy. Downregulation of TAB182 sensitized cancer cells to ionizing radiation, particularly in radioresistant TE‐1R cells that spontaneously overexpress TAB182. Mechanically, TAB182 interacts with FHL2 to induce G2‐M arrest through wiring the CHK2/CDC25C/CDC2 signaling pathway. Finally, overexpression of shRNA‐resistant TAB182 restored the checkpoint and radioresistance. Conclusion TAB182 potentiates the radioresistance of ESCC cells by modulating the G2‐M checkpoint through its interaction with FHL2. Thus, TAB182 may become an ideal biomarker and therapeutic target of ESCC radiotherapy.
Collapse
Affiliation(s)
- Yuandong Cao
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Aidi Gao
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| | - Xiaoqing Li
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| | - Han Min
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| | - Chao He
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Wei-Qun Ding
- Department of Pathology, University of Oklahoma Health Science Center, Oklahoma City, OK, USA
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, P.R. China
| |
Collapse
|
182
|
Peng Z, Gong Y, Liang X. Role of FAT1 in health and disease. Oncol Lett 2021; 21:398. [PMID: 33777221 PMCID: PMC7988705 DOI: 10.3892/ol.2021.12659] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 02/25/2021] [Indexed: 01/15/2023] Open
Abstract
FAT atypical cadherin 1 (FAT1), which encodes a protocadherin, is one of the most frequently mutated genes in human cancer. Over the past 20 years, the role of FAT1 in tissue growth and in the development of diseases has been extensively studied. There is definitive evidence that FAT1 serves a substantial role in the maintenance of organs and development, and its expression appears to be tissue-specific. FAT1 activates a variety of signaling pathways through protein-protein interactions, including the Wnt/β-catenin, Hippo and MAPK/ERK signaling pathways, which affect cell proliferation, migration and invasion. Abnormal FAT1 expression may lead to the development of tumors and may affect prognosis. Therefore, FAT1 may have potential in tumor therapy. The structural and functional changes mediated by FAT1, its tissue distribution and changes in FAT1 expression in human diseases are described in the present review, which provides further insight for understanding the role of FAT1 in development and disease.
Collapse
Affiliation(s)
- Zizhen Peng
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang School of Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yanyu Gong
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang School of Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiaoqiu Liang
- Hunan Province Key Laboratory of Tumor Cellular and Molecular Pathology, Cancer Research Institute, Hengyang School of Medicine, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
183
|
Wu X, Zhang H, Sui Z, Wang Y, Yu Z. The biological role of the CXCL12/CXCR4 axis in esophageal squamous cell carcinoma. Cancer Biol Med 2021; 18:j.issn.2095-3941.2020.0140. [PMID: 33710803 PMCID: PMC8185864 DOI: 10.20892/j.issn.2095-3941.2020.0140] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/30/2020] [Indexed: 12/12/2022] Open
Abstract
Esophageal cancer is the eighth most common malignant tumor and the sixth leading cause of cancer-related death worldwide. Esophageal squamous cell carcinoma (ESCC) is the main histological type of esophageal cancer, and accounts for 90% of all cancer cases. Despite the progress made in surgery, chemotherapy, and radiotherapy, the mortality rate from esophageal cancer remains high, and the overall 5-year survival rate is less than 20%, even in developed countries. The C-X-C motif chemokine ligand 12 (CXCL12) is a member of the CXC chemokine subgroup, which is widely expressed in a variety of tissues and cells. CXCL12 participates in the regulation of many physiological and pathological processes by binding to its specific receptor, C-X-C motif chemokine receptor type 4 (CXCR4), where it causes embryonic development, immune response, and angiogenesis. In addition, increasing evidence indicates that the CXCL12/CXCR4 axis plays an important role in the biological processes of tumor cells. Studies have shown that CXCL12 and its receptor, CXCR4, are highly expressed in ESCC. This abnormal expression contributes to tumor proliferation, lymph node and distant metastases, and worsening prognosis. At present, antagonists and imaging agents against CXCL12 or CXCR4 have been developed to interfere with the malignant process and monitor metastasis of tumors. This article summarizes the structure, function, and regulatory mechanism of CXCL12/CXCR4 and its role in the malignancy of ESCC. Current results from preclinical research targeting CXCL12/CXCR4 are also summarized to provide a reference for the clinical diagnosis and treatment of ESCC.
Collapse
Affiliation(s)
- Xianxian Wu
- Departments of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Hongdian Zhang
- Departments of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhilin Sui
- Departments of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yang Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| | - Zhentao Yu
- Departments of Esophageal Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
184
|
Hoshino I, Ishige F, Iwatate Y, Gunji H, Kuwayama N, Nabeya Y, Yokota H, Takeshita N, Iida K, Nagase H, Matsubara H. Cell-free microRNA-1246 in different body fluids as a diagnostic biomarker for esophageal squamous cell carcinoma. PLoS One 2021; 16:e0248016. [PMID: 33690683 PMCID: PMC7946279 DOI: 10.1371/journal.pone.0248016] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Esophageal squamous cell carcinoma is a malignant tumor with unfavorable prognosis. In this study, we investigated the usefulness of microRNA (miR)-1246 detection in various body fluids as a biomarker for this disease. A total of 72 patients with esophageal squamous cell carcinoma were enrolled, and their blood, urine, and saliva samples were collected prior to treatment. Reverse transcription-polymerase chain reaction of miR-1246 was performed, and pre- and postoperative and intraday fluctuations in its expression were examined. The expression of miR-1246 in the blood and urine was significantly higher in the patients with esophageal squamous cell carcinoma than in 50 healthy control subjects. Receiver operating characteristic curves showed that the area under the curve values were 0.91 (sensitivity 91.7%, specificity 76.0%), 0.82 (sensitivity 90.3%, specificity 62.0%), and 0.80 (sensitivity 83.3%, specificity 66.0%) in the serum, urine, and saliva, respectively. A relatively high diagnostic performance of miR-1246 was observed in all samples, which was better than that of the existing biomarkers squamous cell carcinoma antigen, carcinoembryonic antigen, and cytokeratin 19 fragment. No clear correlation was observed in the levels of miR-1246 expression among the three body fluids. Postoperatively, serum samples displayed significantly decreased miR-1246 levels. Although not significant, changes in the miR-1246 levels were observed at all collection times, with large fluctuations in the saliva. Meanwhile, serum miR-1246 expression was found to be associated with the disease prognosis. The results indicate that the levels of miR-1246 in the urine, saliva, and serum are a useful biomarker for esophageal squamous cell carcinoma and support the use of urine samples instead of blood samples for noninvasive diagnosis.
Collapse
Affiliation(s)
- Isamu Hoshino
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
- * E-mail:
| | - Fumitaka Ishige
- Department of Hepatobiliary and Pancreatic Surgery, Chiba Cancer Center, Chiba, Japan
| | - Yosuke Iwatate
- Department of Hepatobiliary and Pancreatic Surgery, Chiba Cancer Center, Chiba, Japan
| | - Hisashi Gunji
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Naoki Kuwayama
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Yoshihiro Nabeya
- Division of Gastroenterological Surgery, Chiba Cancer Center, Chiba, Japan
| | - Hajime Yokota
- Department of Diagnostic Radiology and Radiation Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Nobuyoshi Takeshita
- Division of Surgical Technology, National Cancer Center Hospital East, Chiba, Japan
| | - Keiko Iida
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroki Nagase
- Laboratory of Cancer Genetics, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Hisahiro Matsubara
- Department of Frontier Surgery, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
185
|
Shu J, Ma J, Ren X, Wang J, Wang Y, Zhang K, Yu H, Guo X, Li Z. The Abnormal Glycopatterns of Salivary Glycoproteins in Esophageal Squamous Cell Carcinoma Patients. Front Chem 2021; 9:637730. [PMID: 33748076 PMCID: PMC7969727 DOI: 10.3389/fchem.2021.637730] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/25/2021] [Indexed: 01/04/2023] Open
Abstract
Glycosylation is one of the most crucial posttranslational modifications of proteins, containing a remarkable amount of biological information. The alteration of glycosylation is closely associated with certain diseases. Exploring glyco-code in the development of diseases is a hot topic in recent years. Esophageal squamous cell carcinoma (ESCC) is the primary pathological histology in developing countries and a severe threat to human health. Although the glycan profiles in the blood samples of ESCC patients were analyzed using glycomic and glycoproteomic methods, the difference of salivary glycopatterns between healthy subjects and ESCC patients is not explicit yet. In the present study, ESCC patients (n = 16) and healthy volunteers (HVs, n = 25) were enrolled. The glycomic strategy combining lectin microarray and lectin blotting was employed to investigate and confirm the altered salivary glycopatterns. Datura stramonium (DSA) was selected to isolate the GlcNAc or Galβ1-4GlcNA-containing glycoproteins due to the distinct difference between ESCC patients and HVs. The N-glycans from DSA-enriched glycoproteins were released by PNGase F and further identified by MALDI-TOF/TOF-MS to obtain the precise structural information of the altered glycans. As a result, the glycopatterns recognized by 13 lectins (e.g., ECA, RCA120, and DSA) showed significant alterations in ESCC patients' saliva. The ESCC patients showed higher levels of GalNAc and Gal, sialic acid, and GlcNAc expression profiles and lower levels of mannose and fucose expression profiles. The MALDI-TOF/TOF-MS results indicated that the proportion of the GlcNAc or Galβ1-4GlcNAc-containing N-glycans was increased in ESCC patients (79.04%) compared with HV (63.20%), which was consistent with the results of lectin microarrays. Our findings provide comprehensive information to understand the complex physiological changes in ESCC patients. And the altered salivary glycopatterns such as GlcNAc or Galβ1-4GlcNAc-containing N-glycans recognized by DSA might serve as potential biomarkers for the diagnosis of ESCC patients.
Collapse
Affiliation(s)
- Jian Shu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Jun Ma
- Institute of Digestive Disease of Zhengzhou University, Zhengzhou, China
- Department of Clinical Laboratory, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiameng Ren
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Jian Wang
- Department of Oncology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Wang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Kun Zhang
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Hanjie Yu
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| | - Xiangqian Guo
- Institute of Biomedical Informatics, Cell Signal Transduction Laboratory, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Zheng Li
- Laboratory for Functional Glycomics, College of Life Sciences, Northwest University, Xi'an, China
| |
Collapse
|
186
|
Amanuma Y, Horimatsu T, Ohashi S, Tamaoki M, Muto M. Association of local complete response with prognosis after salvage photodynamic therapy for esophageal squamous cell carcinoma. Dig Endosc 2021; 33:355-363. [PMID: 32434267 DOI: 10.1111/den.13730] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/14/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Photodynamic therapy (PDT) is an effective salvage endoscopic treatment for local failure at the primary site after chemoradiotherapy (CRT) in esophageal cancer patients. However, the contribution of local control by salvage PDT to the prognosis is unclear. We investigated whether complete response at primary site by salvage PDT could improve the prognosis. METHODS Between January 2008 and March 2016, 34 patients received salvage PDT for local failure of esophageal cancer limited to stage T1-2 after definitive CRT or radiotherapy. Local complete response (L-CR) rate, adverse events, overall survival (OS), and progression-free survival (PFS) were assessed retrospectively. RESULTS Local complete response rates after PDT were 68% (23/34; 95% CI, 50-83%) in all patients: 81% (17/21; 95% CI, 58-95%) for stage T1 and 46% (6/13; 95% CI, 19-75%) for stage T2 patients. Grade 3 esophageal stricture occurred in one patient. The median follow-up was 26.0 months (range, 3.7-93.6 months); 21 patients died. The median survival times were 54.3 months in patients who achieved L-CR after PDT (L-CR group) and 19.8 months in those who did not (non-CR group). The 2-year OS rates were 79% (95% CI, 54-92%) in the L-CR group and 40% (95% CI, 11-68%) in the non-CR group (P = 0.0389; log-rank test). The median PFS was 21.2 months in the L-CR group and 1.9 months in the non-CR group (P < 0.001; log-rank test). CONCLUSION Achieving L-CR by salvage PDT for local failure after CRT in esophageal cancer was associated with good prognosis.
Collapse
Affiliation(s)
- Yusuke Amanuma
- Department of Clinical Oncology, Kyoto University, Kyoto, Japan.,Clinical Trial Promotion Department, Chiba Cancer Center, Chiba, Japan
| | | | - Shinya Ohashi
- Department of Clinical Oncology, Kyoto University, Kyoto, Japan
| | - Masashi Tamaoki
- Department of Clinical Oncology, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Clinical Oncology, Kyoto University, Kyoto, Japan
| |
Collapse
|
187
|
Tarazi M, Chidambaram S, Markar SR. Risk Factors of Esophageal Squamous Cell Carcinoma beyond Alcohol and Smoking. Cancers (Basel) 2021; 13:cancers13051009. [PMID: 33671026 PMCID: PMC7957519 DOI: 10.3390/cancers13051009] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/20/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the sixth most common cause of death worldwide. Incidence rates vary internationally, with the highest rates found in Southern and Eastern Africa, and central Asia. Initial observational studies identified multiple factors associated with an increased risk of ESCC, with subsequent work then focused on developing plausible biological mechanistic associations. The aim of this review is to summarize the role of risk factors in the development of ESCC and propose future directions for further research. A systematic search of the literature was conducted by screening EMBASE, MEDLINE/PubMed, and CENTRAL for relevant publications. In total, 73 studies were included that sought to identify risk factors associated with the development of esophageal squamous cell carcinoma. Risk factors were divided into seven subcategories: genetic, dietary and nutrition, gastric atrophy, infection and microbiome, metabolic, epidemiological and environmental and other risk factors. Risk factors from each subcategory were summarized and explored with mechanistic explanations for these associations. This review highlights several current risk factors of ESCC. These risk factors were explored, and explanations dissected. Most studies focused on investigating genetic and dietary and nutritional factors, whereas this review identified other potential risk factors that have yet to be fully explored. Furthermore, there is a lack of literature on the association of these risk factors with tumor factors and disease prognosis. Further research to validate these results and their effects on tumor biology is absolutely necessary.
Collapse
Affiliation(s)
- Munir Tarazi
- Department of Surgery and Cancer, Imperial College London, London W2 1NY, UK; (M.T.); (S.C.)
| | - Swathikan Chidambaram
- Department of Surgery and Cancer, Imperial College London, London W2 1NY, UK; (M.T.); (S.C.)
| | - Sheraz R. Markar
- Department of Surgery and Cancer, Imperial College London, London W2 1NY, UK; (M.T.); (S.C.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, 17164 Stockholm, Sweden
- Correspondence:
| |
Collapse
|
188
|
Ma Z, Dong Z, Yu D, Mu M, Feng W, Guo J, Cheng B, Guo J, Ma J. IL-32 Promotes the Radiosensitivity of Esophageal Squamous Cell Carcinoma Cell through STAT3 Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6653747. [PMID: 33681363 PMCID: PMC7904356 DOI: 10.1155/2021/6653747] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 12/30/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE This study is set out to determine the relationship between IL-32 and radiosensitivity of esophageal squamous cell carcinoma (ESCC). METHODS Western blot was adopted for measuring IL-32 expression in Eca-109 and TE-10 cells. Eca-109 and TE-10 cells with interference or overexpression of IL-32 were treated with the presence or absence of X-ray irradiation. Then, the use of CCK8 assay was to detect proliferation ability, and effects of IL-32 expression on radiosensitivity of ESCC were tested by colony formation assay. The cell apoptosis was detected using flow cytometry. STAT3 and p-STAT expression, and apoptotic protein Bax were detected by western blot. RESULTS Colony formation assay and CCK8 assay showed that compared with the NC group without treatment, the growth of the ESCC cells, that is Eca-109 and TE-10, was significantly inhibited in the OE+IR group with highly expressed IL-32 and irradiation. In flow cytometry analysis, in Eca-109 and TE-10 cells, highly expressed IL-32 combined with irradiation significantly increased apoptosis compared with the control group. Highly expressed IL-32 has a synergistic effect with irradiation, inhibiting STAT3 and p-STAT3 expression and increasing apoptotic protein Bax expression. CONCLUSION IL-32 can improve the radiosensitivity of ESCC cells by inhibiting the STAT3 pathway. Therefore, IL-32 can be used as a new therapeutic target to provide a new attempt for radiotherapy of ESCC.
Collapse
Affiliation(s)
- Zhiyu Ma
- First Department of Radiotherapy, Wanbei Coal-Electricity Group General Hospital, Suzhou, 234000 Anhui, China
| | - Zhen Dong
- Department of Radiotherapy, BenQ Medical Center Affiliated to Nanjing Medical University, Nanjing, 210000 Jiangsu, China
| | - Dingyue Yu
- Department of Radiotherapy, Bengbu Second People's Hospital Affiliated to Bengbu Medical Collage, Bengbu, 233000 Anhui, China
| | - Mingchen Mu
- Department of Radiotherapy, Lianyungang Municipal Oriental Hospital Affiliated to Bengbu Medical Collage, Lianyungang, 222042 Jiangsu, China
| | - Wanwen Feng
- Translational Medicine Center, Lianyungang Municipal Oriental Hospital Affiliated to Bengbu Medical Collage, Lianyungang, 222042 Jiangsu, China
| | - Jiayi Guo
- Department of Radiotherapy, Lianyungang Municipal Oriental Hospital Affiliated to Bengbu Medical Collage, Lianyungang, 222042 Jiangsu, China
| | - Beibei Cheng
- Department of Radiotherapy, Lianyungang Municipal Oriental Hospital Affiliated to Bengbu Medical Collage, Lianyungang, 222042 Jiangsu, China
| | - Jiayou Guo
- Department of Radiotherapy, Lianyungang Municipal Oriental Hospital Affiliated to Bengbu Medical Collage, Lianyungang, 222042 Jiangsu, China
| | - Jianxin Ma
- Department of Radiotherapy, Lianyungang Municipal Oriental Hospital Affiliated to Bengbu Medical Collage, Lianyungang, 222042 Jiangsu, China
| |
Collapse
|
189
|
Zang HL, Ji FJ, Ju HY, Tian XF. Circular RNA AKT3 governs malignant behaviors of esophageal cancer cells by sponging miR-17-5p. World J Gastroenterol 2021; 27:240-254. [PMID: 33519139 PMCID: PMC7814369 DOI: 10.3748/wjg.v27.i3.240] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/04/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Recent studies have demonstrated that circular RNA AKT3 (circAKT3) plays a crucial role in regulating the malignant phenotypes of tumor cells. However, the potential effects of circAKT3 on esophageal cancer have not been investigated.
AIM To illuminate the role of circAKT3 in malignant behaviors of esophageal cancer cells and its underlying mechanism.
METHODS Clinical samples were collected to detect the expression of circAKT3. The role of circAKT3 in proliferation, migration, invasion, and apoptosis of esophageal cancer cells was evaluated using Cell Counting Kit-8, wound healing assays, Transwell assays, and fluorescence analysis, respectively. The target of circAKT3 was screened and identified using an online database and luciferase reporter assay. A xenograft nude mouse model was established to investigate the role of circAKT3 in vivo.
RESULTS In vitro assays showed that proliferative, migratory, and invasive capacities of esophageal cancer cells were significantly enhanced by circAKT3 overexpression. Furthermore, miR-17-5p was screened as the target of circAKT3, and miR-17-5p antagonized the effects of circAKT3 on esophageal cancer cells. Moreover, we identified RHOC and STAT3 as the direct target molecules of miR-17-5p, and circAKT3 facilitated expression of RHOC and STAT3 by inhibiting miR-17-5p. In vivo assays showed circAKT3 knockdown inhibited growth of esophageal cancer.
CONCLUSION CircAKT3 contributed to the malignant behaviors of esophageal cancer in vitro and in vivo by sponging miR-17-5p thus providing a potential target for treatment of esophageal cancer.
Collapse
Affiliation(s)
- Hong-Liang Zang
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Fu-Jian Ji
- Department of Colorectal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Hai-Ying Ju
- Department of Hematology, Jilin Province Blood Center, Changchun 130000, Jilin Province, China
| | - Xiao-Feng Tian
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
190
|
Repeated talaporfin sodium photodynamic therapy for esophageal cancer: safety and efficacy. Esophagus 2021; 18:817-824. [PMID: 34106353 PMCID: PMC8387249 DOI: 10.1007/s10388-021-00853-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/22/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND Talaporfin sodium photodynamic therapy (tPDT) is an effective salvage treatment for local failure after chemoradiotherapy for esophageal cancer. Repeated tPDT could also be indicated for local recurrence or residue after the first salvage tPDT. However, the safety and efficacy of repeated tPDT have not been elucidated. METHODS We reviewed 52 patients with esophageal cancer who were treated with the first tPDT at Kyoto University Hospital between October 2015 and April 2020. RESULTS Among 52 patients, repeated tPDT after the first tPDT was indicated for 13 patients (25%), of which six had residual tumor, four had local recurrence after complete response (CR) after the first tPDT at the primary site, and six had metachronous lesion. The total session of repeated tPDT was 25; 16 were for primary sites and nine were for metachronous sites. Among them, six patients (46.2%) achieved local (L)-CR and nine lesions (56.3%) achieved lesion L-CR. By session, 10 sessions (40%) achieved L-CR. There were no severe adverse events except for one patient; this patient showed grade 3 esophageal stenosis and perforation after the third tPDT on the same lesion that was previously treated with porfimer sodium photodynamic therapy four times. CONCLUSION Repeated tPDT could be an effective and safe treatment for local failure even after salvage tPDT for esophageal cancer.
Collapse
|
191
|
Zhu P, Huang H, Gu S, Liu Z, Zhang X, Wu K, Lu T, Li L, Dong C, Zhong C, Zhou Y. Long Noncoding RNA FAM225A Promotes Esophageal Squamous Cell Carcinoma Development and Progression via Sponging MicroRNA-197-5p and Upregulating NONO. J Cancer 2021; 12:1073-1084. [PMID: 33442405 PMCID: PMC7797645 DOI: 10.7150/jca.51292] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/16/2020] [Indexed: 12/18/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the major subclass of esophageal cancer and one of the most life-threatening malignancies with high morbidity and mortality. Long noncoding RNAs (lncRNAs) participate in tumorigenesis and metastasis of various tumors. Here, we investigated the function of a newly identified lncRNA FAM225A in ESCC. LncRNA FAM225A expression was significantly higher in ESCC and predicted poor prognosis of ESCC patients. We confirmed that upregulation of FAM225A in ESCC and overexpression of FAM225A was associated with poor outcome in ESCC patients using TCGA ESCC cohort. Knockdown of FAM225A significantly inhibited cell growth, migration and invasion of ESCC cells in vitro and inhibited ESCC xenograft development in vivo. Mechanistically, we demonstrated that lncRNA FAM225A functioned as a competing endogenous RNA (ceRNA) via sponging miR-197-5p. LncRNA FAM225A exerted its regulatory function on ESCC proliferation and metastasis via modulating expression of miR-197-5p. MiR-197-5p overexpression antagonized the function of FAM225A, with decreased cell growth and invasion. Moreover, we identified that RNA binding protein NONO was a direct target of miR-197-5p and miR-197-5p negatively regulated NONO expression and TGF-β signaling in ESCC cells. In summary, our findings suggest that lncRNA FAM225A promotes ESCC development and progression via sponging miR-197-5p and upregulating NONO expression. These results suggest that lncRNA FAM225A could be explored as a new therapy target in ESCC treatment.
Collapse
Affiliation(s)
- Pengyuan Zhu
- Department of thoracic and Cardiovascular Surgery, the Second Affiliated Hospital of Nantong University, School of Medicine, Nantong University, Nantong, 226001, China
| | - Haitao Huang
- Department of thoracic and Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Shaorui Gu
- Department of thoracic and Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Zhenchuan Liu
- Department of thoracic and Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xin Zhang
- Department of thoracic and Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Kaiqin Wu
- Department of thoracic and Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Tiancheng Lu
- Department of thoracic and Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Lei Li
- Department of thoracic and Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Chenglai Dong
- Department of thoracic and Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Chongjun Zhong
- Department of thoracic and Cardiovascular Surgery, the Second Affiliated Hospital of Nantong University, School of Medicine, Nantong University, Nantong, 226001, China
| | - Yongxin Zhou
- Department of thoracic and Cardiovascular Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| |
Collapse
|
192
|
Qin B, Dong M, Wang Z, Wan J, Xie Y, Jiao Y, Yan D. Long non‑coding RNA CASC15 facilitates esophageal squamous cell carcinoma tumorigenesis via decreasing SIM2 stability via FTO‑mediated demethylation. Oncol Rep 2020; 45:1059-1071. [PMID: 33650646 PMCID: PMC7860005 DOI: 10.3892/or.2020.7917] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/27/2020] [Indexed: 01/18/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are involved in the regulation of esophageal squamous cell carcinoma (ESCC) progression. However, the function and mechanism of lncRNA cancer susceptibility candidate 15 (CASC15) are poorly defined. In the present study, tumor and normal adjacent tissues were collected from 45 patients with ESCC. Expression levels of CASC15, fat mass and obesity-associated (FTO) protein and single-minded 2 (SIM2) were examined via reverse transcription-quantitative PCR and western blot assays. Cell proliferation and apoptosis were evaluated via MTT, flow cytometry and caspase-3 activity assays, respectively. Additionally, an ESCC mouse xenograft model was used to assess the function of CASC15 in vivo. The interaction between FTO and CASC15/SIM2 was analyzed via RNA immunoprecipitation and RNA pull-down assays. The results revealed that CASC15 expression was elevated in ESCC tissues, and patients with ESCC exhibiting high CASC15 expression had a poor prognosis. CASC15-knockdown inhibited ESCC cell proliferation and facilitated apoptosis. Additionally, CASC15-knockdown decreased the growth of ESCC xenograft tumors. CASC15 decreased SIM2 stability via FTO-mediated demethylation. Additionally, FTO loss markedly weakened CASC15-mediated pro-proliferative and anti-apoptotic effects in ESCC cells. SIM2 downregulation weakened the effect of CASC15-knockdown on cell proliferation and inhibited the increase of the apoptotic rate and caspase-3 activity induced by CASC15 depletion in ESCC cells. In conclusion, CASC15 promoted ESCC tumorigenesis by decreasing SIM2 stability via FTO-mediated demethylation.
Collapse
Affiliation(s)
- Bo Qin
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Meng Dong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhengyang Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jiajia Wan
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yingying Xie
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yi Jiao
- Yongcheng Coal & Electricity Holding Group Co., Ltd., Shangqiu, Henan 476000, P.R. China
| | - Dan Yan
- Translational Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
193
|
Enesca A. Enhancing the Photocatalytic Activity of SnO 2-TiO 2 and ZnO-TiO 2 Tandem Structures Toward Indoor Air Decontamination. Front Chem 2020; 8:583270. [PMID: 33324610 PMCID: PMC7723902 DOI: 10.3389/fchem.2020.583270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/16/2020] [Indexed: 12/29/2022] Open
Abstract
ZnO-TiO2 and SnO2-TiO2 tandem structures were developed using the doctor blade technique. It was found that by employing organic hydrophilic and hydrophobic as additives into the precursor it is possible to tailor the film density and morphology with direct consequences on the photocatalytic activity of the tandem structures. The highest photocatalytic efficiency corresponds to ZnO-TiO2 and can reach 74.04% photocatalytic efficiency toward acetaldehyde when a hydrophilic additive is used and 70.93% when a hydrophobic additive is employed. The snO2-TiO2 tandem structure presents lower photocatalytic properties (61.35 % when the hydrophilic additive is used) with a constant rate reaction of 0.07771 min-1.
Collapse
Affiliation(s)
- Alexandru Enesca
- Product Design, Mechatronics and Environmental Department, Transilvania University of Brasov, Braşov, Romania
| |
Collapse
|
194
|
Wen Q, Yang Z, Zhu J, Qiu Q, Dai H, Feng A, Xing L. Pretreatment CT-Based Radiomics Signature as a Potential Imaging Biomarker for Predicting the Expression of PD-L1 and CD8+TILs in ESCC. Onco Targets Ther 2020; 13:12003-12013. [PMID: 33244242 PMCID: PMC7685373 DOI: 10.2147/ott.s261068] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/13/2020] [Indexed: 11/29/2022] Open
Abstract
Background The present study constructed and validated models to predict PD-L1 and CD8+TILs expression levels in esophageal squamous cell carcinoma (ESCC) patients using radiomics features and clinical factors. Patients and Methods This retrospective study randomly assigned 220 ESCC patients to a discovery dataset (n= 160) and validation dataset (n= 60). A total of 462 radiomics features were extracted from the segmentation of regions of interest (ROIs) based on pretreatment CT images of each patient. The LASSO algorithm was applied to reduce the dimensionality of the data and select features. A multivariable logistic regression analysis was adopted to build radiomics signatures. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the predictive accuracy of these models. Results There was no significant difference between the training and validation datasets for any clinical factors in patients with ESCC. The PD-L1 expression level correlated with the differentiation degree (p= 0.011) and tumor stage (p= 0.032). Smoking status (p= 0.043) and differentiation degree (p= 0.025) were associated with CD8+TILs expression levels. The radiomics signatures achieved good performance in predicting PD-L1 and CD8+TILs with AUCs= 0.784 and 0.764, respectively. The combined model showed a favorable predictive ability compared to radiomics signatures or clinical factors alone and improved the AUCs from 0.669 to 0.871 for PD-L1 and from 0.672 to 0.832 for CD8+TILs. These results were verified in the validation dataset with the AUCs of 0.817 and 0.795, respectively. Conclusion CT-based radiomics features have a potential value for classifying patients according to PD-L1 and CD8+TILs expression levels. The combination of clinical factors and radiomics signatures significantly improved the predictive performance in ESCC.
Collapse
Affiliation(s)
- Qiang Wen
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan 250021, People's Republic of China
| | - Zhe Yang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan 250021, People's Republic of China
| | - Jian Zhu
- Department of Radiation Physics, Shandong Cancer Hospital and Institute, Shandong First Medical University, Jinan 250117, People's Republic of China
| | - Qingtao Qiu
- Department of Radiation Physics, Shandong Cancer Hospital and Institute, Shandong First Medical University, Jinan 250117, People's Republic of China
| | - Honghai Dai
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan 250021, People's Republic of China
| | - Alei Feng
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan 250021, People's Republic of China
| | - Ligang Xing
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Jinan 250117, People's Republic of China
| |
Collapse
|
195
|
Wei L, Wang B, Hu L, Xu Y, Li Z, Shen Y, Huang H. MEX3A is upregulated in esophageal squamous cell carcinoma (ESCC) and promotes development and progression of ESCC through targeting CDK6. Aging (Albany NY) 2020; 12:21091-21113. [PMID: 33188661 PMCID: PMC7695430 DOI: 10.18632/aging.103196] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 04/13/2020] [Indexed: 02/07/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most commonly diagnosed malignant tumors worldwide and identified as a serious threat to human health. The role of MEX3A in ESCC remains unclear. In this study, we found that MEX3A was upregulated in tumor tissues of ESCC and positively associated with more advanced tumor stage, higher risk of lymphatic metastasis and poor prognosis. The downregulation of MEX3A in ESCC cell lines could induce inhibition of cell proliferation, colony formation, cell migration, and the promotion of cell apoptosis, while MEX3A overexpression exhibited opposite effects. In vivo experiments also verified the inhibition of ESCC induced by MEX3A knockdown. Moreover, we identified CDK6 as a potential target of MEX3A, which was also upregulated in ESCC. Further studies demonstrated that knockdown of CDK6 showed similar effects on the development of ESCC with MEX3A. More importantly, it was illustrated that CDK6 knockdown could alleviate the promotion effects of MEX3A overexpression on ESCC. In conclusion, MEX3A was identified as a tumor promotor in the development and progression of ESCC by targeting CDK6, which may be considered as a novel prognostic indicator and therapeutic target in treatment of ESCC.
Collapse
Affiliation(s)
- Lei Wei
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing 210002, China
| | - Bo Wang
- Department of Thoracic Surgery, Nanjing Chest Hospital, Nanjing 210029, China
| | - Liwen Hu
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing 210002, China
| | - Yang Xu
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing 210002, China
| | - Zhongdong Li
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing 210002, China
| | - Yi Shen
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing 210002, China
| | - Hairong Huang
- Department of Cardiothoracic Surgery, Jinling Hospital, Nanjing 210002, China
| |
Collapse
|
196
|
Predicting the risk of esophageal high-grade lesions in opportunistic screening. Gastrointest Endosc 2020; 92:1136-1137. [PMID: 33160493 DOI: 10.1016/j.gie.2020.05.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 02/08/2023]
|
197
|
Liu T, Du LT, Wang YS, Gao SY, Li J, Li PL, Sun ZW, Binang H, Wang CX. Development of a Novel Serum Exosomal MicroRNA Nomogram for the Preoperative Prediction of Lymph Node Metastasis in Esophageal Squamous Cell Carcinoma. Front Oncol 2020; 10:573501. [PMID: 33123480 PMCID: PMC7573187 DOI: 10.3389/fonc.2020.573501] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Preoperative prediction of lymph node (LN) metastasis is accepted as a crucial independent risk factor for treatment decision-making for esophageal squamous cell carcinoma (ESCC) patients. Our study aimed to establish a non-invasive nomogram to identify LN metastasis preoperatively in ESCC patients. Construction of the nomogram involved three sequential phases with independent patient cohorts. In the discovery phase (N = 20), LN metastasis-associated microRNAs (miRNAs) were selected from next-generation sequencing (NGS) assay of human ESCC serum exosome samples. In the training phase (N = 178), a nomogram that incorporated exosomal miRNA model and clinicopathologic was developed by multivariate logistic regression analysis to preoperatively predict LN status. In the validation phase (n = 188), we validated the predicted nomogram's calibration, discrimination, and clinical usefulness. Four differently expressed miRNAs (chr 8-23234-3p, chr 1-17695-5p, chr 8-2743-5p, and miR-432-5p) were tested and selected in the serum exosome samples from ESCC patients who have or do not have LN metastasis. Subsequently, an optimized four-exosomal miRNA model was constructed and validated in the clinical samples, which could effectively identify ESCC patients with LN metastasis, and was significantly superior to preoperative computed tomography (CT) report. In addition, a clinical nomogram consisting of the four-exosomal miRNA model and CT report was established in training cohort, which showed high predictive value in both training and validation cohorts [area under the receiver operating characteristic curve (AUC): 0.880 and 0.869, respectively]. The Hosmer–Lemeshow test and decision curve analysis implied the nomogram's clinical applicability. Our novel non-invasive nomogram is a robust prediction tool with promising clinical potential for preoperative LN metastasis prediction of ESCC patients, especially in T1 stage.
Collapse
Affiliation(s)
- Tong Liu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lu-Tao Du
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Yun-Shan Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Shan-Yu Gao
- Department of Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Pei-Long Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Zhao-Wei Sun
- Department of Surgery, The Affiliated Hospital of Medical College Qingdao University, Qingdao, China
| | - Helen Binang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chuan-Xin Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| |
Collapse
|
198
|
Deng L, Zhu X, Yu Z, Li Y, Qin L, Liu Z, Feng L, Guo R, Zheng Y. Novel T7-Modified pH-Responsive Targeted Nanosystem for Co-Delivery of Docetaxel and Curcumin in the Treatment of Esophageal Cancer. Int J Nanomedicine 2020; 15:7745-7762. [PMID: 33116498 PMCID: PMC7553263 DOI: 10.2147/ijn.s257312] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/16/2020] [Indexed: 12/16/2022] Open
Abstract
Background Although single-drug chemotherapy is still an effective treatment for esophageal cancer, its long-term application is limited by severe side-effects, poor bioavailability, and drug-resistance. Increasing attention has been paid to nanomedicines because of their good biological safety, targeting capabilities, and high-efficiency loading of multiple drugs. Herein, we have developed a novel T7 peptide-modified pH-responsive targeting nanosystem co-loaded with docetaxel and curcumin for the treatment of esophageal cancer. Methods Firstly, CM-β-CD-PEI-PEG-T7/DTX/CUR (T7-NP-DC) was synthesized by the double emulsion (W/O/W) method. The targeting capacity of the nanocarrier was then investigated by in vitro and in vivo assays using targeted (T7-NP) and non-targeted nanoparticles (NP). Furthermore, the anti-tumor efficacy of T7-NP-DC was studied using esophageal cancer cells (KYSE150 and KYSE510) and a KYSE150 xenograft tumor model. Results T7-NP-DC was synthesized successfully and its diameter was determined to be about 100 nm by transmission electron microscopy and dynamic light scattering. T7-NP-DC with docetaxel and curcumin loading of 10% and 6.1%, respectively, had good colloidal stability and exhibited pH-responsive drug release. Good biosafety was observed, even when the concentration was as high as 800 μg/mL. Significant enhancement of T7-NP uptake was observed 6 hours after intravenous injection compared with NP. In addition, the therapeutic efficacy of T7-NP-DC was better than NP-DC and docetaxel in terms of growth suppression in the KYSE150 esophageal cancer model. Conclusion The findings demonstrated that T7-NP-DC is a promising, non-toxic, and controllable nanoparticle that is capable of simultaneous delivery of the chemotherapy drug, docetaxel, and the Chinese Medicine, curcumin, for treatment of esophageal cancer. This novel T7-modified targeting nanosystem releases loaded drugs when exposed to the acidic microenvironment of the tumor and exerts a synergistic anti-tumor effect. The data indicate that the nanomaterials can safely exert synergistic anti-tumor effects and provide an excellent therapeutic platform for combination therapy of esophageal cancer.
Collapse
Affiliation(s)
- Lian Deng
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Xiongjie Zhu
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Zhongjian Yu
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Ying Li
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Lingyu Qin
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Zhile Liu
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| | - Longbao Feng
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Rui Guo
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yanfang Zheng
- Department of Oncology, Zhujiang Hospital of Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
199
|
Zhang H, Zhao H, He X, Xi F, Liu J. JAK-STAT Domain Enhanced MUC1-CAR-T Cells Induced Esophageal Cancer Elimination. Cancer Manag Res 2020; 12:9813-9824. [PMID: 33116840 PMCID: PMC7549884 DOI: 10.2147/cmar.s264358] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/31/2020] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Chimeric antigen receptor (CAR)-T cells have shown to play a vital role in anti-tumor functions in hematological malignancies, but have poor efficacy in solid tumors. To improve the activation and proliferation of CAR-T cell in solid tumors, we constructed an enhanced CAR-T cells to increase the survival of esophageal cancer. MATERIALS AND METHODS To construct enhanced CAR-T cells, we chose MUC1 as the target of CAR-T cells. Long-term co-culture of target cells and effector cells was applied to verify the antitumor activity of these enhanced MUC1-CAR-T cells in vitro. Moreover, a mouse xenograft model was established to investigate the effects of enhanced MUC1-CAR-T cells on tumor elimination in vivo. RESULTS In vitro studies showed that enhanced MUC1-CAR-T cells have long-lasting tumor killing and proliferative capabilities. Moreover, animal experiments verified that enhanced MUC1-CAR-T cells had significant antitumor function and a prolonged half-life by subcutaneous transplantation models of esophageal cancer and PDX models of esophageal cancer, in vivo. CONCLUSION These results indicated that enhanced MUC1-CAR-T cells have a significant cytotoxic effect on esophageal cancer, and may likely to provide a novel strategy for the treatment of esophageal cancer.
Collapse
Affiliation(s)
- Heng Zhang
- School of Public Health, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, People’s Republic of China
| | - Hui Zhao
- Department of Radiation Therapy, Xinjiang Uygur Autonomous Region People’s Hospital, Urumqi, Xinjiang Uygur Autonomous Region, People’s Republic of China
| | - Xiaolei He
- Department of Hepatology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, People’s Republic of China
| | - Feng Xi
- Medical Department, Xinjiang Uygur Autonomous Region People’s Hospital, Urumqi, Xinjiang Uygur Autonomous Region, People’s Republic of China
| | - Jiwen Liu
- School of Public Health, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region, People’s Republic of China
| |
Collapse
|
200
|
Yu D, Ma Y, Feng C, Ma Z, Guo J, Chen H, He T, Guo J, Sun X, Qin Q, Sun X, Ma J. PBX1 Increases the Radiosensitivity of Oesophageal Squamous Cancer by Targeting of STAT3. Pathol Oncol Res 2020; 26:2161-2168. [PMID: 32170580 DOI: 10.1007/s12253-020-00803-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/04/2020] [Indexed: 10/24/2022]
Abstract
The radioresistance of oesophageal squamous cell carcinoma (OSCC) is a critical factor leading to a poor prognosis among patients. The expression of PBX1 is abnormally high in a broad range of human tissues, and this gene plays a key role in tumour proliferation. This research intended to explore the radiosensitization of OSCC by silencing PBX1. The OSCC cell lines KYSE450 and KYSE150 were subjected to PBX1 silencing and/or irradiation (IR). Cell proliferation, colony formation, and apoptosis were tested to evaluate the radiosensitization ability of PBX1 silencing. The levels of STAT3 and p-STAT3 in the OSCC cells were tested by Western blotting. Furthermore, KYSE150 cells with or without PBX1 silencing were xenografted into nude mice with or without radiation exposure. Concomitant PBX1 silencing and IR can obviously suppress growth and enhance radiosensitivity in OSCC cells and xenografts. Moreover, the downregulation of PBX1 inhibits the expression of STAT3 and p-STAT3. The downregulation of PBX1 may increase radiosensitivity in OSCC cells and xenografts via the PBX1/STAT3 pathway. Our findings demonstrate that PBX1 may be a potential target for promoting the effect of radiation therapy in OSCC patients.
Collapse
Affiliation(s)
- Dingyue Yu
- Department of radiotherapy, The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, 57 Zhonghua West Road, Lianyungang, 222042, Jiangsu Province, China
| | - Yuanyuan Ma
- Department of radiotherapy, The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, 57 Zhonghua West Road, Lianyungang, 222042, Jiangsu Province, China
| | - Chen Feng
- Oncology Department, First Hospital of Bengbu Medical College, Bengbu, 233000, AnHui, China
| | - Zhiyu Ma
- Department of radiotherapy, The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, 57 Zhonghua West Road, Lianyungang, 222042, Jiangsu Province, China
| | - Jiayou Guo
- Department of radiotherapy, The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, 57 Zhonghua West Road, Lianyungang, 222042, Jiangsu Province, China
| | - Hui Chen
- Department of radiotherapy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Tianli He
- Department of Respiratory, Changxing People's Hospital, Huzhou, 313000, Zhejiang, China
| | - Jiayi Guo
- Department of radiotherapy, The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, 57 Zhonghua West Road, Lianyungang, 222042, Jiangsu Province, China
| | - Xingbang Sun
- Department of radiotherapy, The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, 57 Zhonghua West Road, Lianyungang, 222042, Jiangsu Province, China
| | - Qin Qin
- Department of radiotherapy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Xinchen Sun
- Department of radiotherapy, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Jianxin Ma
- Department of radiotherapy, The Lianyungang municipal oriental Hospital Affiliated to Bengbu Medical College, 57 Zhonghua West Road, Lianyungang, 222042, Jiangsu Province, China.
| |
Collapse
|