1
|
Duan L, Cao S, Zhao F, Du X, Gao Z, Wang X, Bian F. Effects of FAP+ fibroblasts on cell proliferation migration and immunoregulation of esophageal squamous carcinoma cells through the CXCL12/CXCR4 axis. Mol Cell Biochem 2025:10.1007/s11010-025-05226-x. [PMID: 39934460 DOI: 10.1007/s11010-025-05226-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Cancer-associated fibroblasts (CAFs) secrete and synthesize fibroblast activation protein (FAP), which could promote proliferation and immunosuppression of multiple cancers including esophageal squamous cell carcinoma (ESCC). CXCL12/CXCR4 signaling could be revitalized by CAFs in cancer cells. Nevertheless, the significance of this interaction in ESCC has yet to be elucidated. Herein, we investigated whether FAP+ CAF cells could promote ESCC cells proliferation, migration and regulate immunity through the CXCL12/CXCR4 pathway in vitro and in vivo. The protein expression level of FSP1, FAP, CD8+ and Ki-67 in different sample was estimated by IHC and western blot. qPCR was used to quantify the mRNA level of FSP1, FAP, CD8+ and Ki-67 in different sample. The cell viability, proliferation, migration and invasion of different sample were evaluated by CCK-8, EdU staining, wound healing assay and Transwell assay, respectively. The ELISA was carried out to measure the protein level of IFN-γ, TNF-α, GZMB and IL-2. ESCC xenograft mice model was established to assess the impact of FAP+ CAF. FSP1, FAP, CD8+ and Ki-67 are greatly up-regulated in hESCC tissues. Through CXCL12/CXCR4 axis, FAP-positive CAF was capable of promoting the cell proliferation, migration and invasion of ESCC tumor cells and preventing the CD8+ T cells from secreting cytokine. Blocking this signaling with selective CXCR4 antagonist could counteract the effects caused by high-expression of FAP. FAP+ CAFs could inhibit the occurrence and development of tumors. These results indicated that FAP-positive CAF have an impact on cell proliferation migration and immunoregulation of ESCC through the CXCL12/CXCR4 axis.
Collapse
Affiliation(s)
- Lijuan Duan
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China.
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China.
| | - Shasha Cao
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China
| | - Fang Zhao
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China
| | - Xianjuan Du
- Department of Pathology, Anyang Cancer Hospital, Anyang, 455000, Henan Province, People's Republic of China
| | - Zhaowei Gao
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
- Henan Provincial Key Medical Laboratory of Precise Prevention and Treatment of Esophageal Cancer, Anyang, 455000, Henan Province, People's Republic of China
| | - Xiaoxiao Wang
- Central Laboratory, Anyang Cancer Hospital, No.1, Huanbin North Road, Beiguan District, Anyang, 455000, Henan Province, People's Republic of China
| | - Fang Bian
- Department of Pathology, Anyang Cancer Hospital, Anyang, 455000, Henan Province, People's Republic of China
| |
Collapse
|
2
|
Zhan T, Betge J, Schulte N, Dreikhausen L, Hirth M, Li M, Weidner P, Leipertz A, Teufel A, Ebert MP. Digestive cancers: mechanisms, therapeutics and management. Signal Transduct Target Ther 2025; 10:24. [PMID: 39809756 PMCID: PMC11733248 DOI: 10.1038/s41392-024-02097-4] [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: 06/29/2024] [Revised: 10/20/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Cancers of the digestive system are major contributors to global cancer-associated morbidity and mortality, accounting for 35% of annual cases of cancer deaths. The etiologies, molecular features, and therapeutic management of these cancer entities are highly heterogeneous and complex. Over the last decade, genomic and functional studies have provided unprecedented insights into the biology of digestive cancers, identifying genetic drivers of tumor progression and key interaction points of tumor cells with the immune system. This knowledge is continuously translated into novel treatment concepts and targets, which are dynamically reshaping the therapeutic landscape of these tumors. In this review, we provide a concise overview of the etiology and molecular pathology of the six most common cancers of the digestive system, including esophageal, gastric, biliary tract, pancreatic, hepatocellular, and colorectal cancers. We comprehensively describe the current stage-dependent pharmacological management of these malignancies, including chemo-, targeted, and immunotherapy. For each cancer entity, we provide an overview of recent therapeutic advancements and research progress. Finally, we describe how novel insights into tumor heterogeneity and immune evasion deepen our understanding of therapy resistance and provide an outlook on innovative therapeutic strategies that will shape the future management of digestive cancers, including CAR-T cell therapy, novel antibody-drug conjugates and targeted therapies.
Collapse
Affiliation(s)
- Tianzuo Zhan
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johannes Betge
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nadine Schulte
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lena Dreikhausen
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Hirth
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Moying Li
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Philip Weidner
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Antonia Leipertz
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Andreas Teufel
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P Ebert
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- DKFZ Hector Cancer Institute at University Medical Center Mannheim, Mannheim, Germany.
- Mannheim Cancer Center, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
3
|
Yuan X, Chen J, Shi D, Song J, Wang P, Cheng D, Yang C, Qiu X, Zhai C. Advanced esophageal cancer with bone metastases: Prognostic biomarkers and palliative treatment. Heliyon 2024; 10:e23510. [PMID: 38170113 PMCID: PMC10758821 DOI: 10.1016/j.heliyon.2023.e23510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/16/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
Esophageal cancer (EC) is a common and devastating tumor of the upper digestive tract. Unfortunately, by the time any symptoms have manifested, the disease has often progressed to an advanced stage and is accompanied by macro- and micrometastases, including in the bones. The treatment of esophageal cancer with bone metastases remains clinically challenging, given the poor prognosis associated with this condition. Effective prognostic biomarkers can help medical staff choose the appropriate operation and treatment plan, that is for most beneficial for making patients. Current treatments for esophageal cancer with bone metastases include pain-relieving drugs, surgical therapy, radiotherapy (RT), chemotherapy (CT, including molecular-targeted drug therapy), endocrine therapy (ET), bisphosphonates (BPs) and interventional therapy. Of these robust measures, radiotherapy has emerged as a particularly promising therapy for bone metastases from esophageal cancer. Substantial progress has been made in radiation therapy techniques since the discovery of X-rays by Roentgen in 1895. In its palliative capacity, the key goals of radiotherapy are to relieve the patients' bone pain and debilitate effects, including relieving spinal cord compression, correcting the spinal deformity and restoring spinal stability. However, it is worth mentioning that RT for esophageal cancer has various side effects. Currently, the available studies focused exclusively on radiotherapy for ECBM are too small to draw any definitive conclusions, and each of these studies has significant limitations. In this review, in addition to the epidemiology described at the beginning, we will explore the current prognostic biomarkers and radiotherapy for esophageal cancer, with a particular focus on those with bone metastases.
Collapse
Affiliation(s)
- Xiaofeng Yuan
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jun Chen
- Department of Orthopedics, Yixing People's Hospital, Yixing, China
| | - Dingsen Shi
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jiaxun Song
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Pu Wang
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dong Cheng
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Cheng Yang
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xubin Qiu
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Chenjun Zhai
- Department of Orthopedics, Yixing People's Hospital, Yixing, China
| |
Collapse
|
4
|
Darvish M. LncRNA FTH1P3: A New Biomarker for Cancer-Related Therapeutic Development. Curr Mol Med 2024; 24:576-584. [PMID: 37491858 DOI: 10.2174/1566524023666230724141353] [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: 04/15/2023] [Revised: 06/17/2023] [Accepted: 06/22/2023] [Indexed: 07/27/2023]
Abstract
Cancer is a persistent and urgent health problem that affects the entire world. Not long ago, regulatory biomolecules referred to as long noncoding RNAs (lncRNAs) might have value for their innate abundance and stability. These single-stranded RNAs potentially interfere with several physiological and biochemical cellular processes involved in many human pathological situations, particularly cancer diseases. Ferritin heavy chain1 pseudogene 3 (FTH1P3), a lncRNA that is ubiquitously transcribed and belongs to the ferritin heavy chain (FHC) family, represents a novel class of lncRNAs primarily found in oral squamous cell carcinoma. Further research has shown that FTH1P3 is involved in other malignancies such as uveal melanoma, glioma, esophageal squamous cell carcinoma, non-small cell lung cancer, breast cancer, laryngeal squamous cell carcinoma, and cervical cancer. Accordingly, FTH1P3 significantly enhances cancer symptoms, including cell proliferation, invasion, metastasis, chemoresistance, and inhibition of apoptosis through many specific mechanisms. Notably, the clinical data significantly demonstrated the association of FTH1P3 overexpression with poor prognosis and poor overall survival within the examined samples. Here, we summarize all the research published to date (13 articles) on FTH1P3, focusing on the biological function underlying the regulatory mechanism and its possible clinical relevance.
Collapse
Affiliation(s)
- Maryam Darvish
- Department of Medical Biotechnology, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| |
Collapse
|
5
|
Wang L, Zhao QF, Yang BB, Liang HJ, Zhang XE, Zhang XY, Yang WJ, Guo ZY, Xu X, Tian F, Wu QH. STAT3-EphA7 axis contributes to the progression of esophageal squamous cell carcinoma. Acta Oncol 2023; 62:1757-1766. [PMID: 37738252 DOI: 10.1080/0284186x.2023.2259601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023]
Abstract
BACKGROUND Our previous study has revealed that EphA7 was upregulated in patient-derived esophageal squamous cell carcinoma (ESCC) xenografts with hyper-activated STAT3, but its mechanism was still unclear. MATERIALS AND METHODS To assess the association between EphA7 and STAT3, western blotting, immunofluorescence, ChIP assay, and qRT-PCR were conducted. Truncated mutation and luciferase assay were performed to examine the promoter activity of EphA7. CCK-8 assay and colony formation were performed to assess the proliferation of ESCC. Cell-derived xenograft models were established to evaluate the effects of EphA7 on ESCC tumor growth. RNA-seq analyses were used to assess the effects of EphA7 on related signals. RESULTS In this study, EphA7 was found upregulated in ESCC cell lines with high STAT3 activation, and immunofluorescence also showed that EphA7 was co-localized with phospho-STAT3 in ESCC cells. Interestingly, suppressing STAT3 activation by the STAT3 inhibitor Stattic markedly inhibited the protein expression of EphA7 in ESCC cells, in contrast, activation of STAT3 by IL-6 obviously upregulated the protein expression of EphA7. Moreover, the transcription of EphA7 was also mediated by the activation of STAT3 in ESCC cells, and the -2000∼-1500 region was identified as the key promoter of EphA7. Our results also indicated that EphA7 enhanced the cell proliferation of ESCC, and silence of EphA7 significantly suppressed ESCC tumor growth. Moreover, EphA7 silence markedly abolished STAT3 activation-derived cell proliferation of ESCC. Additionally, RNA-seq analyses indicated that several tumor-related signaling pathways were significantly changed after EphA7 downregulation in ESCC cells. CONCLUSION Our results showed that the transcriptional expression of EphA7 was increased by activated STAT3, and the STAT3 signaling may act through EphA7 to promote the development of ESCC.
Collapse
Affiliation(s)
- Li Wang
- Department of Obstetrics and Gynecology, Center of Genetics and Prenatal Diagnosis, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Qiao-Feng Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
| | - Bing-Bing Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, P. R. China
| | - Hui-Jie Liang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
| | - Xian-E Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
- Second People's Hospital of Henan Province, Zhengzhou, P. R. China
| | - Xiao-Yan Zhang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
| | - Wan-Jing Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
| | - Zhi-Yu Guo
- SanQuan College of XinXiang Medical University, Xinxiang, P. R. China
| | - Xin Xu
- College of Biotechnology, Center for Self-Propelled Nanotechnologies, Suzhou Industrial Park Institute of Services Outsourcing, Suzhou, P. R. China
- Translational Cancer Research Laboratory, Suzhou Acumen Medical Technology, Suzhou, P. R. China
| | - Fang Tian
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
- Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, P. R. China
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, P. R. China
| | - Qing-Hua Wu
- Department of Obstetrics and Gynecology, Center of Genetics and Prenatal Diagnosis, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| |
Collapse
|
6
|
Pan J, Zang Y. LINC00667 Promotes Progression of Esophageal Cancer Cells by Regulating miR-200b-3p/SLC2A3 Axis. Dig Dis Sci 2022; 67:2936-2947. [PMID: 34313922 DOI: 10.1007/s10620-021-07145-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/30/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Recently, more and more evidence indicated that the long non-coding RNA was strictly related to the occurrence and progression of human cancers, including esophageal cancer (EC). We observed that LINC00667 was increased in EC, but the function of LINC00667 was unclear. Therefore, the function and potential molecular mechanism of LINC00667 in the progression of EC need to be further studied. METHODS Quantitative real-time PCR was used to investigate the levels of LINC00667, miR-200b-3p, and SLC2A3. The levels of protein involved in cell cycle, cell apoptosis, epithelial-mesenchymal transition, as well as SLC2A3 were quantitatived by western blot. The role of LINC00667 in the proliferative, migratory and invasive capabilities of EC cells were measured by cell counting kit-8 assay, EdU assay, flow cytometry assay, wound healing assay and transwell assay, respectively. Interaction between LINC00667 and miR-200b-3p or miR-200b-3p and SLC2A3 were confirmed using a luciferase reporter assay. RESULTS In this work, we found that LINC00667 expression was up-regulated in EC cell lines, and LINC00667 knockdown inhibited cell proliferation, migration, and invasion in EC cells. In addition, it showed that LINC00667 functioned as competitive endogenous RNA for miR-200b-3p by the DIANA-LncBase database. Moreover, we used targetscan online software to predict SLC2A3 as a target gene of miR-200b-3p. Subsequently, rescue experiments confirmed that knocking out SLC2A3 could reverse the inhibitory effect of miR-200b-3p on EC cells transfected with sh-LINC00667. CONCLUSION Herein, we revealed the novel mechanism of LINC00667 on regulating metastasis-related gene by sponge regulatory axis during EC metastasis. Our results demonstrated that LINC00667 plays a critical role in metastatic EC by mediating sponge regulatory axis miR-200b-3p/SLC2A3. To explore function of LINC00667/miR-200b-3p/SLC2A3 axis may provide an informative biomarker of malignancy and a highly selective anti-EC therapeutic target.
Collapse
Affiliation(s)
- Jindun Pan
- Department of Gastroenterology, Taishan Hospital, Taian, Shandong Province, China.
| | - Yunhong Zang
- Department of Gastroenterology, Taishan Hospital, Taian, Shandong Province, China
| |
Collapse
|
7
|
Yao L, Zhong X, Huang G, Ma Q, Xu L, Xiao H, Guo X. Investigation on the Potential Correlation Between TP53 and Esophageal Cancer. Front Cell Dev Biol 2021; 9:730337. [PMID: 34778250 PMCID: PMC8578720 DOI: 10.3389/fcell.2021.730337] [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: 06/24/2021] [Accepted: 09/30/2021] [Indexed: 12/24/2022] Open
Abstract
Background:TP53 family members play an indispensable role in various human cancers, while the gene expression profiles, prognostic value, and potential mechanism in esophageal cancer (ESCA) are yet unclear. Methods: The expression and roles of TP53 family members in ESCA were investigated using the Cancer Genome Atlas (TCGA), Tumor Immune Estimation Resource (TIMER), Kaplan–Meier plotter, gene set enrichment analysis (GSEA), and UALCAN databases. The expression of TP53 between ESCA and the corresponding adjacent tissues was validated using qRT-PCR. Furthermore, the effects of TP53 on esophageal squamous cell carcinoma (ESCC) cell migration and proliferation were examined using the Transwell assay, scratch test, and crystal violet assay. The correlation between TP53 and mTOR pathways was evaluated by Western blotting. Results: This study showed a correlation between high mRNA expression of TP53 members (TP53, TP63, and TP73) and clinical cancer stages and nodal metastasis status in ESCA patients. Moreover, the expression of TP53 was significantly associated with the overall survival (OS) of ESCA patients. Additional experiments verified that the mRNA of TP53 was upregulated in ESCC patients. Moreover, the downregulated expression of TP53 significantly retarded ESCC cell migration and proliferation and might activate the mTOR signaling pathway and inhibit TP53-dependent autophagy. Conclusion:TP53 has a prognostic value in ESCA and may be a leading factor in promoting ESCA pathogenesis.
Collapse
Affiliation(s)
- Lihua Yao
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaowu Zhong
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, China
| | - Guangcheng Huang
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Qiang Ma
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Lei Xu
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, China
| | - Hong Xiao
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiaolan Guo
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
8
|
Min Q, Wang Y, Wu Q, Li X, Teng H, Fan J, Cao Y, Fan P, Zhan Q. Genomic and epigenomic evolution of acquired resistance to combination therapy in esophageal squamous cell carcinoma. JCI Insight 2021; 6:150203. [PMID: 34494553 PMCID: PMC8492345 DOI: 10.1172/jci.insight.150203] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/21/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUNDTargeted arterial infusion of verapamil combined with chemotherapy (TVCC) is an effective clinical interventional therapy for esophageal squamous cell carcinoma (ESCC), but multidrug resistance (MDR) remains the major cause of relapse or poor prognosis, and the underlying molecular mechanisms of MDR, temporal intratumoral heterogeneity, and clonal evolutionary processes of resistance have not been determined.METHODSTo elucidate the roles of genetic and epigenetic alterations in the evolution of acquired resistance during therapies, we performed whole-exome sequencing on 16 serial specimens from 7 patients with ESCC at every cycle of therapeutic intervention from 3 groups, complete response, partial response, and progressive disease, and we performed whole-genome bisulfite sequencing for 3 of these 7 patients, 1 patient from each group.RESULTSPatients with progressive disease exhibited a substantially higher genomic and epigenomic temporal heterogeneity. Subclonal expansions driven by the beneficial new mutations were observed during combined therapies, which explained the emergence of MDR. Notably, SLC7A8 was identified as a potentially novel MDR gene, and functional assays demonstrated that mutant SLC7A8 promoted the resistance phenotypes of ESCC cell lines. Promoter methylation dynamics during treatments revealed 8 drug resistance protein-coding genes characterized by hypomethylation in promoter regions. Intriguingly, promoter hypomethylation of SLC8A3 and mutant SLC7A8 were enriched in an identical pathway, protein digestion and absorption, indicating a potentially novel MDR mechanism during treatments.CONCLUSIONOur integrated multiomics investigations revealed the dynamics of temporal genetic and epigenetic inter- and intratumoral heterogeneity, clonal evolutionary processes, and epigenomic changes, providing potential MDR therapeutic targets in treatment-resistant patients with ESCC during combined therapies.FUNDINGNational Natural Science Foundation of China, Science Foundation of Peking University Cancer Hospital, CAMS Innovation Fund for Medical Sciences, Major Program of Shenzhen Bay Laboratory, Guangdong Basic and Applied Basic Research Foundation, and the third round of public welfare development and reform pilot projects of Beijing Municipal Medical Research Institutes.
Collapse
Affiliation(s)
- Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xianfeng Li
- Department of Gastroenterology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Huajing Teng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiawen Fan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yiren Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Pingsheng Fan
- Department of Medical Oncology, Anhui Provincial Cancer Hospital, Hefei, China
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China
- Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Zhang H, Ju L, Hu P, Ye J, Yang C, Huang J. Circular RNA 0014715 Facilitates Cell Proliferation and Inhibits Apoptosis in Esophageal Squamous Cell Carcinoma. Cancer Manag Res 2021; 13:4735-4749. [PMID: 34163248 PMCID: PMC8214545 DOI: 10.2147/cmar.s314882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/01/2021] [Indexed: 12/24/2022] Open
Abstract
Background Circular RNAs (circRNAs) have recently been verified to have multiple biological functions and participate in diverse biological processes in different malignant tumors, including esophageal squamous cell carcinoma (ESCC). Nonetheless, the function of circular RNA 0014715 (hsa_circ_0014715, circ_0014715) in ESCC has not been described. Materials and Methods We investigated clinical data from sixty-seven patients undergoing surgery for esophageal cancer. The clinical data were collected. And we analyzed the correlation between the clinical characteristics of these patients and the expression of circ_0014715. Besides, we explored the expression of circ_0014715 in ESCC cell lines. We used cell counting kit-8, colony formation, transwell assay, and flow cytometry to detect changes in cell proliferation, migration, apoptosis, and cell cycle progression. Results We found that circ_0014715 was highly expressed in esophageal squamous cell carcinoma tissues and cell lines. The correlation analysis of clinicopathological features and gene expression revealed that high expression of circ_0014715 was related to nerve invasion, vascular invasion, more advanced tumor-node-metastasis (TNM) stage and poor differentiation grade. Receiver operating characteristic (ROC) curves revealed that circ_0014715 might have diagnostic value for ESCC. Experiments with cultured cells showed that knockdown of circ_0014715 significantly restrained cell proliferation, migration, invasion, wound healing and accelerated cell apoptosis. And cell cycle arrest at G2 phase was observed via flow cytometry. Overexpression of circ_0014715 caused the opposite effects. Collectively, these studies show that circ_0014715 is closely connected with the pathogenesis and development of ESCC. The excess expression of circ_0014715 may have promoting effects on the progression of esophageal cell carcinoma. Conclusion Our finding revealed that circ_0014715 promoted tumor growth and cell proliferation. All of these suggest that targeting circ_0014715 has potential therapeutic value in the treatment of ESCC.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, Jiangsu, People's Republic of China
| | - Linling Ju
- Nantong Institute of Liver Diseases, Nantong Third People's Hospital, Nantong University, Nantong, 226000, Jiangsu, People's Republic of China
| | - Peipei Hu
- Department of Pain Medicine, Nantong Hospital of Traditional Chinese Medicine, Nantong Hospital Affiliated to Nanjing University of Chinese Medicine, Nantong, 226000, Jiangsu, People's Republic of China
| | - Jun Ye
- Institute of Clinical Medicine, Taizhou People's Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, Jiangsu, People's Republic of China
| | - Canlin Yang
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, Jiangsu, People's Republic of China
| | - Junxing Huang
- Department of Oncology, Taizhou People's Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, 225300, Jiangsu, People's Republic of China
| |
Collapse
|
10
|
Zhou S, Guo Z, Zhou C, Zhang Y, Wang S. circ_NRIP1 is oncogenic in malignant development of esophageal squamous cell carcinoma (ESCC) via miR-595/SEMA4D axis and PI3K/AKT pathway. Cancer Cell Int 2021; 21:250. [PMID: 33957921 PMCID: PMC8101145 DOI: 10.1186/s12935-021-01907-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 04/07/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The hsa_circ_0004771 derived from NRIP1 (called circ_NRIP1) is a recently identified oncogenic circRNA. Here, we intended to investigate the role and mechanism of circ_NRIP1 in esophageal squamous cell carcinoma (ESCC), a prevalent and aggressive type of esophageal cancer. METHODS Expression of circ_NRIP1, miRNA-595-5p (miR-595) and semaphorin 4D (SEMA4D) was detected by RT-qPCR and western blotting. Cell growth was assessed by colony formation assay, MTS assay, flow cytometry, and xenograft experiment; migration and invasion were evaluated by transwell assay and western blotting. Dual-luciferase reporter assay identified the relationship among circ_NRIP1, miR-595 and SEMA4D. Western blotting measured phosphatidylinositol-3-hydroxykinase (PI3K)/AKT pathway-related proteins. RESULTS Expression of circ_NRIP1 was upregulated in ESCC tissues and cells. Knockdown of circ_NRIP1 could enhance apoptosis rate and E-cadherin expression, but suppress colony formation, cell viability, migration, invasion, and snail expression in KYSE30 and KYSE450 cells, as well as retarded tumor growth in mice. The suppressive role of circ_NRIP1 knockdown in cell growth, migration and invasion in vitro was abated by blocking miR-595; meanwhile, miR-595 overexpression elicited similar anti-tumor role in KYSE30 and KYSE450 cells, which was abrogated by restoring SEMA4D. Notably, circ_NRIP1 was a sponge for miR-595, and SEMA4D was a target of miR-595. Besides, PI3K/AKT signal was inhibited by circ_NRIP1 knockdown and/or miR-595 overexpression via indirectly or directly regulating SEMA4D. CONCLUSION circ_NRIP1 functioned as an oncogene in ESCC, and modulated ESCC cell growth, migration and invasion both in vitro and in vivo via targeting miR-595/SEMA4D axis and inhibiting PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Shifan Zhou
- Henan University of Chinese Medicine, No.156 Jinshui East Road, Zhengzhou, 450046, Henan, China. .,Department of Oncology, The Second Affiliated Hospital of Henan University of Chinese Medicine, No.6 Dongfeng Road, Jinshui District, Zhengzhou, 450002, Henan, China.
| | - Zhizhong Guo
- Department of Oncology, The Second Affiliated Hospital of Henan University of Chinese Medicine, No.6 Dongfeng Road, Jinshui District, Zhengzhou, 450002, Henan, China
| | - Chaofeng Zhou
- Department of Oncology, The Second Affiliated Hospital of Henan University of Chinese Medicine, No.6 Dongfeng Road, Jinshui District, Zhengzhou, 450002, Henan, China
| | - Yu Zhang
- Department of Oncology, The Second Affiliated Hospital of Henan University of Chinese Medicine, No.6 Dongfeng Road, Jinshui District, Zhengzhou, 450002, Henan, China
| | - Sai Wang
- Department of Oncology, The Second Affiliated Hospital of Henan University of Chinese Medicine, No.6 Dongfeng Road, Jinshui District, Zhengzhou, 450002, Henan, China
| |
Collapse
|
11
|
Yang J, Guo T, Liang X, Zhai Y, Cheng Y, Sun H, Cui Y, Cheng X. Cadmium inhibits apoptosis of human esophageal epithelial cells by upregulating CDK6. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2020; 205:111146. [PMID: 32827965 DOI: 10.1016/j.ecoenv.2020.111146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 07/22/2020] [Accepted: 08/06/2020] [Indexed: 06/11/2023]
Abstract
Cadmium (Cd) exposure in environment is associated with development of esophageal cancer. However, the mechanisms of Cd-induced carcinogenesis are still not been fully cleared, and the present study aimed to explore the possible etiological mechanism of Cd-induced esophageal cancer. Human esophageal epithelial cell lines (HET-1A and KYSE450) were treated with CdCl2 at 0.05 mg/l for 12, 24 h, and the then the apoptosis were detected using flow cytometry with annexin-V-FITC/PI staining. Results showed that apoptosis of treatment groups was significantly inhibited, and decreased reactive oxygen species (ROS) production played a key role in the inhibitory effects by upregulating Bcl-2 and downregulating Caspase-3/9. The relief of oxidative stress during Cd exposure was actively promoted by the increased nicotinamide adenine dinucleotide phosphoric acid and glutathione levels. To investigate the causes of enhanced intracellular antioxidant capacity, the activity of pyruvate kinase (PK), a key enzyme of glycolysis, was detected. Our results showed that PK activity was inhibited, suggesting that glycolysis process was blocked which promoted more intermediate metabolites of glycolysis to be used for reduced nicotinamide adenine dinucleotide phosphoric acid (NADPH) or other antioxidants synthesis. PK activity was closely correlated with phosphorylation of pyruvate kinase M2 (PKM2), and a highly negative correlation (correlation coefficients: -0.835, p < 0.05) between them was found. Western blotting showed the overphosphorylation of PKM2 in Cd-exposed cells, resulting from increased expression of cyclin-dependent kinases 6 (CDK6). These results suggested a possible mechanism of carcinogenic: Cd-induced upregulation of CDK6 in esophageal cell lines caused PKM2 overphosphorylation inhibiting PK activity, thereby shunting glucose-derived carbon into the pentose phosphate pathway and promoting the production of NADPH and reduced glutathione (GSH) to neutralize ROS, which finally results in the inhibited apoptosis.
Collapse
Affiliation(s)
- Jian Yang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China; Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, PR China
| | - Tianyi Guo
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, PR China; Department of Colorectal Anorectum, Shanxi Provincial People's Hospital Affilicated of Shanxi Medical University, Taiyuan, PR China
| | - Xiao Liang
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, PR China; Department of Anatomy, Shanxi Medical University, Taiyuan, PR China; Department of Orthopedics, Second Hospital of Shanxi Medical University, Taiyuan, PR China
| | - Yuanfang Zhai
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, PR China; Department of Anatomy, Shanxi Medical University, Taiyuan, PR China
| | - Yikun Cheng
- College of Letter & Science, University of California Berkeley, Berkeley, USA
| | - Hui Sun
- Institute of Science & Technology of Shanxi, Taiyuan, PR China
| | - Yongping Cui
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China; Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, PR China.
| | - Xiaolong Cheng
- Department of Pathology & Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, PR China; Department of Anatomy, Shanxi Medical University, Taiyuan, PR China.
| |
Collapse
|
12
|
Yu J, Wu X, Lv M, Zhang Y, Zhang X, Li J, Zhu M, Huang J, Zhang Q. A model for predicting prognosis in patients with esophageal squamous cell carcinoma based on joint representation learning. Oncol Lett 2020; 20:387. [PMID: 33193847 PMCID: PMC7656101 DOI: 10.3892/ol.2020.12250] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 08/25/2020] [Indexed: 12/31/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the deadliest cancer types with a poor prognosis due to the lack of symptoms in the early stages and a delayed diagnosis. The present study aimed to identify the risk factors significantly associated with prognosis and to search for novel effective diagnostic modalities for patients with early-stage ESCC. mRNA and methylation data of patients with ESCC and the corresponding clinical information were downloaded from The Cancer Genome Atlas (TCGA) database, and the representation features were screened using deep learning autoencoder. The univariate Cox regression model was used to select the prognosis-related features from the representation features. K-means clustering was used to cluster the TCGA samples. Support vector machine classifier was constructed based on the top 75 features mostly associated with the risk subgroups obtained from K-means clustering. Two ArrayExpress datasets were used to verify the reliability of the obtained risk subgroups. The differentially expressed genes and methylation genes (DEGs and DMGs) between the risk subgroups were analyzed, and pathway enrichment analysis was performed. A total of 500 representation features were produced. Using K-means clustering, the TCGA samples were clustered into two risk subgroups with significantly different overall survival rates. Joint multimodal representation strategy, which showed a good model fitness (C-index=0.760), outperformed early-fusion autoencoder strategy. The joint representation learning-based classification model had good robustness. A total of 1,107 DEGs and 199 DMGs were screened out between the two risk subgroups. The DEGs were involved in 70 pathways, the majority of which were correlated with metastasis and proliferation of various cancer types, including cytokine-cytokine receptor interaction, cell adhesion molecules PPAR signaling pathway, pathways in cancer, transcriptional misregulation in cancer and ECM-receptor interaction pathways. The two survival subgroups obtained via the joint representation learning-based model had good robustness, and had prognostic significance for patients with ESCC.
Collapse
Affiliation(s)
- Jun Yu
- Department of Molecular Biology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Xuanwu, Nanjing 210009, P.R. China
| | - Xiaoliu Wu
- Department of Molecular Biology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Xuanwu, Nanjing 210009, P.R. China
| | - Min Lv
- Department of Pathology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Xuanwu, Nanjing 210009, P.R. China
| | - Yuanying Zhang
- Department of Molecular Biology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Xuanwu, Nanjing 210009, P.R. China
| | - Xiaomei Zhang
- Department of Molecular Biology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Xuanwu, Nanjing 210009, P.R. China
| | - Jintian Li
- Department of Molecular Biology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Xuanwu, Nanjing 210009, P.R. China
| | - Ming Zhu
- Department of Molecular Biology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Xuanwu, Nanjing 210009, P.R. China
| | - Jianfeng Huang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Xuanwu, Nanjing 210009, P.R. China
| | - Qin Zhang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and The Affiliated Cancer Hospital of Nanjing Medical University, Xuanwu, Nanjing 210009, P.R. China
| |
Collapse
|
13
|
Abstract
As a new kind of RNA, circular RNA (circRNA) is a endogenous non-coding RNA with circular structure, which has the characteristics of universality, stability, conservatism and specificity. CircRNA can specifically bind to microRNAs (miRNAs) in the form of competitive endogenous RNA, thus directly or indirectly regulating the expression of related genes. In addition to the role of sponge, circRNA also regulates parental gene expression, transcriptional translation and protein modification; and it can be used as a biomarker to develop potential diagnosis and treatment methods and evaluate prognosis. Due to changes in dietary habits and genetic factors, the morbidity and mortality of esophageal cancer (EC) in the world are still high, and are prone to early metastasis. Although the diagnosis and treatment techniques have been improved in recent years, the early diagnosis of EC is not common, and the 5-year survival rate of patients is still very low. This article reviews the function and significance of circRNA and discusses the research progress of circRNA as biomarkers in EC.
Collapse
|
14
|
Lu P, Gu J, Zhang N, Sun Y, Wang J. Risk factors for precancerous lesions of esophageal squamous cell carcinoma in high-risk areas of rural China: A population-based screening study. Medicine (Baltimore) 2020; 99:e21426. [PMID: 32756148 PMCID: PMC7402764 DOI: 10.1097/md.0000000000021426] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although many studies in China have found that environmental or lifestyle factors are major contributors to the etiology of esophageal cancer, most of the patients in the above studies are in the middle and late stages, the early-stage patients account for a small proportion. To clarify the risk/protective factors contributing to early lesions, we conducted the present cross-sectional study.A total of 2925 healthy controls and 402 patients with esophageal precancerous lesions were included in our study by endoscopic examination. Information on risk/protective factors was collected by personal interview, and unconditional logistic regression was used to determine adjusted odds ratios (AORs) by the maximum-likelihood method.Smoking >20 pack-years (AOR = 1.48), duration of drinking >30 years (AOR = 1.40), alcohol consumption >100 mL/d (AOR = 1.44), gastroesophageal reflux disease (AOR = 1.75), esophagitis (AOR = 1.25), a family history of esophageal cancer (AOR = 1.92), or stomach cancer (AOR = 1.92) were significant risk factors for esophageal precancerous lesions. There was a negative correlation between abdominal obesity and early esophageal cancer and precancerous lesions (AOR = 0.75). In addition, we found that there was a synergistic effect between a family history of esophageal cancer and drinking (AOR = 3.00) and smoking (AOR = 2.90).Lifestyle risk factors, genetic factors, and upper gastrointestinal diseases are associated with the development of esophageal precancerous lesions. These results highlight the need for primary prevention to reduce the future burden of cancer and other chronic diseases in high-risk areas of rural China.
Collapse
Affiliation(s)
- Peipei Lu
- School of Medicine and Life Sciences, Shandong Academy of Medical Sciences, University of Jinan
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan
| | - Jianhua Gu
- Office of National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer
- Department of Epidemiology Cancer Institute and Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Nan Zhang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan
| | - Yawen Sun
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan
| | - Jialin Wang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan
| |
Collapse
|
15
|
Liu M, An H, Zhang Y, Sun W, Cheng S, Wang R, Wang X, Feng L. Molecular analysis of Chinese oesophageal squamous cell carcinoma identifies novel subtypes associated with distinct clinical outcomes. EBioMedicine 2020; 57:102831. [PMID: 32580137 PMCID: PMC7317223 DOI: 10.1016/j.ebiom.2020.102831] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/20/2020] [Accepted: 05/27/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Oesophageal squamous cell carcinoma (ESCC) is a highly heterogeneous cancer with a distinct incidence and prognosis. Molecular events driving ESCC subtypes and prognosis have not been established, and little is known regarding Chinese ESCC patients in Xinjiang, China. METHODS Here, we first integrated the genomic and transcriptomic data of 125 Chinese ESCC patients from Xinjiang Tumor Hospital (Urumqi, China). Two independent datasets of GSE53624 and The Cancer Genome Atlas (TCGA) ESCC were used to confirm the results of this study. DNA mutation and overall survival (OS) were analysed independently in the Chinese ESCC cohorts. FINDINGS Genomic analyses revealed a consistent mutation signatures and discordance among mutated genes across the different ESCC cohorts. In addition, transcriptomic profiling identified three Chinese ESCC subtypes associated with clinical and molecular attributes, including patient survival, lymph node status and genetic profile. Moreover, Chinese ESCC subtypes have distinct metabolic, inflammatory, metastatic, and cell proliferation features and unique potential therapeutics. Furthermore, the expression of cell cycle- and/or cell proliferation-related genes was higher in cyclin D1 (CCND1)-amplified tumours than in CCND1-normal tumours from Chinese ESCC patients, suggesting that CCND1 amplification promoted cell proliferation. INTERPRETATION Our findings provide a framework to facilitate the rational categorization of ESCC in Chinese patients and a foundation for new therapies. FUNDING This study was supported by the Research Fund of Key Laboratory of Xinjiang oncology (Grant no.2017D04006) and the Outstanding Youth Science and technology training project fund of Xinjiang, China (Grant no. 2017Q058).
Collapse
Affiliation(s)
- Meng Liu
- Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi 830011, Xinjiang, China.
| | - Haiyin An
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.
| | - Yuan Zhang
- Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China.
| | - Wei Sun
- Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China.
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.
| | - Ruozheng Wang
- Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China; Key Laboratory of Cancer Immunotherapy and Radiotherapy, Chinese Academy of Medical Sciences, Urumqi 830011, Xinjiang, China.
| | - Xiyan Wang
- Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang, China.
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
16
|
Chen J, Yang HM, Zhou HC, Peng RR, Niu ZX, Kang CY. PRR11 and SKA2 promote the proliferation, migration and invasion of esophageal carcinoma cells. Oncol Lett 2020; 20:639-646. [PMID: 32565988 PMCID: PMC7285799 DOI: 10.3892/ol.2020.11615] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 04/03/2020] [Indexed: 01/09/2023] Open
Abstract
Proline-rich protein 11 (PRR11) together with its upstream adjacent gene, spindle and kinetochore associated 2 (SKA2), represent a classic, head-to-head gene pair. The role of the PRR11 and SKA2 gene pair has been described in various types of cancer, including breast cancer, non-small cell lung cancer, hepatocellular carcinoma and ovarian carcinoma. However, its role in esophageal carcinoma (ESCC) remains unclear. The mRNA expression levels of PRR11 and SKA2 were examined in ESCC surgical specimens. In addition, the role of PRR11 and SKA2 in the proliferation and migratory and invasive capacities of EC9706 and EC109 cell lines was examined. The results from the present study demonstrated that PRR11 and SKA2 expression levels were upregulated in ESCC tissues compared with adjacent normal tissues. Furthermore, PRRl1 and SKA2 knockdown significantly inhibited the proliferation and migratory and invasive capacities of ESCC cells. Conversely, PRRl1 and SKA2 overexpression significantly promoted the proliferation and migratory and invasive capacities of ESCC cell lines via activation of the AKT signaling pathway and certain markers of epithelial-mesenchymal transition, including Snail and N-cadherin. The results from the present study suggested that the PRR11 and SKA2 gene pair may represent a potential target in the diagnosis and treatment of ESCC.
Collapse
Affiliation(s)
- Jie Chen
- Department of Pathophysiology, Henan Medical College, Zhengzhou, Henan 450000, P.R. China
| | - Hong-Mei Yang
- Department of Pathophysiology, Henan Medical College, Zhengzhou, Henan 450000, P.R. China
| | - Hui-Chong Zhou
- Department of Gastroenterology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Rui-Rui Peng
- Department of Pathophysiology, Henan Medical College, Zhengzhou, Henan 450000, P.R. China
| | - Zhao-Xiao Niu
- Department of Pathophysiology, Henan Medical College, Zhengzhou, Henan 450000, P.R. China
| | - Chun-Yan Kang
- Department of Pathophysiology, Henan Medical College, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
17
|
Li X, Geng J, Ren Z, Xiong C, Li Y, Liu H. WAVE3 upregulation in esophageal squamous cell carcinoma and its effect on the migration of human esophageal cancer cell lines in vitro. Mol Med Rep 2020; 22:465-473. [PMID: 32377706 PMCID: PMC7248532 DOI: 10.3892/mmr.2020.11126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 03/31/2020] [Indexed: 12/22/2022] Open
Abstract
The present study aimed to investigate the role of Wiskott-Aldrich syndrome verprolin-homologous protein 3 (WAVE3) in the progression of esophageal squamous cell carcinoma (ESCC), and to explore its effect on the migration of esophageal cancer cell lines in vitro. The expression level of WAVE3 in ESCC tissues was determined via immunohistochemistry, and the relative levels of WAVE3 mRNA and micro (mi)RNA200b were assessed in the serum of patients with ESCC using reverse transcription-quantitative PCR (RT-qPCR). Following cell transfection, the levels of miRNA200b and WAVE3 protein were determined via RT-qPCR and western blot analysis, and cell migration was examined using a Transwell assay. Subsequently, the clinical parameters were used to analyze whether the expression of WAVE3 in tissues and serum was associated with the occurrence and development of ESCC. The results demonstrated that the expression of WAVE3 was increased in ESCC tissues compared with normal tissues. The results also revealed increased expression levels of WAVE3 and decreased expression levels of miRNA200b in the serum of patients with ESCC, compared with healthy volunteers. High expression of WAVE3 was significantly associated with tumor TNM stage, invasion depth and lymphatic invasion of ESCC. In cells transfected with miRNA200b mimic, the miRNA200b was overexpressed, WAVE3 protein was downregulated and cell migration ability was decreased. The results of the present study suggest that WAVE3 may serve as an oncogene in ESCC, and its inhibition via miRNA200b decreased tumor cell migration. Therefore, WAVE3 may serve as a novel biological marker and therapeutic target for ESCC.
Collapse
Affiliation(s)
- Xuebing Li
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jie Geng
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhenzhen Ren
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Chao Xiong
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yuqing Li
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hongchun Liu
- Department of Medical Laboratory, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
18
|
Zhang Z, Li X, Xiong F, Ren Z, Han Y. Hsa_circ_0012563 promotes migration and invasion of esophageal squamous cell carcinoma by regulating XRCC1/EMT pathway. J Clin Lab Anal 2020; 34:e23308. [PMID: 32185826 PMCID: PMC7439410 DOI: 10.1002/jcla.23308] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Recent reports have indicated that circular RNA (circRNA) may regulate tumorigenesis development. However, the function of circRNAs in esophageal squamous cell carcinoma (ESCC) is unclear. MATERIAL AND METHOD The RT-qPCR assay was performed to detect hsa_circ_0012563 expression in ESCC tissues and cell lines. Then, the MTT assay, colony formation assay, flow cytometric assay, and cell migration and invasion assay were performed to examine the function of hsa_circ_0012563. In addition, the RT-PCR and Western blot were used to detect XRCC1 and epithelial-to-mesenchymal transition (EMT) related gene expression. RESULTS The RT-qPCR revealed that the hsa_circ_0012563 expression was remarkably upregulated in ESCC tissue and ESCC cell lines. Functionally, downregulation of hsa_circ_0012563 suppressed cell proliferation, migration, and invasion and promoted cell apoptosis. Mechanically, the knockdown of hsa_circ_0012563 inhibited XRCC1-mediated EMT pathway to suppress cell migration and invasion. CONCLUSIONS Therefore, these results reveal hsa_circ_0012563 is a critical oncogene and may be a novel biomarker in ESCC.
Collapse
Affiliation(s)
- Zhuo Zhang
- Department of Thoracic Surgery, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China
| | - Xueman Li
- Department of Thoracic Surgery, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China
| | - Fei Xiong
- Department of Thoracic Surgery, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China
| | - Zhangtao Ren
- Department of Pharmaceutical Sciences, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan, China
| | - Yongming Han
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
19
|
Zhou R, Tang X, Li L, Zhang F, Sun J, Ju C, Zhou Y, Liu R, Liang Y, Lv B, Zhang Z, Hu H, Lv XB. Identification of BRMS1L as Metastasis Suppressing Gene in Esophageal Squamous Cell Carcinoma. Cancer Manag Res 2020; 12:531-539. [PMID: 32021462 PMCID: PMC6987535 DOI: 10.2147/cmar.s232632] [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: 09/28/2019] [Accepted: 01/04/2020] [Indexed: 02/05/2023] Open
Abstract
Introduction Breast cancer metastasis suppressor 1 like (BRMS1-like)was first reported to be a component of the Sin3-HDAC complex, but the role in the progression of cancers was largely unknown. Our previous study reported that BRMS1L promoted the metastasis of breast cancer through facilitating the recruitment of HDAC complex to the promoter FZD10, and hence suppressing the transcription of FZD10. Methods In this study, we detected the expression level of BRMS1L in esophageal squamous cell carcinoma (ESCC). The effect of BRMS1L in TE-1D (knockdown) and ECA-109 (overexpression) cell lines was explored by transwell assays, wound healing assays, and cell adhesion assays. Quantitative real‑time PCR, Western blot analysis, and luciferase assays were used to detect the interaction of the CBP/P300-BRMS1L-ITGA7 axis. Results In the present study, we found that knockdown of BRMS1L promoted the migration, invasion, and epithelial-mesenchymal transition (EMT). Conversely, overexpression of BRMS1L inhibited the migration and invasion of ESCC. Mechanistically, BRMS1L exerted their metastasis-suppressing role via transcriptionally repress ITGA7 expression. Moreover, we revealed that CBP/p 300 regulated the expression of BRMS1L and might be responsible for the down-regulation of BRMS1L in ESCC. Conclusion Collectively, we identified the role of CBP/p300-BRMS1L-ITGA7 axis in the metastasis of ESCC.
Collapse
Affiliation(s)
- Ruihao Zhou
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang 30008, People's Republic of China.,Department of Pain Management, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, People's Republic of China
| | - Xiaofeng Tang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang 30008, People's Republic of China
| | - Liping Li
- Department of Clinical Laboratory, The Third Affiliated Hospital of Nanchang University, Nanchang 330008, People's Republic of China
| | - Feifei Zhang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang 30008, People's Republic of China
| | - Jun Sun
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang 30008, People's Republic of China
| | - Cheng Ju
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang 30008, People's Republic of China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University,Nanchang 330008, People's Republic of China
| | - Yan Zhou
- Department of Oncology,Shanghai Jiao Tong University Affiliated Sixth People's Hospital of Shanghai, Shanghai 200233, People's Republic of China
| | - Renfeng Liu
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang 30008, People's Republic of China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University,Nanchang 330008, People's Republic of China
| | - Yiping Liang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang 30008, People's Republic of China
| | - Bin Lv
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang 30008, People's Republic of China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University,Nanchang 330008, People's Republic of China
| | - Zhiping Zhang
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang 30008, People's Republic of China.,Department of Orthopedics, The Third Affiliated Hospital of Nanchang University,Nanchang 330008, People's Republic of China
| | - Haiyan Hu
- Department of Oncology,Shanghai Jiao Tong University Affiliated Sixth People's Hospital of Shanghai, Shanghai 200233, People's Republic of China
| | - Xiao-Bin Lv
- Jiangxi Key Laboratory of Cancer Metastasis and Precision Treatment, The Third Affiliated Hospital of Nanchang University, Nanchang 30008, People's Republic of China
| |
Collapse
|
20
|
Emerging Role of Non-Coding RNAs in Esophageal Squamous Cell Carcinoma. Int J Mol Sci 2019; 21:ijms21010258. [PMID: 31905958 PMCID: PMC6982002 DOI: 10.3390/ijms21010258] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/04/2019] [Accepted: 12/14/2019] [Indexed: 12/14/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly prevalent tumor and is associated with ethnicity, genetics, and dietary intake. Non-coding RNAs (ncRNAs), specifically microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs) have been reported as functional regulatory molecules involved in the development of many human cancers, including ESCC. Recently, several ncRNAs have been detected as oncogenes or tumor suppressors in ESCC progression. These ncRNAs influence the expression of specific genes or their associated signaling pathways. Moreover, interactions of ncRNAs are evident in ESCC, as miRNAs regulate the expression of lncRNAs, and further, lncRNAs and circRNAs function as miRNA sponges to compete with the endogenous RNAs. Here, we discuss and summarize the findings of recent investigations into the role of ncRNAs (miRNAs, lncRNAs, and circRNAs) in the development and progression of ESCC and how their interactions regulate ESCC development.
Collapse
|
21
|
Yu Z, Ni F, Chen Y, Zhang J, Cai J, Shi W. miR-125b suppresses cell proliferation and metastasis by targeting HAX-1 in esophageal squamous cell carcinoma. Pathol Res Pract 2019; 216:152792. [PMID: 31899048 DOI: 10.1016/j.prp.2019.152792] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/27/2019] [Accepted: 12/12/2019] [Indexed: 01/22/2023]
Affiliation(s)
- Zhijun Yu
- Department of Thoracic Surgery, The Second People's Hospital of Nantong, Nantong, Jiangsu, China
| | - Feng Ni
- Department of Radiation Oncology, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Yongmei Chen
- Department of Thoracic Surgery, The Second People's Hospital of Nantong, Nantong, Jiangsu, China
| | - Jie Zhang
- Department of Thoracic Surgery, The Second People's Hospital of Nantong, Nantong, Jiangsu, China
| | - Jing Cai
- Department of Radiation Oncology, Nantong Tumor Hospital, Nantong, Jiangsu, China.
| | - Weidong Shi
- Department of Thoracic Surgery, The Second People's Hospital of Nantong, Nantong, Jiangsu, China.
| |
Collapse
|
22
|
Wang KL, Chen XL, Lei L, Li P, Hong LL, Huang XC, Mao WM, Mukaisho K, Ling ZQ. Validation study of susceptibility loci for esophageal squamous cell carcinoma identified by GWAS in a Han Chinese subgroup from Eastern China. J Cancer 2019; 10:3624-3631. [PMID: 31333779 PMCID: PMC6636302 DOI: 10.7150/jca.32810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 05/04/2019] [Indexed: 11/05/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) occurs at a relatively high frequency in China and is one of the most prevalent cancers in the world. Genome-wide association studies (GWAS) have identified 24 single-nucleotide polymorphisms (SNPs) that could be associated with ESCC in Chinese patients. This retrospective study aimed to validate the association between these 24 SNPs and ESCC in a Han Chinese subgroup from East China. A total of 2280 and 1900 patients with ESCC (case group) and non-esophageal cancer (control group) were included from a single center. Genotyping of the 24 polymorphisms was performed using the Sequenom MassARRAY system. Unconditional logistic regression analyses were conducted for every polymorphism. It was found that rs12188136 (P=0.027, OR=1.158, 95% CI=1.016-1.319 for AG/AA) was associated with ESCC. Binary logistic regression analyses revealed a significant negative association of rs875339 in RORA (P=0.014, OR=0.762, 95% CI=0.613-0.947 for TT/CC). Under the dominant model, rs6854472 was slightly associated with ESCC risk (P=0.048, OR=1.192, 95% CI=1.002-1.418). Under the recessive model, a significant negative association was observed for rs875339 (P=0.010, OR=0.758, 95% CI=0.615-0.935). In a word, this large-scale replication study validated that rs12188136 and rs6854472 are associated with ESCC in a Han Chinese subgroup from Eastern China, and that rs875339 is negative associated with ESCC.
Collapse
Affiliation(s)
- Kai-Lai Wang
- Zhejiang Cancer Institute, Zhejiang Cancer Hospital, No.1 Banshan East Rd., Gongshu District, Hangzhou 310022, P.R.China
| | - Xiang-Liu Chen
- Zhejiang Cancer Institute, Zhejiang Cancer Hospital, No.1 Banshan East Rd., Gongshu District, Hangzhou 310022, P.R.China
| | - Lan Lei
- Zhejiang Cancer Institute, Zhejiang Cancer Hospital, No.1 Banshan East Rd., Gongshu District, Hangzhou 310022, P.R.China
| | - Pei Li
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Lian-Lian Hong
- Zhejiang Cancer Institute, Zhejiang Cancer Hospital, No.1 Banshan East Rd., Gongshu District, Hangzhou 310022, P.R.China
| | - Xian-Chong Huang
- Zhejiang Cancer Institute, Zhejiang Cancer Hospital, No.1 Banshan East Rd., Gongshu District, Hangzhou 310022, P.R.China
| | - Wei-Min Mao
- Department of Thoracic Tumor Surgery, Zhejiang Cancer Hospital, No.1 Banshan East Rd., Gongshu District, Hangzhou 310022, P.R.China.,Zhejiang Key Laboratory of Diagnosis & Treatment Technology on Thoracic Oncology (Lung and Esophagus), Hangzhou 310022, China
| | - Kenichi Mukaisho
- Department of Pathology, Division of Molecular Diagnostic Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Zhi-Qiang Ling
- Zhejiang Cancer Institute, Zhejiang Cancer Hospital, No.1 Banshan East Rd., Gongshu District, Hangzhou 310022, P.R.China.,Zhejiang Key Laboratory of Diagnosis & Treatment Technology on Thoracic Oncology (Lung and Esophagus), Hangzhou 310022, China
| |
Collapse
|
23
|
Guan C, Liu Z, Lu C, Xiao M, Shi H, Ni R, Bian Z. Nucleolar spindle-associated protein 1 promotes tumorigenesis and predicts poor prognosis in human esophageal squamous cell carcinoma. J Cell Biochem 2019; 120:11726-11737. [PMID: 30793360 DOI: 10.1002/jcb.28452] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 01/24/2023]
Abstract
The microtubule binding protein, nucleolar spindle-associated protein 1 (NUSAP1), has a crucial function in mitosis and its expression is closely associated with carcinogenesis. Herein, we aimed to determine the function of NUSAP1 in the development of human esophageal squamous cell carcinoma (ESCC), and the association of NUSAP1 expression with ESCC. Immunohistochemical staining of ESCC tissue sections indicated that NUSAP1 was expressed to a higher degree in tumor tissues than in adjacent nontumor tissues. NUSAP1 levels were relevant closely to histological differentiation (P = 0.049). Overall survival was longer in patients with lower NUSAP1 levels ( P < 0.001). NUSAP1 expression ( P = 0.002), histological differentiation ( P < 0.001), tumor depth ( P = 0.045), lymph node metastases ( P < 0.001), and tumor-node-metastasis staging ( P = 0.008) were greatly associated with overall survival using univariate analysis. Multivariate analysis suggested that histological differentiation ( P = 0.014) and NUSAP1 expression ( P = 0.026) could be independent prognostic markers for ESCC. Additionally, the biological behavior of ESCC cells was investigated in vitro and in vivo. Suppression of NUSAP1 inhibited cellular proliferation and invasion, and induced cell cycle arrest and apoptosis in vitro. More importantly, knockdown of NUSAP1 led to inhibition of tumor formation in nude mice. These findings indicated that NUSAP1 is a potential prognostic biomarker in ESCC, and is an ESCC oncogene. Thus, NUSAP1 could represent a therapeutic target for ESCC.
Collapse
Affiliation(s)
- Chengqi Guan
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zhaoxiu Liu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Cuihua Lu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Mingbing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hui Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Runzhou Ni
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Zhaolian Bian
- Department of Gastroenterology and Hepatology, Nantong Institute of Liver Disease, Nantong Third People's Hospital Affiliated to Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
24
|
Wu Y, Yang Y, Xian YS. HCRP1 inhibits cell proliferation and invasion and promotes chemosensitivity in esophageal squamous cell carcinoma. Chem Biol Interact 2019; 308:357-363. [PMID: 31152734 DOI: 10.1016/j.cbi.2019.05.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 04/27/2019] [Accepted: 05/20/2019] [Indexed: 12/17/2022]
Abstract
Hepatocellular carcinoma related protein 1 (HCRP1), which is essential for internalization and degradation of ubiquitinated membrane receptors, was reported to play crucial roles in cancer pathogenesis and progression. However, the functional roles of HCRP1 in esophageal squamous cell carcinoma (ESCC) remain unknown. In this study, we investigated the effects of HCRP1 on ESCC cells and the underlying mechanism. Our results demonstrated that HCRP1 was lowly expressed in ESCC tissues and cell lines. Overexpression of HCRP1 significantly suppressed ESCC cell proliferation and invasion as well as the epithelial-mesenchymal transition (EMT) process. Furthermore, HCRP1 increased the sensitivity of ESCC cells towards cisplatin/fluorouracil. Mechanistically, HCRP1 inhibited the activity of Wnt/β-catenin signaling pathway in ESCC cells. In conclusion, these findings indicated that HCRP1 suppressed ESCC cell proliferation and invasion through regulation of the Wnt/β-catenin pathway. Therefore, HCRP1 may function as a tumor suppressor in ESCC progression.
Collapse
Affiliation(s)
- Yu Wu
- Department of Thoracic Surgery, Shaanxi People's Hospital, Xi'an, 710068, China
| | - Ye Yang
- Department of Thoracic Surgery, Shaanxi People's Hospital, Xi'an, 710068, China
| | - Yin-Sheng Xian
- Department of Oncosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
25
|
Zhang J, Liu S, Zhang D, Ma Z, Sun L. Homeobox D10, a tumor suppressor, inhibits the proliferation and migration of esophageal squamous cell carcinoma. J Cell Biochem 2019; 120:13717-13725. [PMID: 30938888 DOI: 10.1002/jcb.28644] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/31/2019] [Accepted: 02/04/2019] [Indexed: 12/18/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common types of esophageal cancer, which is the sixth leading cause of cancer death globally. Homeobox D10 (HOXD10) is a member of the homeobox (HOX) gene family and has been reported to act as a tumor suppressor. However, the potential role of HOXD10 in ESCC has not been reported. Thus, the aim of this study was to examine the expression and function of HOXD10 in ESCC. The expressions of HOXD10 in human ESCC tissues and cell lines were detected by quantitative reverse transcription polymerase chain reaction and Western blot. The HOXD10 overexpressing cell lines were established, then CCK-8 and Transwell assays were performed to examine cell proliferation, migration, and invasion, respectively. The expression of EMT-related proteins and signaling pathway-related proteins were detected by Western blot. Our results showed that HOXD10 is lowly expressed in ESCC tissues as well as in ESCC cell lines. Ectopic overexpression of HOXD10 inhibited cell proliferation, migration, and invasion of ESCC cells (P < 0.05). HOXD10 overexpression repressed the epithelial-mesenchymal transition (EMT) process in ESCC cells. Besides, HOXD10 overexpression suppressed the activation of PI3K/AKT/mTOR signaling pathway. PI3K/Akt agonist, insulin-like growth factor-1, reversed the inhibitory effects of HOXD10 on cell proliferation and migration in ESCC cells. Additional in vivo study proved that ectopic expression of HOXD10 caused an obvious inhibitory effect on the tumor growth. These findings indicated that overexpression of HOXD10 suppressed the proliferation, migration, and invasion via regulating the PI3K/AKT/mTOR signaling pathway in ESCC cells. Thus, targeting HOXD10 may be considered as a therapeutic strategy for ESCC treatment.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Shiyuan Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Danjie Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zhenchuan Ma
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Liangzhang Sun
- Department of Thoracic Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
26
|
Su Z, Zou GR, Mao YP, OuYang PY, Cao XL, Xie FY, Li Q. Prognostic impact of family history of cancer in Southern Chinese patients with esophageal squamous cell cancer. J Cancer 2019; 10:1349-1357. [PMID: 31031844 PMCID: PMC6485237 DOI: 10.7150/jca.26511] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 12/05/2018] [Indexed: 12/30/2022] Open
Abstract
Background: First degree family history of cancer is associated with developing esophageal cancer and sparse data is about the impact on poor survival among established esophageal squamous cell cancer (ESCC) patients. In this study, we investigated the prognoses of patients with ESCC with a family history. Methods: A total of 479 ESCC patients were retrospectively enrolled from a Southern Chinese institution. A positive family history was defined as having malignant cancer among parents and siblings. Kaplan-Meier plots and Cox proportional hazards regressions were applied for overall survival (OS) and progression-free survival (PFS). Results: Among 479 patients, 119 (24.8%) and 68 (14.2%) reported a first-degree family history of cancer and digestive tract cancer, respectively. Compared with patients without a family history of cancer, the adjusted hazard ratios (HR) among those with it were 1.40 (95% CI, 1.08-1.82, p=0.011) for death, 1.36 (95% CI, 1.05-1.76, p=0.018) for progression. Similar results were observed in those with a family history of digestive tract cancer (HR=1.69, 95%CI, 1.24-1.98, p=0.001 for death and HR=1.77, 95%CI, 1.30-2.37, p<0.001 for progression, respectively). Furthermore, there was a trend for increasing risk of overall mortality (p=0.021, p=0.004, respectively), and progression (p=0.022, p=0.001, respectively) with an increasing number of affected family members. Conclusion: A first-degree family history of cancer, especially digestive tract cancer is associated with poor survival for established ESCC patients and plays an important role in prognosis. The patients with a family history of cancer might need a greater intensity of treatment and more frequent follow-up.
Collapse
Affiliation(s)
- Zhen Su
- Panyu central hospital, Cancer Institute of Panyu, Guangzhou, China
| | - Guo-Rong Zou
- Panyu central hospital, Cancer Institute of Panyu, Guangzhou, China
| | - Yan-Ping Mao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Pu-Yun OuYang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Xiao-Long Cao
- Panyu central hospital, Cancer Institute of Panyu, Guangzhou, China
| | - Fang-Yun Xie
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Qun Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
27
|
Zhong S, Yan H, Chen Z, Li Y, Shen Y, Wang Y, Li L, Sheng S, Wang Y. Overexpression of TAF1L Promotes Cell Proliferation, Migration and Invasion in Esophageal Squamous Cell Carcinoma. J Cancer 2019; 10:979-989. [PMID: 30854104 PMCID: PMC6400815 DOI: 10.7150/jca.26504] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 12/05/2018] [Indexed: 02/05/2023] Open
Abstract
Currently, it reported that TAF1L gene mutation is found in a number of carcinomas, but its pathophysiological function has not been well studied. We focused on investigating expressive levels of TAF1L gene and protein in esophageal squamous cell carcinoma (ESCC) with two tissue microarrays, forty fresh paired ESCC and paracancer samples using immunohistochemistry, real-time PCR or Western blot in this study. Furthermore, we executed TAF1L silence with siRNA in ESCC cell lines to evaluate effects of TAF1L expression on cell proliferation, migration and invasion of ESCC via CCK-8, wound healing and transwell chamber assays. Moreover, key proteins related to ESCC development were also analyzed by Western blot. Results from this study showed that the expression of TAF1L mRNA and protein in ESCC tissues were significantly higher than that in matched paracancer tissues. However, its abnormal expression was not associated with other clinic features, such as the age, gender and pathological grade, except of TNM-N stage. Furthermore, the proliferation, migration and invasion of ESCC cells were inhibited after TAF1L gene silencing. As a consequence, the expression of c-Myc and phosphorylated Akt in esophageal squamous cell line after TAF1L-siRNA treatment were inversely decreased, while p53 was increased significantly, compared those to control group. Taken together, the results from this study suggest that TAF1L gene might be served as an oncogene, and its overexpression could accelerate to the tumorigenesis of ESCC via promoting the malignant cell proliferation and tumor metastasis.
Collapse
Affiliation(s)
- Shan Zhong
- Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518055, P. R. China
- Laboratory of Molecular Pathology, Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Hongfei Yan
- Pathology Laboratory, Shantou University Medical College, Cancer Hospital, Shantou, Guangdong 515041, P. R. China
| | - Zhengshan Chen
- Laboratory of Molecular Pathology, Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Yanpeng Li
- Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518055, P. R. China
| | - Yanqin Shen
- Laboratory of Molecular Pathology, Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Yongyu Wang
- Laboratory of Molecular Pathology, Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Lan Li
- Laboratory of Molecular Pathology, Shantou University Medical College, Shantou, Guangdong 515041, P. R. China
| | - Sitong Sheng
- Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518055, P. R. China
| | - Yun Wang
- Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong 518055, P. R. China
- ✉ Corresponding author: Center for Research and Technology of Precision Medicine, College of Life Sciences and Oceanography, Shenzhen University (Xili Campus), No. 1066, Xueyuan Ave, Nanshan Distract, Shenzhen, Guangdong 518055, P.R. China. Email address:
| |
Collapse
|
28
|
Xue WH, Fan ZR, Li LF, Lu JL, Ma BJ, Kan QC, Zhao J. Construction of an oesophageal cancer-specific ceRNA network based on miRNA, lncRNA, and mRNA expression data. World J Gastroenterol 2018; 24:23-34. [PMID: 29358879 PMCID: PMC5757122 DOI: 10.3748/wjg.v24.i1.23] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 11/07/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To explore the expression profiles of microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and mRNAs in oesophageal squamous cell carcinoma (ESCC) in order to construct an oesophageal cancer-specific competing endogenous RNA (ceRNA) network.
METHODS In this work, the expression data of miRNAs, lncRNAs, and mRNAs in ESCC were obtained. An oesophageal cancer-specific ceRNA network was then constructed and investigated.
RESULTS CeRNAs have the ability to reduce the targeting activity of miRNAs, leading to the de-repression of specific mRNAs with common miRNA response elements. CeRNA interactions have a critical effect in gene regulation and cancer development.
CONCLUSION This study suggests a novel perspective on potential oesophageal cancer mechanisms as well as novel pathways for modulating ceRNA networks for treating cancers.
Collapse
MESH Headings
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/pathology
- Cluster Analysis
- Computational Biology
- Databases, Genetic
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/metabolism
- Esophageal Neoplasms/mortality
- Esophageal Neoplasms/pathology
- Esophageal Squamous Cell Carcinoma
- Gene Expression Regulation, Neoplastic
- Gene Regulatory Networks
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
Collapse
Affiliation(s)
- Wen-Hua Xue
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Zhi-Rui Fan
- Cancer Centre, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Li-Feng Li
- Cancer Centre, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jing-Li Lu
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Bing-Jun Ma
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Quan-Cheng Kan
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Jie Zhao
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
29
|
Zheng H, Ren W, Pan X, Zhang Q, Liu B, Liu S, He J, Zhou Z. Role of intravoxel incoherent motion MRI in early assessment of the response of esophageal squamous cell carcinoma to chemoradiotherapy: A pilot study. J Magn Reson Imaging 2018; 48:349-358. [PMID: 29297204 DOI: 10.1002/jmri.25934] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 12/07/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Since definitive concurrent chemoradiotherapy (CRT) is standard therapy for inoperable esophageal squamous cell carcinoma (ESCC), early evaluation of treatment response is crucial for patients and would be useful in assessing response, especially in patients with severe side effects. PURPOSE To explore the feasibility of intravoxel incoherent motion (IVIM) MRI in the early assessment of treatment response to CRT. STUDY TYPE Prospective. POPULATION Twenty-three inoperable ESCC patients. SEQUENCE IVIM 3T MRI of nine b values (0, 25, 50, 75, 100, 150, 200, 500 and 800 s/mm2 ) was performed at four timepoints: pre-CRT (within 5 days before CRT), mid-CRT (2-3 weeks after the start of CRT), end-CRT (within 5 days after the end of CRT), and post-CRT (1 month after the end of CRT). ASSESSMENT IVIM-based parameters and ADC were analyzed independently by two radiologists and treatment response was assessed by the Response Evaluation Criteria in Solid Tumors (RECIST). STATISTICAL TESTS Analyses of variance for repeated measurements were conducted to observe dynamic changes of IVIM-based parameters (D, f, and D*) and ADC during CRT. The parameters and their change percentages (Δ%) were compared between complete response (CR) and partial response (PR) by Mann-Whitney U-test. Diagnostic performance of parameters in predicting response was tested with receiver-operating characteristic curve analysis. RESULTS ADC, D, and f increased significantly during CRT (P < 0.001, < 0.001, and 0.001, respectively). ADC, f, Δ%ADC, and Δ%D at mid-CRT in CR group were significantly higher than those in the PR group (P = 0.002, 0.013, 0.005, and 0.011, respectively). D combined with f and ADC had highest area under curve (0.917) in identifying CR from PR. DATA CONCLUSION IVIM parameters proved useful in assessing response to definitive concurrent CRT for inoperable ESCC and combined with ADC at an early stage of treatment was a good predictor of response. LEVEL OF EVIDENCE 2 Technical Efficacy: Stage 4 J. MAGN. RESON. IMAGING 2018;48:349-358.
Collapse
Affiliation(s)
- Huanhuan Zheng
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wei Ren
- Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Xia Pan
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qinglei Zhang
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Baorui Liu
- Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Song Liu
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jian He
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhengyang Zhou
- Department of Radiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| |
Collapse
|
30
|
Yang B, Yan T, Cui H, Xu E, Ma Y, Cheng C, Zhang L, Kong P, Wang F, Qian Y, Yang J, Li Y, Li H, Bi Y, Hu X, Wang J, Song B, Yang J, Gao W, Liu J, Zou B, Shi R, Zhang Y, Liu H, Liu Y, Zhai Y, Chang L, Wang Y, Zhang Y, Jia Z, Chen X, Xi Y, Li G, Liang J, Guo J, Guo S, Zhang R, Cheng X, Cui Y. The macro-evolutionary events in esophageal squamous cell carcinoma. Oncotarget 2017; 8:112770-112782. [PMID: 29348864 PMCID: PMC5762549 DOI: 10.18632/oncotarget.22625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/28/2017] [Indexed: 11/25/2022] Open
Abstract
Understanding the evolutionary processes operative in cancer genome may provide insights into clinical outcome and drug-resistance. However, studies focus on genomic signatures, especially for macro-evolutionary events, in esophageal squamous cell carcinoma (ESCC) are limited. Here, we integrated published genomic sequencing data to investigate underlying evolutionary characteristics in ESCC. We found most of ESCC genomes were polyploidy with high genomic instability. Whole genome doubling that acts as one of mechanisms for polyploidy was predicted as a late event in the majority of ESCC genome. Moreover, loss of heterozygosity events were more likely to occur in chromosomes harboring tumor suppressor genes in ESCC. The 40% of neutral loss of heterozygosity events was not a result of genome doubling, suggesting an alternative mechanism for neutral loss of heterozygosity formation. Importantly, deconstruction of copy number alterations extending to telomere revealed that telomere-bounded copy number alterations play a critical role for amplification/deletion of oncogenes/suppressor genes. For well-known genes SOX2, PIK3CA and TERT, nearly all of their amplifications were telomere bounded, which was further confirmed in a Japanese ESCC cohort. Furthermore, we provide evidence that karyotype evolution was mostly punctuated in ESCC. Collectively, our data reveal the potential biological role of whole genome doubling, neutral loss of heterozygosity and telomere-bounded copy number alterations, and highlight mecro-evolution in ESCC tumorigenesis.
Collapse
Affiliation(s)
- Bin Yang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China.,Department of Tumor Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Ting Yan
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Heyang Cui
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Enwei Xu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China.,Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Yanchun Ma
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Caixia Cheng
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China.,Department of Pathology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ling Zhang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Pengzhou Kong
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fang Wang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yu Qian
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jian Yang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaoping Li
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China.,Department of Colorectal & Anal Surgery, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Hongyi Li
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanghui Bi
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoling Hu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Juan Wang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Bin Song
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jie Yang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wei Gao
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Liu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Department of General Surgery, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Binbin Zou
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruyi Shi
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanyan Zhang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Department of General Surgery, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haiyan Liu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yiqian Liu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yuanfang Zhai
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lu Chang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yi Wang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yingchun Zhang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Zhiwu Jia
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xing Chen
- Department of Endoscopy, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Yanfeng Xi
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Guodong Li
- Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Jianfang Liang
- Department of Pathology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiansheng Guo
- Department of General Surgery, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shiping Guo
- Department of Tumor Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Rongsheng Zhang
- Department of Tumor Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, China
| | - Xiaolong Cheng
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongping Cui
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, China.,Shanxi Key Laboratory of Carcinogenesis and Translational Research on Esophageal Cancer, Shanxi Medical University, Taiyuan, Shanxi, China.,Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
31
|
Bulibu J, Wang W, Tang Y, Li N, Saifuding K. Association Between Polymorphisms in the Promoter Region of microRNA-34b/c and the Chemoradiotherapy Efficacy for Locally Advanced Esophageal Squamous Cell Carcinoma in Chinese Han Population. Pathol Oncol Res 2017; 25:421-427. [PMID: 29270777 DOI: 10.1007/s12253-017-0366-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/29/2017] [Indexed: 01/07/2023]
Abstract
The study aims to explore the association between polymorphisms in the promoter region of microRNA-34b/c (miR-34b/c) and the chemoradiotherapy efficacy for locally advanced esophageal squamous cell carcinoma (ESCC) in Chinese Han population. A total of 175 locally advanced ESCC cases and 186 healthy individuals were enrolled as the case and control groups. Denaturing high performance liquid chromatography (DHPLC) was applied to determine the genotypes of subjects. Subjects in the case group were classified into complete response (CR), partial response (PR), stable disease (SD) and progressive disease (PD). CR + PR were defined as the sensitive group, and SD + PD were defined as the resistance group. All patients were followed up for 3 ~ 36 months. Receiver operating characteristic (ROC) curve was used to evaluate the predictive value of rs4938723 in the promoter region of miR-34b/c in the chemoradiotherapy efficacy for patients with locally advanced ESCC. The distribution of genotype and allele of rs4938723 in the promoter region of miR-34b/c was significantly different between the case and control group (both P < 0.05), and CC genotype and C allele could decrease the risk of ESCC (CC genotype: OR = 0.57, 95%CI = 0.32 ~ 0.99, P = 0.045; C allele: OR = 0.72, 95%CI = 0.54 ~ 0.97, P = 0.032). MiR-34b/c rs4938723 was associated with ESCC TNM staging, differentiation degree, and lymph node metastasis (LNM) for ES CC patients (all P < 0.05). The chemoradiotherapy efficacy of patients with CC genotype was better than that of patients with (TT + TC) genotypes (P < 0.05). ROC curve results showed that the area under curve (AUC), sensitivity and specificity were 0.777, 85.1% and 71.3%, respectively. The average median progression free survival (PFS) of patients with (TT + TC) genotypes was significantly shorter than those patients with CC genotype (P < 0.05). Our study provides evidence that miR-34b/c rs4938723 is closely related with the chemoradiotherapy efficacy for locally advanced ESCC.
Collapse
Affiliation(s)
- Jilisihan Bulibu
- Department of Gastroenterology, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789, Suzhou East Street, Urumqi, 830000, the Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Wei Wang
- Department of Gastroenterology, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789, Suzhou East Street, Urumqi, 830000, the Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Yong Tang
- Department of Gastroenterology, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789, Suzhou East Street, Urumqi, 830000, the Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Na Li
- Department of Gastroenterology, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789, Suzhou East Street, Urumqi, 830000, the Xinjiang Uygur Autonomous Region, People's Republic of China
| | - Keyoumu Saifuding
- Department of Gastroenterology, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789, Suzhou East Street, Urumqi, 830000, the Xinjiang Uygur Autonomous Region, People's Republic of China.
| |
Collapse
|
32
|
The effect of celecoxib on DNA methylation of CDH13, TFPI2, and FSTL1 in squamous cell carcinoma of the esophagus in vivo. Anticancer Drugs 2017; 27:848-53. [PMID: 27400374 DOI: 10.1097/cad.0000000000000396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This study examined the in-vivo effect of the NSAID celecoxib on DNA methylation in the promoter region of the tumor-suppressor genes cadherin 13, tissue factor pathway inhibitor 12, and follistatin-like protein 1, and on apoptosis, in esophageal squamous cell carcinoma (ESCC). Forty-five patients who underwent an esophagectomy for ESCC were allocated to either a treatment group (n=22) or a control group (n=23). Patients in the treatment group were administered 800 mg/day of celecoxib for 14 days before surgery. Patients in the control group did not take any type of NSAID. Biopsies of the tumor were collected before surgery and tissue from the resection specimens after surgery. Methylation-specific PCR was used to measure DNA methylation and apoptosis was measured by flow cytometry. There was no difference in the proportion of patients with methylation for each of the genes between the patient groups before treatment. In those patients with pretreatment methylation, there was a significant reduction in the proportion with methylation and a significant increase in the corresponding messenger RNA expression after treatment with celecoxib. In those tissues in which there was a reduction in methylation following celecoxib treatment, there was a significant increase in the percentage of apoptotic cells, but not in the tissues with no change in methylation. In ESCC, in-vivo treatment with celecoxib is associated with a reduction in DNA methylation and increase in messenger RNA expression of tumor-suppressor genes, and increases in apoptosis.
Collapse
|
33
|
The correlation between XIAP gene polymorphisms and esophageal squamous cell carcinoma susceptibility and prognosis in a Chinese population. Pathol Res Pract 2017; 213:1482-1488. [PMID: 29037837 DOI: 10.1016/j.prp.2017.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 09/30/2017] [Accepted: 10/07/2017] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study aims to explore the correlation between X-linked inhibitor of apoptosis protein (XIAP) gene polymorphisms (rs8371 and rs9856) with the susceptibility and prognosis of esophageal squamous cell carcinoma (ESCC), providing a potential treatment for ESCC. METHOD A total of 170 ESCC patients (case group) and 191 healthy people (control group) were enrolled in our study. Genotyping was conducted on the basis of the ligase detection reaction (LDR). The expressions of XIAP polymorphisms were detected. The patients were followed up every three months until death or the last follow-up day. The overall survival (OS) and progression free survival (PFS) were recorded by Kaplan-Meier survival curve, and the relationship between XIAP gene polymorphism and risk and prognosis of ESCC was assessed by Cox multivariate analysis. RESULT TT+CT genotype and T allele frequencies of XIAP rs8371 and rs9856 in the case group were significantly lower compared to those of the control group (all P<0.05), suggesting that TT+CT genotype of XIAP rs8371 and rs9856 was associated with ESCC susceptibility. XIAP rs8371 and rs9856 polymorphisms were associated with tumor node metastasis (TNM) staging, depth of invasion and lymph node metastasis. The OS and PFS of TT+CT genotype carriers of rs8371 were longer than those of CC genotype carriers. Smoking, alcohol, TNM staging, depth of invasion, and lymph node metastasis were significantly associated with the OS and PFS in ESCC patients. Higher TNM staging, depth of invasion, and presence of lymph node metastasis were independent risk factors, while XIAP rs8371 was an independent protective factor for the prognosis of ESCC patients. CONCLUSION The present study demonstrates that XIAP rs8371 and rs9856 are associated with susceptibility to ESCC, and rs8371 polymorphisms might serve as an indicator for improved clinical efficacy and prognosis of ESCC patients.
Collapse
|
34
|
Zhai S, Xue J, Wang Z, Hu L. High expression of special AT-rich sequence binding protein-1 predicts esophageal squamous cell carcinoma relapse and poor prognosis. Oncol Lett 2017; 14:7455-7460. [PMID: 29344188 DOI: 10.3892/ol.2017.7081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/21/2017] [Indexed: 02/05/2023] Open
Abstract
Previous studies of the roles of special AT-rich sequence binding protein-1 (SATB1) in the development and progression of cancer have suggested that SATB1 promotes cancer cell metastasis. The aim of the present study is to evaluate the role of SATB1 in the progression and prognosis of esophageal squamous cell carcinoma (ESCC). ESCC tissues were collected from 102 patients and SATB1 mRNA expression was measured by reverse transcription-quantitative polymerase chain reaction. The association between expression of SATB1 mRNA with clinicopathological features and prognosis was assessed, and the prognosis of ESCC was evaluated using Kaplan-Meier survival curves. In the 102 ESCC tissues, SATB1 mRNA expression correlated with the clinical tumor node metastasis stage (P<0.05), but not with any other clinicopathological features (including age, gender, tumor differentiation grade, adjuvant radio/chemotherapy, or the consumption of alcohol and use of cigarettes) (P>0.05). The disease-free survival (DFS) and overall survival (OS) of patients with high SATB1 expression was decreased compared with those with low SATB1 expression. The present study indicated that SATB1 mRNA expression was associated with the postoperative recurrent and poor prognosis in ESCC. SATB1 may be a novel marker for predicting the recurrent and worse prognosis of ESCC.
Collapse
Affiliation(s)
- Songhui Zhai
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jianxin Xue
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Zheng Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lijuan Hu
- Department of Immunology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
35
|
Davidson M, Chau I, Cunningham D, Khabra K, Iveson T, Hickish T, Seymour M, Starling N. Impact of tumour histological subtype on chemotherapy outcome in advanced oesophageal cancer. World J Gastrointest Oncol 2017; 9:333-340. [PMID: 28868114 PMCID: PMC5561045 DOI: 10.4251/wjgo.v9.i8.333] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 04/20/2017] [Accepted: 05/19/2017] [Indexed: 02/05/2023] Open
Abstract
AIM To investigate the impact of histology on outcome in advanced oesophageal cancer treated with first-line fluoropyrimidine-based chemotherapy.
METHODS Individual patient data were pooled from three randomised phase III trials of fluoropyrimidine-based chemotherapy ± platinum/anthracycline in patients with advanced, untreated gastroesophageal adenocarcinoma or squamous cell carcinoma (SCC) randomised between 1994 and 2005. The primary endpoint was overall survival of oesophageal cancer patients according to histology. Secondary endpoints were response rates and a toxicity composite endpoint.
RESULTS Of the total 1836 randomised patients, 973 patients (53%) were eligible (707 patients with gastric cancer were excluded), 841 (86%) had adenocarcinoma and 132 (14%) had SCC. There was no significant difference in survival between patients with adenocarcinoma and SCC, with median overall survivals of 9.5 mo vs 7.6 mo (HR = 0.85, 95%CI: 0.70-1.03, P = 0.09) and one-year survivals of 38.8% vs 28.2% respectively. The overall response rate to chemotherapy was 44% for adenocarcinoma vs 33% for SCC (P = 0.01). There was no difference in the frequency of the toxicity composite endpoint between the two groups.
CONCLUSION There was no significant difference in survival between adenocarcinoma and SCC in patients with advanced oesophageal cancer treated with fluoropyrimidine-based chemotherapy despite a trend for worse survival and less chemo-sensitivity in SCC. Tolerance to treatment was similar in both groups. This analysis highlights the unmet need for SCC-specific studies in advanced oesophageal cancer and will aid in the design of future trials of targeted agents.
Collapse
|
36
|
Silencing of Rab3D suppresses the proliferation and invasion of esophageal squamous cell carcinoma cells. Biomed Pharmacother 2017; 91:402-407. [DOI: 10.1016/j.biopha.2017.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/23/2017] [Accepted: 04/10/2017] [Indexed: 12/27/2022] Open
|
37
|
Erkizan HV, Johnson K, Ghimbovschi S, Karkera D, Trachiotis G, Adib H, Hoffman EP, Wadleigh RG. African-American esophageal squamous cell carcinoma expression profile reveals dysregulation of stress response and detox networks. BMC Cancer 2017; 17:426. [PMID: 28629367 PMCID: PMC5477112 DOI: 10.1186/s12885-017-3423-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 06/12/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Esophageal carcinoma is the third most common gastrointestinal malignancy worldwide and is largely unresponsive to therapy. African-Americans have an increased risk for esophageal squamous cell carcinoma (ESCC), the subtype that shows marked variation in geographic frequency. The molecular architecture of African-American ESCC is still poorly understood. It is unclear why African-American ESCC is more aggressive and the survival rate in these patients is worse than those of other ethnic groups. METHODS To begin to define genetic alterations that occur in African-American ESCC we conducted microarray expression profiling in pairs of esophageal squamous cell tumors and matched control tissues. RESULTS We found significant dysregulation of genes encoding drug-metabolizing enzymes and stress response components of the NRF2- mediated oxidative damage pathway, potentially representing key genes in African-American esophageal squamous carcinogenesis. Loss of activity of drug metabolizing enzymes would confer increased sensitivity of esophageal cells to xenobiotics, such as alcohol and tobacco smoke, and may account for the high incidence and aggressiveness of ESCC in this ethnic group. To determine whether certain genes are uniquely altered in African-American ESCC we performed a meta-analysis of ESCC expression profiles in our African-American samples and those of several Asian samples. Down-regulation of TP53 pathway components represented the most common feature in ESCC of all ethnic groups. Importantly, this analysis revealed a potential distinctive molecular underpinning of African-American ESCC, that is, a widespread and prominent involvement of the NRF2 pathway. CONCLUSION Taken together, these findings highlight the remarkable interplay of genetic and environmental factors in the pathogenesis of African-American ESCC.
Collapse
Affiliation(s)
- Hayriye Verda Erkizan
- Institute for Clinical Research, Department of Veteran Affairs Medical Center (VAMC), Washington, D.C., USA
| | - Kory Johnson
- Bioinformatics Neuroscience Group, Information Technology Program, National Institute of Neurological Disorders & Stroke, Bethesda, MD, USA
| | - Svetlana Ghimbovschi
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, D.C., USA
| | - Deepa Karkera
- Institute for Clinical Research, Department of Veteran Affairs Medical Center (VAMC), Washington, D.C., USA
| | | | - Houtan Adib
- Radiology Service, VAMC, Washington, D.C., USA
| | - Eric P Hoffman
- Research Center for Genetic Medicine, Children's National Medical Center, Washington, D.C., USA
- Present address: School of Pharmacy, Binghamton University - SUNY, Binghamton, NY, USA
| | - Robert G Wadleigh
- Institute for Clinical Research, Department of Veteran Affairs Medical Center (VAMC), Washington, D.C., USA.
- Oncology Section, Washington DC VAMC, 50 Irving St. NW, Washington DC, 20422, USA.
| |
Collapse
|
38
|
Das M, Sharma SK, Sekhon GS, Mahanta J, Phukan RK, Jalan BK. p16 gene silencing along with p53 single-nucleotide polymorphism and risk of esophageal cancer in Northeast India. Tumour Biol 2017; 39:1010428317698384. [PMID: 28459370 DOI: 10.1177/1010428317698384] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The high incidence of esophageal cancer in Northeast India and the unique ethnic background and dietary habits provide a great opportunity to study the molecular genetics behind esophageal squamous cell carcinoma in this part of the region. We hypothesized that in addition to currently known environmental risk factors for esophageal cancer, genetic and epigenetic factors are also involved in esophageal carcinogenesis in Northeast India. Therefore, in this study, we explored the possible association between the two important G1 cell cycle regulatory genes p16 and p53 and environmental risk factors and risk of esophageal carcinogenesis. A total of 100 newly diagnosed esophageal cancer cases along with equal number of age-, sex-, and ethnicity-matched controls were included in this study. Methylation-specific polymerase chain reaction was used to determine the p16 promoter methylation status. Single-nucleotide polymorphism at codon 72 of p53 gene was assessed by the polymerase chain reaction-restriction fragment length polymorphism method. Aberrant methylation of p16 gene was seen in 81% of esophageal cancer cases. Hypermethylation of p16 gene was not found in healthy controls. p53 Pro/Pro genotype was found to be a risk genotype in Northeast India compared with Arg/Pro and Arg/Arg. p53 variant/polymorphism was significantly associated with esophageal cancer risk in the study population under all three genetic models, namely, dominant model (Arg/Pro + Pro/Pro vs Arg/Arg odds ratio = 2.25, confidence interval = 1.19-4.26; p = 0.012), recessive model (Arg/Arg + Arg/Pro vs Pro/Pro odds ratio = 2.35, confidence interval = 1.24-4.44; p = 0.008), and homozygous model (Pro/Pro vs Arg/Arg odds ratio = 3.33, confidence interval = 1.54-7.20; p = 0.002). However, p53 variant/polymorphism was not statistically associated with esophageal cancer risk under the heterozygous model (Pro/Pro vs Arg/Pro). In the case-only analysis based on p16 methylation, the p53 variant/polymorphism (Pro/Pro or Arg/Pro) showed significant association for esophageal cancer risk (odds ratio = 3.33, confidence interval = 1.54-7.20; p = 0.002). Gene-gene and gene-environment interaction using the case-only approach revealed a strong association between p16 methylation, p53 single-nucleotide polymorphism, and environmental factors and esophageal cancer risk. Cases with p16 methylation and p53 variant/polymorphism (Pro/Pro or Arg/Pro) along with both betel quid and tobacco chewing habit (odds ratio = 8.29, confidence interval = 1.14-60.23; p = 0.037) conferred eightfold increased risk toward esophageal cancer development. This study reveals a synergistic interaction between epigenetic, genetic, and environmental factors and risk of esophageal cancer in this high-incidence region of Northeast India. The inactivation of either p16 or p53 in a majority of esophageal cancer cases in this study suggests the possible crosstalk between the important cell cycle genes.
Collapse
Affiliation(s)
- Mandakini Das
- 1 Regional Medical Research Centre, NE Region (ICMR), Dibrugarh, India
| | | | | | - Jagadish Mahanta
- 1 Regional Medical Research Centre, NE Region (ICMR), Dibrugarh, India
| | - Rup Kumar Phukan
- 1 Regional Medical Research Centre, NE Region (ICMR), Dibrugarh, India
| | | |
Collapse
|
39
|
Tan B, Wang J, Song Q, Wang N, Jia Y, Wang C, Yao B, Liu Z, Zhang X, Cheng Y. Prognostic value of PAX9 in patients with esophageal squamous cell carcinoma and its prediction value to radiation sensitivity. Mol Med Rep 2017; 16:806-816. [PMID: 28560390 PMCID: PMC5482201 DOI: 10.3892/mmr.2017.6626] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 03/27/2017] [Indexed: 12/25/2022] Open
Abstract
Abnormal paired box 9 (PAX9) expression is associated with tumorigenesis, cancer development, invasion and metastasis. The present study investigated the prognostic significance of PAX9 in esophageal squamous cell carcinoma (ESCC) and its role in predicting radiation sensitivity. A total of 52.8% (121/229) ESCC tissues were positive for PAX9. The 1-, 3- and 5-year disease-free survival (DFS) rates were 72.2, 35.2 and 5.6%, respectively, and the overall survival (OS) rates were and 86.1, 44.4, and 23.1%, respectively, in PAX9-positive tumors. In PAX9-negative tumors, the one-, three- and five-year DFS rates were 76.9, 47.9 and 24.0%, and the OS rates were 90.9, 57.9 and 38.8%, respectively. Univariate analysis revealed that PAX9, differentiation, T stage, lymph node metastasis, and tumor-node-metastasis stage were associated with OS. Multivariate analysis of DFS and OS revealed that the hazard ratios for PAX9 were 0.624 (95% CI: 0.472–0.869, P=0.004) and 0.673 (95% CI: 0.491–0.922, P=0.014), respectively. Patients that received adjuvant therapy exhibited significant differences in the 5-year DFS (P<0.001) and OS (P<0.001). PAX9-positive ESCC patients who received post-surgery radiotherapy had a significantly greater 5-year DFS (P=0.011) and OS (P=0.009) than patients who received surgery only. Thus, PAX9 may be an independent prognostic factor for the surgical treatment of ESCC and a possible predictor of radiation sensitivity.
Collapse
Affiliation(s)
- Bingxu Tan
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Jianbo Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qingxu Song
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Nana Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yibin Jia
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Cong Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Bin Yao
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhulong Liu
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiaomei Zhang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
40
|
Zhang P, Li XM, Zhao XK, Song X, Yuan L, Shen FF, Fan ZM, Wang LD. Novel genetic locus at MHC region for esophageal squamous cell carcinoma in Chinese populations. PLoS One 2017; 12:e0177494. [PMID: 28493959 PMCID: PMC5426749 DOI: 10.1371/journal.pone.0177494] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
Background Our previous genome-wide association study (GWAS) identified three independent single nucleotide polymorphisms (SNPs) in human major histocompatibility complex (MHC) region showing association with esophageal squamous cell carcinoma (ESCC). In this study, we increased GWAS sample size on MHC region and performed validation in an independent ESCC cases and normal controls with aim to find additional loci at MHC region showing association with an increased risk to ESCC. Methods The 1,077 ESCC cases and 1,733 controls were genotyped using Illumina Human 610-Quad Bead Chip, and 451 cases and 374 controls were genotyped using Illumina Human 660W-Quad Bead Chip. After quality control, the selected SNPs were replicated by TaqMan genotyping assay on another 2,026 ESCC cases and 2,384 normal controls. Results By excluding low quality SNPs in primary GWAS screening, we selected 2,533 SNPs in MHC region for association analysis, and identified 5 SNPs with p <10−4. Further validation analysis in an independent case-control cohort confirmed one of the 5 SNPs (rs911178) that showed significant association with ESCC. rs911178 (PGWAS = 6.125E-04, OR = 0.644 and Preplication = 1.406E-22, OR = 0.489) was located at upstream of SCAND3. Conclusion The rs911178 (SCAND3 gene) in MHC region is significantly associated with high risk of ESCC. This study not only reveal the potential role of MHC region for the pathogenesis of ESCC, but also provides important clues for the establishment of tools and methods for screening high risk population of ESCC.
Collapse
Affiliation(s)
- Peng Zhang
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin-Min Li
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Pathology, The Maternal and Child Health Care Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Xue-Ke Zhao
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xin Song
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ling Yuan
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Fang-Fang Shen
- The Key Laboratory for Tumor Translational Medicine, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Zong-Min Fan
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Li-Dong Wang
- Henan Key Laboratory for Esophageal Cancer Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- * E-mail:
| |
Collapse
|
41
|
Miao Y, Liu R, Pu Y, Yin L. Trends in esophageal and esophagogastric junction cancer research from 2007 to 2016: A bibliometric analysis. Medicine (Baltimore) 2017; 96:e6924. [PMID: 28514311 PMCID: PMC5440148 DOI: 10.1097/md.0000000000006924] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND This study aimed to analyze the scientific outputs of esophageal and esophagogastric junction (EGJ) cancer and construct a model to quantitatively and qualitatively evaluate pertinent publications from the past decade. METHODS Publications from 2007 to 2016 were retrieved from the Web of Science Core Collection database. Microsoft Excel 2016 (Redmond, WA) and the CiteSpace (Drexel University, Philadelphia, PA) software were used to analyze publication outcomes, journals, countries, institutions, authors, research areas, and research frontiers. RESULTS A total of 12,978 publications on esophageal and EGJ cancer were identified published until March 23, 2017. The Journal of Clinical Oncology had the largest number of publications, the USA was the leading country, and the University of Texas MD Anderson Cancer Center was the leading institution. Ajani JA published the most papers, and Jemal A had the highest co-citation counts. Esophageal squamous cell carcinoma ranked the first in research hotspots, and preoperative chemotherapy/chemoradiotherapy ranked the first in research frontiers. CONCLUSION The annual number of publications steadily increased in the past decade. A considerable number of papers were published in journals with high impact factor. Many Chinese institutions engaged in esophageal and EGJ cancer research but significant collaborations among them were not noted. Jemal A, Van Hagen P, Cunningham D, and Enzinger PC were identified as good candidates for research collaboration. Neoadjuvant therapy and genome-wide association study in esophageal and EGJ cancer research should be closely observed.
Collapse
Affiliation(s)
- Yan Miao
- Key Laboratory of Environmental Medicine Engineering of Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu Province, China
| | | | | | | |
Collapse
|
42
|
Lu Z, Wang J, Zheng Y, Yang S, Liu M, Chen X, Wang C, Hou G. Wild-type phosphatase and tensin homolog deleted on chromosome 10 improved the sensitivity of cells to rapamycin through regulating phosphorylation of Akt in esophageal squamous cell carcinoma. Dis Esophagus 2017; 30:1-8. [PMID: 26725440 DOI: 10.1111/dote.12448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most frequently diagnosed cancers in China, but the etiology and mode of carcinogenesis of this disease remain poorly understood. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN), as a negative regulator of Akt/mTOR pathway, frequently mutates or is inactive in many cancers. Although mTOR has been thought a promising cancer therapeutic target, the sensitivity of tumor cells to rapamycin was still to be revaluated. In this study, we measured the effects of rapamycin on cell proliferation and phosphorylation of Akt in ESCC cells with varying degrees of differentiation. And then, the relationship between PTEN status and the sensitivity of cells to rapamycin was investigated in EC9706 cells with or without wild-type PTEN in vitro and in vivo. The results demonstrated ESCC cells with poor differentiation were insensitive to rapamycin of high concentration and rapamycin obviously promoted the phosphorylation of Akt in these cells, but it had no obvious effects on p-Akt in cells with well differentiation. Also, we showed that wild-type PTEN improved the sensitivity of poor differentiation cells to rapamycin through inhibiting phosphorylation of Akt in vitro and in vivo. This study explored the possible molecular mechanism of some ESCC cells insensitive to rapamycin and provided a measure for treating ESCC patients with PTEN inactivation using mTOR inhibitors.
Collapse
Affiliation(s)
- Z Lu
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China
| | - J Wang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China
| | - Y Zheng
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China
| | - S Yang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China
| | - M Liu
- Oncology Department, People's Hospital of Henan Province, China
| | - X Chen
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China
| | - C Wang
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China.,New Drug Research and Development Centre of Zhengzhou University, Zhengzhou, China
| | - G Hou
- Department of Clinical Pharmacy, School of Pharmaceutical Sciences, Zhengzhou University, China.,New Drug Research and Development Centre of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
43
|
Ma Z, Feng J, Guo Y, Kong R, Ma Y, Sun L, Yang X, Zhou B, Li S, Zhang W, Jiang J, Zhang J, Qiao Z, Cheng Y, Zha D, Liu S. Knockdown of DDX5 Inhibits the Proliferation and Tumorigenesis in Esophageal Cancer. Oncol Res 2016; 25:887-895. [PMID: 28244855 PMCID: PMC7841059 DOI: 10.3727/096504016x14817158982636] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
DEAD (Asp-Glu-Ala-Asp) box protein 5 (DDX5), a prototypical member of the DEAD/H-box protein family, has been involved in several human malignancies. However, the expression and biological role of DDX5 in esophageal cancer (EC) remain largely unknown. In this study, we examined the role of DDX5 in regulating EC cell proliferation and tumorigenesis and explored its possible molecular mechanism. We found that DDX5 was overexpressed in human EC cell lines. In addition, knockdown of DDX5 significantly inhibited the proliferation of EC cells in vitro and the growth of EC xenografts in vivo. Knockdown of DDX5 also suppressed the migration/invasion and epithelial-to-mesenchymal transition (EMT) phenotype in EC cells. Furthermore, we observed that knockdown of DDX5 inhibited the expression of β-catenin, c-Myc, and cyclin D1 in EC cells. In conclusion, our findings provide the first evidence that siRNA-DDX5 inhibited the proliferation and invasion of EC cells through suppressing the Wnt/β-catenin signaling pathway. Therefore, DDX5 may be a novel potential therapeutic target for the prevention and treatment of EC.
Collapse
|
44
|
Can CT-PET and Endoscopic Assessment Post-Neoadjuvant Chemoradiotherapy Predict Residual Disease in Esophageal Cancer? Ann Surg 2016; 264:831-838. [DOI: 10.1097/sla.0000000000001902] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
45
|
Li F, Zhang L, Li W, Deng J, Zheng J, An M, Lu J, Zhou Y. Circular RNA ITCH has inhibitory effect on ESCC by suppressing the Wnt/β-catenin pathway. Oncotarget 2016; 6:6001-13. [PMID: 25749389 PMCID: PMC4467417 DOI: 10.18632/oncotarget.3469] [Citation(s) in RCA: 563] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/20/2015] [Indexed: 12/17/2022] Open
Abstract
Circular RNAs with exonic sequences represent a special form of non-coding RNAs, discovered by analyzing a handful of transcribed genes. It has been observed that circular RNAs function as microRNA sponges. In the present study, we investigated whether the expression of circular RNAs is altered during the development of esophageal squamous cell carcinoma (ESCC). Using a TaqMan-based reverse transcriptase polymerase chain reaction assay, the relationship between cir-ITCH and ESCC was analyzed in a total of 684 ESCC and paired adjacent non-tumor tissue samples from eastern and southern China. We found that cir-ITCH expression was usually low in ESCC compared to the peritumoral tissue. The functional relevance of cir-ITCH was further examined by biochemical assays. As sponge of miR-7, miR-17, and miR-214, cir-ITCH might increase the level of ITCH. ITCH hyper expression promotes ubiquitination and degradation of phosphorylated Dvl2, thereby inhibiting the Wnt/β-catenin pathway. These results indicate that cir-ITCH may have an inhibitory effect on ESCC by regulating the Wnt pathway.
Collapse
Affiliation(s)
- Fang Li
- Department of Genetics, Medical College of Soochow University, Suzhou, China
| | - Liyuan Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Li
- Department of Genetics, Medical College of Soochow University, Suzhou, China
| | - Jieqiong Deng
- Department of Genetics, Medical College of Soochow University, Suzhou, China
| | - Jian Zheng
- Department of Genetics, Medical College of Soochow University, Suzhou, China
| | - Mingxing An
- Department of Genetics, Medical College of Soochow University, Suzhou, China
| | - Jiachun Lu
- The Institute for Chemical Carcinogenesis, The State Key Lab of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Yifeng Zhou
- Department of Genetics, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
46
|
Overexpression of Suprabasin is Associated with Proliferation and Tumorigenicity of Esophageal Squamous Cell Carcinoma. Sci Rep 2016; 6:21549. [PMID: 26899563 PMCID: PMC4761926 DOI: 10.1038/srep21549] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/27/2016] [Indexed: 12/27/2022] Open
Abstract
Suprabasin is a recently identified oncoprotein that is upregulated in multiple cancers. However, the clinical significance and biological role of suprabasin in human esophageal squamous cell carcinoma (ESCC) remains unclear. In the current study, we reported that suprabasin was markedly overexpressed in ESCC cell lines and tissues at both mRNA and protein levels, and this was associated with advanced clinical stage, tumor-nodes-metastasis (TNM) classification, histological differentiation, tumor size and poorer survival. Furthermore, we found that both proliferation and tumorigenicity of ESCC cells were significantly induced by suprabasin overexpression, but inhibited by suprabasin knock-down. Moreover, we demonstrated that upregulation of suprabasin activated the Wnt/β-catenin signaling pathway and led to nuclear localization of β-catenin and upregulation of Cyclin D1 and c-Myc. Together, our results suggest that suprabasin plays an important oncogenic role in promoting proliferation and tumorigenesis of ESCC.
Collapse
|
47
|
Sasaki Y, Tamura M, Koyama R, Nakagaki T, Adachi Y, Tokino T. Genomic characterization of esophageal squamous cell carcinoma: Insights from next-generation sequencing. World J Gastroenterol 2016; 22:2284-2293. [PMID: 26900290 PMCID: PMC4735002 DOI: 10.3748/wjg.v22.i7.2284] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 09/29/2015] [Accepted: 12/21/2015] [Indexed: 02/06/2023] Open
Abstract
Two major types of cancer occur in the esophagus: squamous cell carcinoma, which is associated with chronic smoking and alcohol consumption, and adenocarcinoma, which typically arises in gastric reflux-associated Barrett’s esophagus. Although there is increasing incidence of esophageal adenocarcinoma in Western counties, esophageal squamous cell carcinoma (ESCC) accounts for most esophageal malignancies in East Asia, including China and Japan. Technological advances allowing for massively parallel, high-throughput next-generation sequencing (NGS) of DNA have enabled comprehensive characterization of somatic mutations in large numbers of tumor samples. Recently, several studies were published in which whole exome or whole genome sequencing was performed in ESCC tumors and compared with matched normal DNA. Mutations were validated in several genes, including in TP53, CDKN2A, FAT1, NOTCH1, PIK3CA, KMT2D and NFE2L2, which had been previously implicated in ESCC. Several new recurrent alterations have also been identified in ESCC. Combining the clinicopathological characteristics of patients with information obtained from NGS studies may lead to the development of effective diagnostic and therapeutic approaches for ESCC. As this research becomes more prominent, it is important that gastroenterologist become familiar with the various NGS technologies and the results generated using these methods. In the present study, we describe recent research approaches using NGS in ESCC.
Collapse
|
48
|
Family history of esophageal cancer increases the risk of esophageal squamous cell carcinoma. Sci Rep 2015; 5:16038. [PMID: 26526791 PMCID: PMC4630623 DOI: 10.1038/srep16038] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 10/07/2015] [Indexed: 01/29/2023] Open
Abstract
A population-based case-control was performed to explore familial aggregation of esophageal squamous cell carcinoma (ESCC). Family history of cancer was assessed by a structured questionnaire, and from which 2 cohorts of relatives of cases and controls were reconstructed. Unconditional logistic regression and Cox proportional hazards regression were applied for case-control design and reconstructed cohort design, respectively. We observed a close to doubled risk of ESCC associated with a positive family history of esophageal cancer among first degree relatives (odds ratio [OR] = 1.85, 95% confidence interval [CI]: 1.42–2.41), after adjusting age, sex, family size and other confounders. The excess risks of ESCC increased with the increasing of first-degree relatives affected by esophageal cancer (p < 0.001). In particular, those individuals whose both parents with esophageal cancer had an 8-fold excess risk of ESCC (95% CI: 1.74–36.32). The reconstructed cohort analysis showed that the cumulative risk of esophageal cancer to age 75 was 12.2% in the first-degree relatives of cases and 7.0% in those of controls (hazard ratio = 1.91, 95% CI: 1.54–2.37). Our results suggest family history of esophageal cancer significantly increases the risk for ESCC. Future studies are needed to understand how the shared genetic susceptibility and/or environmental exposures contribute to the observed excess risk.
Collapse
|
49
|
Appelman HD, Matejcic M, Parker MI, Riddell RH, Salemme M, Swanson PE, Villanacci V. Progression of esophageal dysplasia to cancer. Ann N Y Acad Sci 2015; 1325:96-107. [PMID: 25266019 DOI: 10.1111/nyas.12523] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The following, from the 12th OESO World Conference: Cancers of the Esophagus, includes commentaries on the evolution of low-grade squamous and glandular dysplasia to invasive carcinoma; the mutational spectra of Barrett's esophagus and adenocarcinoma; the risk of p53-immunoreactive glandular dysplasia compared to non-immunoreactive mucosa for progression to cancer; the role of lectins in progression to adenocarcinoma; and the role of racemase immunoreactivity in the prediction of risk of adenocarcinoma.
Collapse
Affiliation(s)
- Henry D Appelman
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | | | | | | | | | | | | |
Collapse
|
50
|
Zhao H, Gu X. Silencing of insulin-like growth factor-1 receptor enhances the radiation sensitivity of human esophageal squamous cell carcinoma in vitro and in vivo. World J Surg Oncol 2014; 12:325. [PMID: 25363593 PMCID: PMC4232704 DOI: 10.1186/1477-7819-12-325] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 10/20/2014] [Indexed: 01/18/2023] Open
Abstract
Background Esophageal squamous cell carcinoma (ESCC) is a prevalent fatal cancer worldwide, and the number of deaths due to this disease is increasing. Due to ESCC resistance to chemotherapy and radiation treatment, new therapies are urgently needed for the improvement of ESCC patient clinical outcomes. Methods Eca-109 and TE-1 cells were transfected with 100 nM IGF-1r siRNA, and a combination of IGF-1r siRNA and radiation therapy was tested in vitro and in vivo. The effects of IGF-1r siRNA were determined through Western blotting and flow cytometry experiments. Results After radiotherapy, the number of IGF-1r siRNA-transfected Eca-109 cells decreased by approximately 67.3%, and a 78.9% reduction was observed in the transfected TE-1 cells. In addition, the Eca-109 and TE-1 cells that were irradiated following IGF-1r knockdown contained 16.2% and 20.3% apoptotic cells, respectively. Conclusions The results of the current study suggest that IGF-1r knockdown may enhance the radiation sensitivity of ESCC and increase the therapeutic effects of radiation both in vitro and in vivo. These results provide strong evidence that the targeted application of siRNA will enable the development of new therapeutic strategies for the clinical treatment of ESCC patients.
Collapse
Affiliation(s)
| | - Xiaomeng Gu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Wenhua Western Road 107, Jinan 250012 Shandong Province, China.
| |
Collapse
|