201
|
Ding H, Shi Y, Liu X, Qiu A. MicroRNA-4513 Promotes Gastric Cancer Cell Proliferation and Epithelial-Mesenchymal Transition Through Targeting KAT6B. HUM GENE THER CL DEV 2020; 30:142-148. [PMID: 31310159 DOI: 10.1089/humc.2019.094] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The purpose of this study was to investigate the expression level of microRNA-4513 (miR-4513) in gastric cancer (GC), and to elucidate the mechanisms underlying its regulation of GC progression. Quantitative real-time PCR (qRT-PCR) was performed to measure the expression level of miR-4513 in GC cells. Transfection efficacy of synthetic miRNAs was examined by qRT-PCR. After synthetic miRNA transfection, cell counting kit-8 assay and transwell invasion assay were conducted to measure biological changes in these groups. The key molecular expression level involved in epithelial-mesenchymal transition (EMT) was analyzed by Western blot. Bioinformatic analysis and Western blot were performed to investigate the connection between miR-4513 and lysine acetyltransferase 6B (KAT6B). qRT-PCR results showed that miR-4513 expression level was upregulated in GC cell lines. Downregulation of miR-4513 expression inhibited GC cell proliferation, invasion, and EMT. KAT6B was validated as a direct target of miR-4513. In addition, KAT6B expression level can be upregulated by miR-4513 inhibitor. Collectively, we showed that miR-4513 is involved in regulating the biological function of GC cells via KAT6B. In addition, miR-4513 may serve as a potential target for the molecular therapy of GC.
Collapse
Affiliation(s)
- Huimin Ding
- Department of General Surgery, The First People's Hospital of Yancheng City, Yancheng, P.R. China
| | - Yuhua Shi
- Department of General Surgery, The Third People's Hospital of Yancheng City, Yancheng, P.R. China
| | - Xiaobing Liu
- Department of General Surgery, The Third People's Hospital of Yancheng City, Yancheng, P.R. China
| | - Aifeng Qiu
- Department of General Surgery, The Third People's Hospital of Yancheng City, Yancheng, P.R. China
| |
Collapse
|
202
|
Wang X, Guo J, Dai M, Wang T, Yang T, Xiao X, Tang Q, Zhang L, Jia L. PLOD2 increases resistance of gastric cancer cells to 5-fluorouracil by upregulating BCRP and inhibiting apoptosis. J Cancer 2020; 11:3467-3475. [PMID: 32284742 PMCID: PMC7150456 DOI: 10.7150/jca.41828] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/29/2020] [Indexed: 01/01/2023] Open
Abstract
Background: Gastric cancer (GC) is one of the most common cancers, and it is the third most common cause of cancer-related mortality worldwide. Fluorouracil (5-FU)-based chemotherapy is frequently used for the treatment of advanced GC. However, a substantial proportion of patients eventually experience refractory disease due to drug resistance. PLOD2 was reported to increase invasion and migration in several GC cell lines, but the roles of PLOD2 in chemoresistance are still unclear. The present study aimed to determine whether PLOD2 could confer 5-FU resistance in GC. Methods: The expression of PLOD2 in GC cell lines was assessed by Western blotting. The cells were transfected by lentiviral transduction. The IC50 values were determined by the CCK-8 assay. The migration and invasion abilities of cells were analyzed by the Transwell assay. The proportion of apoptotic cells was assessed by flow cytometry. The protein levels of P-gp (MDR1), MRP1, BCRP (ABCG2), Bax and Bcl2 were analyzed by Western blotting. Furthermore, tumor xenograft models in nude mice were established to test tumor growth and weight. Result: The knockdown of PLOD2 in BGC823 cells significantly decreased the IC50 values of 5-FU. It also contributed to reducing the cell migration and invasion and promoting the apoptosis of GC cells. The opposite results appeared in PLOD2-overexpressing MGC803 GC cells. In vivo experiments showed that the knockdown of PLOD2 increased the growth inhibition of transplanted tumors in nude mice in response to 5-FU. Our mechanistic studies revealed that PLOD2-overexpressing cells appear to be resistant to the therapeutic characteristics of 5-FU in GC cells by upregulating BCRP and that PLOD2 confers resistance to 5-FU-induced apoptosis in GC cells by affecting the expression of Bax and Bcl2. Conclusion: PLOD2 contributed to increasing resistance of gastric cancer cells to 5-fluorouracil by upregulating BCRP and inhibiting apoptosis.
Collapse
Affiliation(s)
- Xiaohui Wang
- Cancer Center, Bayannur Hospital, Bayannur, Inner Mongolia, PR China.,Department of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, PR China
| | - Jiaojiao Guo
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Meng Dai
- Cancer Center, Bayannur Hospital, Bayannur, Inner Mongolia, PR China.,Bayannur Clinical Medical College, Inner Mongolia Medical University, Bayannur, Inner Mongolia, PR China
| | - Tengqi Wang
- Cancer Center, Bayannur Hospital, Bayannur, Inner Mongolia, PR China.,Bayannur Clinical Medical College, Inner Mongolia Medical University, Bayannur, Inner Mongolia, PR China
| | - Tingting Yang
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Xuejun Xiao
- Department of Pharmacology, Xinjiang Medical University, Wulumuqi, Xinjiang, PR China
| | - Qi Tang
- Department of Pathology, Nanjing Medical University, Nanjing, Jiangsu Province, PR China
| | - Lingli Zhang
- Department of Ophthalmology, Inner Mongolia Autonomous Region People's Hospital, Hohhot, PR China
| | - Lizhou Jia
- Cancer Center, Bayannur Hospital, Bayannur, Inner Mongolia, PR China.,Bayannur Clinical Medical College, Inner Mongolia Medical University, Bayannur, Inner Mongolia, PR China
| |
Collapse
|
203
|
The curcumin analogue WZ35 affects glycolysis inhibition of gastric cancer cells through ROS-YAP-JNK pathway. Food Chem Toxicol 2020; 137:111131. [DOI: 10.1016/j.fct.2020.111131] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 01/09/2020] [Accepted: 01/13/2020] [Indexed: 12/11/2022]
|
204
|
Liu Y, Yin L, Chen C, Zhang X, Wang S. Long non-coding RNA GAS5 inhibits migration and invasion in gastric cancer via interacting with p53 protein. Dig Liver Dis 2020; 52:331-338. [PMID: 31530437 DOI: 10.1016/j.dld.2019.08.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 07/30/2019] [Accepted: 08/04/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The rapid progress of gastric cancer (GC) is mainly due to metastasis. Long non-coding RNA (lncRNA) GAS5 has been identified as a tumor suppressor in numerous cancers, and its downregulation in GC has already been reported. AIMS In this study, we planned to investigate the role of GAS5 in GC metastasis. METHODS Gene expressions were detected by qRT-PCR. ISH staining was applied to assess GAS5 level in clinical tissues. Gain-of-function assays were conducted to evaluate the function of GAS5 in GC metastasis. RNA pull down, RIP and cycloheximide assays were performed to confirm the relationship between GAS5 and p53 protein. RESULTS GAS5 expression was markedly decreased in GC tissues and cell lines, and its low expression was strongly related to GC metastasis and unsatisfactory prognosis. GAS5 overexpression repressed GC cell migration and invasion by targeting p53. Intriguingly, GAS5 relied on the exon 12 to interact with and stabilize p53 protein. CONCLUSION Our data implied that GAS5 is a suppressor in GC metastasis via modulating p53 signaling, suggesting GAS5 as a potential therapeutic target for GC, especially for patients with metastasis.
Collapse
Affiliation(s)
- Yongchao Liu
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Lu Yin
- Center for Diffcult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Chunqiu Chen
- Center for Diffcult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Xiangyu Zhang
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| | - Sheng Wang
- Department of Critical Care Medicine, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| |
Collapse
|
205
|
Hou G, Bai Y, Jia A, Ren Y, Wang Y, Lu J, Wang P, Zhang J, Lu Z. Inhibition of autophagy improves resistance and enhances sensitivity of gastric cancer cells to cisplatin. Can J Physiol Pharmacol 2020; 98:449-458. [PMID: 32058824 DOI: 10.1139/cjpp-2019-0477] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Autophagy plays critical roles in tumorigenesis, while the effects of autophagy on chemoresistance of cancer cells had great disparity. This study aims to explore the impacts of autophagy on the sensitivity and resistance of gastric cancer cells to cisplatin (DDP). We firstly demonstrated that there was stronger autophagy activity in gastric cancer SGC-7901 cells than that in DDP-resisting SGC-7901/DDP cells. Then, we discovered that inhibiting autophagy by chloroquine (CQ) significantly enhanced the proliferation-inhibiting and apoptosis-inducing effects of DDP to SGC-7901 and SGC-7901/DDP cells. Moreover, CQ could partially reverse the resistance of SGC-7901/DDP cells to DDP in a concentration-dependent manner. However, the autophagy inducer everolimus (RAD001) had no obvious effects on the sensitivity of gastric cells to DDP. Mechanistically, we demonstrated that CQ might enhance the sensitivity of SGC-7901cells and improve the resistance of SGC-7901/DDP cells to DDP through inhibiting the mTORC1 pathway, especially to SGC-7901/DDP cells. Additionally, we found interfering Beclin-1 using Beclin-1 shRNA also enhanced the proliferation-inhibiting and apoptosis-inducing effects of DDP on gastric cancer cells by inhibiting phosphorylation of Akt. Our study shows that inhibiting autophagy could improve the chemoresistance and enhanced sensitivity of gastric cancer cells to DDP and provide a rationale for the administration of cisplatin combined with CQ for treating patients with gastric cancer.
Collapse
Affiliation(s)
- Guiqin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Yiru Bai
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China.,First Affiliated Hospital of Henan University of Science and Technology, Luoyang, Henan 471000, People's Republic of China
| | - Ang Jia
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Yandan Ren
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Yang Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Jie Lu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Peng Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Jianying Zhang
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Zhaoming Lu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China.,Collaborative Innovation Center of Cancer Chemoprevention, Henan Province, Zhengzhou 450001, People's Republic of China
| |
Collapse
|
206
|
Li Y, Cheng Z, Gevaert O, He L, Huang Y, Chen X, Huang X, Wu X, Zhang W, Dong M, Huang J, Huang Y, Xia T, Liang C, Liu Z. A CT-based radiomics nomogram for prediction of human epidermal growth factor receptor 2 status in patients with gastric cancer. Chin J Cancer Res 2020; 32:62-71. [PMID: 32194306 PMCID: PMC7072015 DOI: 10.21147/j.issn.1000-9604.2020.01.08] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 12/03/2019] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE To develop and validate a computed tomography (CT)-based radiomics nomogram for predicting human epidermal growth factor receptor 2 (HER2) status in patients with gastric cancer. METHODS This retrospective study included 134 patients with gastric cancer (HER2-negative: n=87; HER2-positive: n=47) from April 2013 to March 2018, who were then randomly divided into training (n=94) and validation (n=40) cohorts. Radiomics features were obtained from the CT images showing gastric cancer. Least absolute shrinkage and selection operator (LASSO) regression analysis was utilized for building the radiomics signature. A multivariable logistic regression method was applied to develop a prediction model incorporating the radiomics signature and independent clinicopathologic risk predictors, which were then visualized as a radiomics nomogram. The predictive performance of the nomogram was assessed in the training and validation cohorts. RESULTS The radiomics signature was significantly associated with HER2 status in both training (P<0.001) and validation (P=0.023) cohorts. The prediction model that incorporated the radiomics signature and carcinoembryonic antigen (CEA) level demonstrated good discriminative performance for HER2 status prediction, with an area under the curve (AUC) of 0.799 [95% confidence interval (95% CI): 0.704-0.894] in the training cohort and 0.771 (95% CI: 0.607-0.934) in the validation cohort. The calibration curve of the radiomics nomogram also showed good calibration. Decision curve analysis showed that the radiomics nomogram was useful. CONCLUSIONS We built and validated a radiomics nomogram with good performance for HER2 status prediction in gastric cancer. This radiomics nomogram could serve as a non-invasive tool to predict HER2 status and guide clinical treatment.
Collapse
Affiliation(s)
- Yexing Li
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Shantou University Medical College, Shantou 515041, China
| | - Zixuan Cheng
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Olivier Gevaert
- Stanford Center for Biomedical Informatics Research, Department of Medicine, and Department of Biomedical Data Science, Stanford University, California 94305, USA
| | - Lan He
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Xin Chen
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Southern Medical University, Guangzhou 510515, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou 510180, China
| | - Xiaomei Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Southern Medical University, Guangzhou 510515, China
| | - Xiaomei Wu
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Wen Zhang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Southern Medical University, Guangzhou 510515, China
- Department of Radiology, Zhuhai Hospital of Traditional Chinese and Western Medicine, Zhuhai 519000, China
| | - Mengyi Dong
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Southern Medical University, Guangzhou 510515, China
| | - Jia Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Shantou University Medical College, Shantou 515041, China
| | - Yucun Huang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Southern Medical University, Guangzhou 510515, China
- Department of Radiology, The Fifth People’s Hospital of Zhuhai, Zhuhai 519055, China
| | - Ting Xia
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Shantou University Medical College, Shantou 515041, China
- Changhong Liang, PhD. Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, No.106 Zhongshan Er Road, Guangzhou 510080, China.
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
- Graduate College, Shantou University Medical College, Shantou 515041, China
- Zaiyi Liu, PhD. Department of Radiology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, No.106 Zhongshan Er Road, Guangzhou 510080, China.
| |
Collapse
|
207
|
Ilson DH, Tabernero J, Prokharau A, Arkenau HT, Ghidini M, Fujitani K, Van Cutsem E, Thuss-Patience P, Beretta GD, Mansoor W, Zhavrid E, Alsina M, George B, Catenacci D, McGuigan S, Makris L, Doi T, Shitara K. Efficacy and Safety of Trifluridine/Tipiracil Treatment in Patients With Metastatic Gastric Cancer Who Had Undergone Gastrectomy: Subgroup Analyses of a Randomized Clinical Trial. JAMA Oncol 2020; 6:e193531. [PMID: 31600365 PMCID: PMC6802061 DOI: 10.1001/jamaoncol.2019.3531] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Question Is trifluridine/tipiracil treatment safe and effective for the subpopulation of patients with previously treated metastatic gastric or gastroesophageal junction cancer who have undergone gastrectomy? Findings In this subgroup analysis of a randomized clinical trial, trifluridine/tipiracil treatment improved overall survival and progression-free survival compared with placebo among patients with previously treated metastatic gastric or gastroesophageal junction cancer and who had or had not undergone gastrectomy. No new safety concerns were reported, and hematologic toxic effects were more frequent among the subgroup who had undergone gastrectomy but were treated using dosing modifications. Meaning Trifluridine/tipiracil is a safe and effective treatment option for patients with pretreated metastatic gastric or gastroesophageal junction cancer regardless of previous gastrectomy. Importance Trifluridine/tipiracil (FTD/TPI) treatment has shown clinical benefit in patients with pretreated metastatic gastric cancer or gastroesophageal junction cancer (mGC/GEJC). Patients who have undergone gastrectomy constitute a significant proportion of patients with mGC/GEJC. Objective To assess the efficacy and safety of FTD/TPI among patients with previously treated mGC/GEJC who had or had not undergone gastrectomy. Design, Setting, and Participants This preplanned subgroup analysis of TAGS (TAS-102 Gastric Study), a phase 3, randomized, placebo-controlled, clinical trial included patients with mGC/GEJC who had received at least 2 previous chemotherapy regimens, and was conducted at 110 academic hospitals in 17 countries in Europe, Asia, and North America, with enrollment between February 24, 2016, and January 5, 2018; the data cutoff was March 31, 2018. Interventions Patients were randomized 2:1 to receive oral FTD/TPI 35 mg/m2 twice daily or placebo twice daily with best supportive care on days 1 through 5 and days 8 through 12 of each 28-day treatment cycle. Main Outcomes and Measures The primary end point was overall survival. This subgroup analysis was conducted to examine potential trends and was not powered for statistical significance. Efficacy and safety end points were evaluated in the subgroups. Results Of 507 randomized patients (369 [72.8%] male; mean [SD] age, 62.5 [10.5] years), 221 (43.6%) had undergone gastrectomy (147 randomized to FTD/TPI and 74 to placebo) and 286 (56.4%) had not undergone gastrectomy (190 randomized to FTD/TPI and 96 to placebo). In the gastrectomy subgroup, the overall survival hazard ratio (HR) in the FTD/TPI group vs placebo group was 0.57 (95% CI, 0.41-0.79), and the progression-free survival HR was 0.48 (95% CI, 0.35-0.65). In the no gastrectomy subgroup, the overall survival HR in the FTD/TPI group vs placebo group was 0.80 (95% CI, 0.60-1.06), and the progression-free survival HR was 0.65 (95% CI, 0.49-0.85). Among FTD/TPI-treated patients, grade 3 or higher adverse events of any cause occurred in 122 of 145 patients (84.1%) in the gastrectomy subgroup and 145 of 190 (76.3%) in the no gastrectomy subgroup: 64 (44.1%) in the gastrectomy subgroup and 50 (26.3%) in the no gastrectomy subgroup had grade 3 or higher neutropenia, 31 (21.4%) in the gastrectomy subgroup and 33 (17.4%) in the no gastrectomy subgroup had grade 3 or higher anemia, and 21 (14.5%) in the gastrectomy subgroup and 10 (5.3%) in the no gastrectomy subgroup hD grade 3 or higher leukopenia. In the gastrectomy subgroup, 94 (64.8%) had dosing modifications because of adverse events vs 101 (53.2%) in the no gastrectomy subgroup; 15 (10.3%) in the gastrectomy group and 28 (14.7%) in the no gastrectomy group discontinued treatment because of adverse events. Treatment exposure was similar between groups. Conclusions and Relevance The FTD/TPI treatment was tolerable and provided efficacy benefits among patients with pretreated mGC/GEJC regardless of previous gastrectomy. Trial Registration ClinicalTrials.gov identifier: NCT02500043
Collapse
Affiliation(s)
- David H Ilson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Josep Tabernero
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Hendrik-Tobias Arkenau
- Drug Development Unit, Sarah Cannon Research Institute, Cancer Institute, University College London, London, United Kingdom
| | - Michele Ghidini
- Department of Oncology, Azienda Ospedaliera di Cremona, Cremona, Italy
| | | | - Eric Van Cutsem
- Digestive Oncology, University Hospitals Gasthuisberg Leuven, KU Leuven, Leuven, Belgium
| | - Peter Thuss-Patience
- Department of Hematology, Oncology, and Tumor Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Wasat Mansoor
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Edvard Zhavrid
- Department of Chemotherapy, Alexandrov National Cancer Centre of Belarus, Minsk, Belarus
| | - Maria Alsina
- Department of Medical Oncology, Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ben George
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee
| | - Daniel Catenacci
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Sandra McGuigan
- Medical Affairs Division, Taiho Oncology Inc, Princeton, New Jersey
| | | | - Toshihiko Doi
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| |
Collapse
|
208
|
Li SS, Lian YF, Huang YL, Huang YH, Xiao J. Overexpressing PLOD family genes predict poor prognosis in gastric cancer. J Cancer 2020; 11:121-131. [PMID: 31892979 PMCID: PMC6930397 DOI: 10.7150/jca.35763] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023] Open
Abstract
Procollagen-lysine, 2-oxoglutarate 5-dioxygenases (PLODs) are a set of enzymes involved in the hydroxylation of lysine and stabilization of collagen by crosslinks. Previous studies have highlighted that overexpressing PLOD genes were related to the progression, migration and progression of different human cancers. However, the diverse expression patterns and prognostic values of PLOD genes remain to be elucidated in gastric cancer (GC). In this study, we mined the expression and survival data in GC patients through ONCOMINE, UALCAN and Kaplan-Meier Plotter database. STRING portal couple with DAVID was used to establish a functional protein interaction network of PLOD family genes and analyze the GO and KEGG enriched pathways. Differential gene expression correlated with PLOD family genes was identified with LinkedOmics. We found that PLOD1, 2 and 3 were up-regulated in GC patients compared with normal tissues. High expression levels of PLOD1 and PLOD3 were associated with shorter overall survival (OS), first progression (FP) and post progression survival (PPS) while high expression level of PLOD2 was only associated with shorter FP in all GC patients. Specifically, only high PLOD2 expression had significant correlation with shorter OS, FP and PPS in the diffuse type GC patients. Furthermore, combinatorial use of expressions of all PLOD genes was a superior prognostic indicator for GC patients. Pathway analysis confirmed that PLOD family genes mainly participate in regulating the collagen metabolism and extracellular matrix constitution, and the cellular adaptor protein SHC1, which helps to transduce an extracellular signal into an intracellular signal, could be the regulatory module mediating PLOD's effect on GC. Therefore, we propose that individual PLOD genes or PLOD family genes as a whole could be potential prognostic biomarkers for GC.
Collapse
Affiliation(s)
- Shan-Shan Li
- Department of Medical Oncology, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yi-Fan Lian
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yan-Lin Huang
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yue-Hua Huang
- Guangdong Provincial Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jian Xiao
- Department of Medical Oncology, the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
209
|
Naeli P, Pourhanifeh MH, Karimzadeh MR, Shabaninejad Z, Movahedpour A, Tarrahimofrad H, Mirzaei HR, Bafrani HH, Savardashtaki A, Mirzaei H, Hamblin MR. Circular RNAs and gastrointestinal cancers: Epigenetic regulators with a prognostic and therapeutic role. Crit Rev Oncol Hematol 2020; 145:102854. [PMID: 31877535 PMCID: PMC6982584 DOI: 10.1016/j.critrevonc.2019.102854] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 02/06/2023] Open
Abstract
Both environmental and genetic factors are involved in the initiation and development of gastrointestinal cancer. Covalent closed circular RNAs (circRNAs) are produced by a mechanism called "back-splicing" from mRNAs. They are highly stable and show cell and tissue specific expression patterns. Although some functions such as "microRNA sponge" and "RNA binding protein sponge" have been reported for a small number of circRNAs, the function of thousands of other circRNAs is still unknown. Dysregulation of circRNAs has been reported in many GI cancers and are involved in metastasis and invasion. CircRNAs have been reported to be useful as prognostic markers and targets for developing new treatments. We first describe the properties and biogenesis of circRNAs. We then summarize recent reports about circRNA functions, expression status, and their potential to be used as biomarkers in GI cancers including, gastric cancer, colorectal cancer, esophageal cancer, hepatocellular carcinoma, gallbladder cancer and pancreatic cancer.
Collapse
Affiliation(s)
- Parisa Naeli
- Department of Biological Sciences, Faculty of Genetics, Tarbiat Modares University, Tehran, Iran.
| | | | - Mohammad Reza Karimzadeh
- Department of Medical Genetics, School of Medicine, Bam University of Medical Sciences, Bam, Iran.
| | - Zahra Shabaninejad
- Department of Nanobiotechnology, School of Basic Sciences, TarbiatModares University, Tehran, Iran; Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ahmad Movahedpour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Hossein Tarrahimofrad
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran.
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hassan Hassani Bafrani
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Amir Savardashtaki
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences Shiraz, Iran.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA, 02114, USA.
| |
Collapse
|
210
|
Zhao X, Hu GF, Shi YF, Xu W. Research Progress in microRNA-Based Therapy for Gastric Cancer. Onco Targets Ther 2019; 12:11393-11411. [PMID: 31920330 PMCID: PMC6935305 DOI: 10.2147/ott.s221354] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 12/10/2019] [Indexed: 12/14/2022] Open
Abstract
Gastric cancer (GC) is one of the leading causes of tumor-related mortality. In addition to surgery and endoscopic resection, systemic therapy remains the main treatment option for GC, especially for advanced-stage disease and for cases not suitable for surgical therapy. Hence, improving the efficacy of systemic therapy is still an urgent problem to overcome. In the past decade, the essential roles of microRNAs (miRNAs) in tumor treatment have been increasingly recognized. In particular, miRNAs were recently shown to reverse the resistance to chemotherapy drugs such as 5-fluorouracil, cisplatin, and doxorubicin. Synthesized nanoparticles loaded with mimics or inhibitors of miRNAs can directly target tumor cells to suppress their growth. Moreover, exosomes may serve as promising safe carriers for mimics or inhibitors of miRNAs to treat GC. Some miRNAs have also been shown to play roles in the mechanism of action of other anti-tumor drugs. Therefore, in this review, we highlight the research progress on microRNA-based therapy in GC and discuss the challenges and prospects associated with this strategy. We believe that microRNA-based therapy has the potential to offer a clinical benefit to GC patients, and this review would contribute to and motivate further research to promote this field toward this ultimate goal.
Collapse
Affiliation(s)
- Xu Zhao
- Department of Hepatology, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Gao-Feng Hu
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, People's Republic of China.,Department of Clinical Laboratory, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| | - Yan-Fen Shi
- Department of Pathology, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Wei Xu
- Department of Clinical Laboratory, The First Hospital of Jilin University, Changchun 130021, People's Republic of China
| |
Collapse
|
211
|
Andreollo NA, Drizlionoks E, Tercioti-Junior V, Coelho-Neto JDS, Ferrer JAP, Carvalheira JBC, Lopes LR. ADJUVANT CHEMORADIOTHERAPY AFTER SUBTOTAL OR TOTAL GASTRECTOMY AND D2 LIMPHADENECTOMY INCREASES SURVIVAL IN ADVANCED GASTRIC CANCER? ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA : ABCD = BRAZILIAN ARCHIVES OF DIGESTIVE SURGERY 2019; 32:e1464. [PMID: 31859917 PMCID: PMC6918727 DOI: 10.1590/0102-672020190001e1464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/17/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The treatment of advanced gastric cancer with curative intent is essentially surgical and chemoradiotherapy is indicated as neo or adjuvant to control the disease and prolong survival. AIM To assess the survival of patients undergoing subtotal or total gastrectomy with D2 lymphadenectomy followed by adjuvant chemoradiotherapy. METHODS Were retrospectively analyzed 87 gastrectomized patients with advanced gastric adenocarcinoma, considered stages IB to IIIC and submitted to adjuvant chemoradiotherapy (protocol INT 0116). Tumors of the esophagogastric junction, with peritoneal implants, distant metastases, and those that had a compromised surgical margin or early death after surgery were excluded. They were separated according to the extention of the gastrectomy and analyzed for tumor site and histopathology, lymph node invasion, staging, morbidity and survival. RESULTS The total number of patients who successfully completed the adjuvant treatment was 45 (51.7%). Those who started treatment and discontinued due to toxicity, tumor-related worsening, or loss of follow-up were 10 (11.5%) and reported as incomplete adjuvant. The number of patients who refused or did not start adjuvant treatment was 33 (48.3%). Subtotal gastrectomy was indicated in 60 (68.9%) and total in 27 (31.1%) and this had a shorter survival. The mean resected lymph nodes was 30.8. Staging and number of lymph nodes affected were predictors of worse survival and the more advanced the tumor. Patients undergoing adjuvant therapy with complete chemoradiotherapy showed a longer survival when compared to those who did it incompletely or underwent exclusive surgery. On the other hand, comparing the T4b (IIIB + IIIC) staging patients who had complete adjuvance with those who underwent the exclusive operation or who did not complete the adjuvant, there was a significant difference in survival. CONCLUSION Adjuvant chemoradiotherapy presents survival gain for T4b patients undergoing surgical treatment with curative intent.
Collapse
Affiliation(s)
| | - Eric Drizlionoks
- Digestive Diseases Surgical Unit and Gastrocenter, Campinas, SP, Brazil
| | | | | | | | - José Barreto Campello Carvalheira
- Division of Oncology, Department of Surgery and Internal Medicine, School of Medical Sciences, State University of Campinas - UNICAMP, Campinas, SP, Brazil
| | | |
Collapse
|
212
|
Zhang Y, Wu Y, Zhou X, Yi B, Wang L. Estrogen Receptor Beta Inhibits The Proliferation, Migration, And Angiogenesis Of Gastric Cancer Cells Through Inhibiting Nuclear Factor-Kappa B Signaling. Onco Targets Ther 2019; 12:9153-9164. [PMID: 31807000 PMCID: PMC6842292 DOI: 10.2147/ott.s214529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022] Open
Abstract
Purpose This study aimed to investigate the regulatory roles of estrogen receptor beta (ERβ) on gastric cancer (GC) cells, and reveal the potential mechanisms relating to nuclear factor-kappa B (NF-κB) signaling. Methods GC cell lines SGC7901 and MKN45 were transfected with pEGFP-C1-ERβ to overexpress ERβ, and treated with PMA (a NF-κB activator) to activate NF-κB signaling. The cell proliferation and migration, as well as the formation of vessel-like structures in human venous endothelial cells (HUVECs) were detected. The expression of ERβ, NF-κB p65, p-NF-κB p65, Ki67 (a proliferation marker), vascular endothelial growth factor A (VEGF-A) and matrix metalloproteinase 2 (MMP-2), the DNA binding activity of NF-κB p65, the content of VEGF-A, and the activity of MMP-2 were detected in SGC7901 and MKN45 cells. Results The transfection of pEGFP-C1-ERβ significantly increased the expression of ERβ in SGC7901 and MKN45 cells (P < 0.05). Overexpression of ERβ in SGC7901 and MKN45 cells significantly decreased the cell activity, cell number in G2/M phase, cell migration, the expression of Ki67, VEGF-A and MMP-2, VEGF-A content, MMP-2 activity, as well as the number of vessel-like structures formed by HUVECs (P < 0.05). Overexpression of ERβ also significantly decreased the DNA binding activity and the expression of p-NF-κB p65 in SGC7901 and MKN45 cells (P < 0.05). The anti-tumor effect of ERβ overexpression on GC cells was reversed by the intervention of PMA (P < 0.05). Conclusion Overexpression of ERβ inhibited the proliferation, migration, and angiogenesis of GC cells through inhibiting NF-κB signaling.
Collapse
Affiliation(s)
- Yiping Zhang
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Yahua Wu
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Xufeng Zhou
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Benyi Yi
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| | - Lili Wang
- Department of Biochemistry and Molecular Biology, Basic Medical College of Jiujiang University, Jiujiang City, Jiangxi Province 332000, People's Republic of China
| |
Collapse
|
213
|
An JX, Ma ZS, Ma MH, Shao S, Cao FL, Dai DQ. MiR-1236-3p serves as a new diagnostic and prognostic biomarker for gastric cancer. Cancer Biomark 2019; 25:127-132. [PMID: 31045511 DOI: 10.3233/cbm-171026] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The microRNA plays an important role in tumor progression. MiR-1236-3p acts as a tumor suppressor in various malignancies. OBJECTIVE The aim of present study was to explore the expression of miR-1236-3p in gastric cancer (GC) and its correlation with clinicopathological features, and evaluate the feasibility of using it as a prognostic biomarker in GC. METHODS Seventy-six pairs of tissue specimens were collected from GC patients. MiR-1236-3p expression level was detected by using qRT-PCR. The diagnostic value of miR-1236-3p was evaluated by receiver operating characteristic curve, and Kaplan-Meier method was used to analyze the overall survival. Prognosis analysis was performed using multivariate cox proportional hazards regression analysis. RESULTS The expression of miR-1236-3p was significantly reduced in tumor tissues (P< 0.001). In addition, miR-1236-3p expression was correlated with TNM stage (P= 0.001), lymph node metastasis (P= 0.005) and differentiated degree (P= 0.001). The area under the curve was 0.7016, and its specificity and sensitivity were 60.53% and 73.68%. Kaplan-Meier survival curves showed that patients with high miR-1236-3p expression had better overall survival than those with low expression (P= 0.0190). Multivariate Cox regression analysis showed that the miR-1236-3p expression (P= 0.033) was an independent prognostic factor for overall survival of GC prognosis. CONCLUSIONS The study showed that miR-1236-3p is downregulated in GC tissues, and low expression of miR-1236-3p is associated with a poor prognosis in GC. It may be a new diagnostic and prognostic biomarker for GC.
Collapse
Affiliation(s)
- Jia-Xiang An
- Department of Surgical Oncology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, China.,Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Zhao-Sheng Ma
- Department of Surgical Oncology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, China
| | - Ming-Hui Ma
- Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shuai Shao
- Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Fei-Lin Cao
- Department of Surgical Oncology, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, China
| | - Dong-Qiu Dai
- Department of Gastroenterological Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
214
|
Benzyl Isothiocyanate Induces Apoptosis via Reactive Oxygen Species-Initiated Mitochondrial Dysfunction and DR4 and DR5 Death Receptor Activation in Gastric Adenocarcinoma Cells. Biomolecules 2019; 9:biom9120839. [PMID: 31817791 PMCID: PMC6995572 DOI: 10.3390/biom9120839] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/26/2019] [Accepted: 12/01/2019] [Indexed: 12/11/2022] Open
Abstract
Benzyl isothiocyanate (BITC) is known to inhibit the metastasis of gastric cancer cells but further studies are needed to confirm its chemotherapeutic potential against gastric cancer. In this study, we observed cell shrinkage and morphological changes in one of the gastric adenocarcinoma cell lines, the AGS cells, after BITC treatment. We performed 3-(4,5-dimethyl-2-thiazolyl)-2,5- diphenyl-2H-tetrazolium bromide (MTT) assay, a cell viability assay, and found that BITC decreased AGS cell viability. Reactive oxygen species (ROS) analyses using 2',7'-dichlorofluorescin diacetate (DCFDA) revealed that BITC-induced cell death involved intracellular ROS production, which resulted in mitochondrial dysfunction. Additionally, cell viability was partially restored when BITC-treated AGS cells were preincubated with glutathione (GSH). Western blotting indicated that BITC regulated the expressions of the mitochondria-mediated apoptosis signaling molecules, B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), and cytochrome c (Cyt c). In addition, BITC increased death receptor DR5 expression, and activated the cysteine-aspartic proteases (caspases) cascade. Overall, our results showed that BITC triggers apoptosis in AGS cells via the apoptotic pathways involved in ROS-promoted mitochondrial dysfunction and death receptor activation.
Collapse
|
215
|
Su J, Su B, Xia H, Liu F, Zhao X, Li J, Zhang J, Shi Y, Zeng Y, Zeng X, Ling H, Wu Y, Su Q. RORα Suppresses Epithelial-to-Mesenchymal Transition and Invasion in Human Gastric Cancer Cells via the Wnt/β-Catenin Pathway. Front Oncol 2019; 9:1344. [PMID: 31867273 PMCID: PMC6909819 DOI: 10.3389/fonc.2019.01344] [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: 09/15/2019] [Accepted: 11/15/2019] [Indexed: 12/24/2022] Open
Abstract
Retinoid-related orphan receptor alpha (RORα) is involved in tumor development. However, the mechanisms underlying RORα inhibiting epithelial-to-mesenchymal transition (EMT) and invasion are poorly understood in gastric cancer (GC). This study revealed that the decreased expression of RORα is associated with GC development, progression, and prognosis. RORα suppressed cell proliferation, EMT, and invasion in GC cells through inhibition of the Wnt/β-catenin pathway. RORα overexpression resulted in the decreased Wnt1 expression and the increased RORα interaction with β-catenin, which could lead to the decreased intranuclear β-catenin and p-β-catenin levels, concomitant with downregulated T-cell factor-4 (TCF-4) expression and the promoter activity of c-Myc. The inhibition of Wnt/β-catenin pathway was coupled with the reduced expression of Axin, c-Myc, and c-Jun. RORα downregulated vimentin and Snail and upregulated E-cadherin protein levels in vitro and in vivo. Inversely, knockdown of RORα attenuated its inhibitory effects on Wnt/β-catenin pathway and its downstream gene expression, facilitating cell proliferation, EMT, migration, and invasion in GC cells. Therefore, RORα could play a crucial role in repressing GC cell proliferation, EMT, and invasion via downregulating Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Jian Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Department of Pathology, Second Affiliated Hospital, University of South China, Hengyang, China
| | - Bo Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Key Laboratory for Pharmacoproteomics of Hunan Provincial University, Institute of Pharmacy and Pharmacology, University of South China, Hengyang, China
| | - Hong Xia
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - Fang Liu
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - XiaoHong Zhao
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Department of Gynaecology, Hainan Maternal and Child Health Hospital, Haikou, China
| | - Juan Li
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Department of Gastroenterology, Loudi Center Hospital, Loudi, China
| | - JiZhen Zhang
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Department of Pathology, Affiliated Hospital, Jinggangshan University, Ji'an, China
| | - Ying Shi
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Department of Pathology and Pathophysiology, Xiamen Medical College, Xiamen, China
| | - Ying Zeng
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - Xi Zeng
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - Hui Ling
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - YouHua Wu
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| | - Qi Su
- Hunan Province Key Laboratory of Cancer Cellular and Molecular Pathology, Cancer Research Institute, University of South China, Hengyang, China.,Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, China
| |
Collapse
|
216
|
Ji L, Liu Z, Zhou B, Cai Y, An F, Wang L, Lv Z, Xia M, Yang J, Yuan J, Wang H, Zhou Z, Yang S, Hu L, Zhan Q. Community-Based Pilot Study of a Screening Program for Gastric Cancer in a Chinese Population. Cancer Prev Res (Phila) 2019; 13:73-82. [PMID: 31796467 DOI: 10.1158/1940-6207.capr-19-0372] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/26/2019] [Accepted: 11/26/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Lin Ji
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zengchao Liu
- Wuxi Xinwu District Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Bin Zhou
- Department of Biotechnology, Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Ying Cai
- Department of Pathology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Fangmei An
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Lei Wang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zhifa Lv
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Min Xia
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Jianbo Yang
- Wuxi Xinwu District Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Jianfen Yuan
- Wuxi Xinwu District Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Hui Wang
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Zhiyi Zhou
- Department of Pathology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Shudong Yang
- Department of Pathology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Lei Hu
- Wuxi Xinwu District Center for Disease Control and Prevention, Wuxi, Jiangsu, China
| | - Qiang Zhan
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China.
| |
Collapse
|
217
|
Yang B, Bai Q, Chen H, Su K, Gao C. LINC00665 induces gastric cancer progression through activating Wnt signaling pathway. J Cell Biochem 2019; 121:2268-2276. [PMID: 31736127 DOI: 10.1002/jcb.29449] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Bo Yang
- Department of Oncology Affiliated Hospital of Xuzhou Medical University Xuzhou Jiangsu China
| | - Qingqing Bai
- Department of Oncology Suqian Hospital of Traditional Chinese Medicine Suqian Jiangsu China
| | - Huidong Chen
- Department of Oncology Suqian Hospital of Traditional Chinese Medicine Suqian Jiangsu China
| | - Kun Su
- Department of Oncology Suqian Hospital of Traditional Chinese Medicine Suqian Jiangsu China
| | - Chao Gao
- Department of Oncology Affiliated Hospital of Xuzhou Medical University Xuzhou Jiangsu China
| |
Collapse
|
218
|
Li M, Wang Y, Liu X, Zhang Z, Wang L, Li Y. miR-629 targets FOXO3 to promote cell apoptosis in gastric cancer. Exp Ther Med 2019; 19:294-300. [PMID: 31853302 PMCID: PMC6909679 DOI: 10.3892/etm.2019.8168] [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: 06/27/2018] [Accepted: 11/01/2019] [Indexed: 12/17/2022] Open
Abstract
Gastric cancer (GC) is one of the most aggressive types of human tumor worldwide, and the 5-year survival rate is less than 25%. The transcriptional factor, forkhead box O3 (FOXO3), is regulated by various micro (mi)RNAs and has been reported to be associated with multiple regulatory signaling pathways involved in tumor development. The current study therefore assessed the impact of miR-629 and FOXO3 on gastric cancer. Reverse transcription-quantitative polymerase chain reaction and western blotting were performed to assess the expression of mRNA and protein, respectively. Additionally, the cell proliferation and apoptosis rate were determined via an MTT assay and flow cytometry, respectively. The online database TargetScan predicted that FOXO3 was a target of miR-629. A luciferase reporter assay was also performed to verify that FOXO3 was the direct target of miR-629. The results demonstrated that miR-629 and FOXO3 was upregulated and downregulated in GC tissue, respectively. Furthermore, following transfection with a miR-629 inhibitor, SGC-7901, cell proliferation and apoptosis rate were inhibited and promoted when compared with the control group, respectively. Moreover, after the treatment with SGC-7901, the expression of FOXO3, Bax, Caspase 3 was upregulated, and Bcl-2 was downregulated. Furthermore, the luciferase reporter assay revealed that FOXO3 was the target of miR-629. The results demonstrated that miR-629 and FOXO3 serve vital roles in the development of gastric cancer and may be a future therapeutic target.
Collapse
Affiliation(s)
- Ming Li
- Department of General Surgery, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Department of General Surgery, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Yingxin Wang
- Department of General Surgery, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Xia Liu
- Department of Forensic Pathology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Zhenduo Zhang
- Department of General Surgery, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Liwei Wang
- Department of General Surgery, The First Hospital of Shijiazhuang City, Shijiazhuang, Hebei 050011, P.R. China
| | - Yong Li
- Department of General Surgery, Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| |
Collapse
|
219
|
Lv X, Zhao Y, Zhang L, Zhou S, Zhang B, Zhang Q, Jiang L, Li X, Wu H, Zhao L, Wei M, He M. Development of a novel gene signature in patients without Helicobacter pylori infection gastric cancer. J Cell Biochem 2019; 121:1842-1854. [PMID: 31633246 DOI: 10.1002/jcb.29419] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 10/08/2019] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC) is one of the most fatal common cancers in worldwide. Helicobacter pylori (H. pylori) infection is closely related to the development of GC, although the mechanism is still unclear. In our study, we aim to develop a robust messenger RNA (mRNA) signature associated with H. pylori (-) GC that can sensitively and efficiently predict the prognostic. The RNA-seq expression profile and corresponding clinical data of 598 gastric cancer samples and 63 normal samples obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus database. Using gene set enrichment analysis H. pylori (+) GC and H. pylori (-) GC patients and normal samples to select certain genes for further analysis. Using univariate and multivariate Cox regression model to establish a gene signature for predicting the overall survival (OS). Finally, we identified G2/M related seven-mRNA signature (TGFB1, EGF, MKI67, ILF3, INCENP, TNPO2, and CHAF1A) closely related to the prognosis of patients with H. pylori (-) GC. The seven-mRNA signature was identified to act as an independent prognostic biomarker by stratified analysis and multivariate Cox regression analysis. It was also validated on two test groups from TCGA and GSE15460 and shown that patients with high-risk scores based on the expression of the seven mRNAs had significantly shorter survival times compared to patients with low-risk scores (P < .0001). In this study, we developed a seven-mRNA signature related to G2/M checkpoint from H. pylori (-) GCs that as an independent biomarker potentially with a good performance in predicting OS and might be valuable for the clinical management for patients with GC.
Collapse
Affiliation(s)
- Xuemei Lv
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Yanyun Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Liwen Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Shuqi Zhou
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Bing Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Qiang Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Longyang Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Xueping Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China.,Liaoning Key Laboratory of Molecular Targeted Antitumor Drug Development and Evaluation, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
220
|
New Insights in the Pathogenesis of Cisplatin-Induced Nephrotoxicity. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2019. [DOI: 10.2478/sjecr-2019-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Cisplatin (cis-diamminedichloroplatinum II) is a widely used chemotherapeutic agent. However, efficacy and clinical utility of this drug is significantly limited by severe side effects such as nephrotoxicity which develops due to renal accumulation and bio-transformation in proximal tubular epithelial cells. Cisplatin-induced nephrotoxicity can be manifested as acute kidney injury (AKI), or as different types of tubulopathies, salt wasting, loss of urinary concentrating ability, and magnesium wasting. The attenuation of cisplatin-caused AKI is currently accomplished by hydration, magnesium supplementation or mannitol-induced forced diuresis. However, mannitol treatment causes over-diuresis and consequent dehydration, indicating an urgent need for the clinical use of newly designed, safe and efficacious renoprotective drug, as an additive therapy for high dose cisplatin-treated patients. Accordingly, we emphasized current knowledge regarding molecular mechanisms responsible for cisplatin-caused nephrotoxicity and we described in detail the main clinical manifestations of cisplatin-induced renal dysfunction in order to pave the way for the design of new therapeutic approaches that can minimize detrimental effects of cisplatin in the kidneys. Having in mind that most of cisplatin-induced cytotoxic effects against renal cells are, at the same time, involved in anti-tumor activity of cisplatin, new nephroprotective therapeutic strategies have to prevent renal injury and inflammation without affecting cisplatin-induced toxicity against malignant cells.
Collapse
|
221
|
Kinsella T, Safran H, Wiersma S, DiPetrillo T, Schumacher A, Rosati K, Vatkevich J, Anderson LW, Hill KD, Kunos C, Collins JM. Phase I and Pharmacology Study of Ropidoxuridine (IPdR) as Prodrug for Iododeoxyuridine-Mediated Tumor Radiosensitization in Advanced GI Cancer Undergoing Radiation. Clin Cancer Res 2019; 25:6035-6043. [PMID: 31337643 PMCID: PMC6801071 DOI: 10.1158/1078-0432.ccr-19-0862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/21/2019] [Accepted: 07/17/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE Iododeoxyuridine (IUdR) is a potent radiosensitizer; however, its clinical utility is limited by dose-limiting systemic toxicities and the need for prolonged continuous infusion. 5-Iodo-2-pyrimidinone-2'-deoxyribose (IPdR) is an oral prodrug of IUdR that, compared with IUdR, is easier to administer and less toxic, with a more favorable therapeutic index in preclinical studies. Here, we report the clinical and pharmacologic results of a first-in-human phase I dose escalation study of IPdR + concurrent radiation therapy (RT) in patients with advanced metastatic gastrointestinal (GI) cancers. PATIENTS AND METHODS Adult patients with metastatic GI cancers referred for palliative RT to the chest, abdomen, or pelvis were eligible for study. Patients received IPdR orally once every day × 28 days beginning 7 days before the initiation of RT (37.5 Gy in 2.5 Gy × 15 fractions). A 2-part dose escalation scheme was used, pharmacokinetic studies were performed at multiple time points, and all patients were assessed for toxicity and response to Day 56. RESULTS Nineteen patients were entered on study. Dose-limiting toxicity was encountered at 1,800 mg every day, and the recommended phase II dose is 1,200 mg every day. Pharmacokinetic analyses demonstrated achievable and sustainable levels of plasma IUdR ≥1 μmol/L (levels previously shown to mediate radiosensitization). Two complete, 3 partial, and 9 stable responses were achieved in target lesions. CONCLUSIONS Administration of IPdR orally every day × 28 days with RT is feasible and tolerable at doses that produce plasma IUdR levels ≥1 μmol/L. These results support the investigation of IPdR + RT in phase II studies.
Collapse
Affiliation(s)
- Timothy Kinsella
- Brown University Oncology Group (BrUOG), Providence, Rhode Island.
- EMEK, Inc., Warwick, Rhode Island
| | - Howard Safran
- Brown University Oncology Group (BrUOG), Providence, Rhode Island
| | | | | | | | - Kayla Rosati
- Brown University Oncology Group (BrUOG), Providence, Rhode Island
| | | | | | - Kimberly D Hill
- Leidos Biomedical Research Inc, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Charles Kunos
- Cancer Therapy Evaluation Program (CTEP), NCI, Bethesda, Maryland
| | - Jerry M Collins
- Developmental Therapeutics Program (DTP), NCI, Bethesda, Maryland
| |
Collapse
|
222
|
Mashima T, Iwasaki R, Kawata N, Kawakami R, Kumagai K, Migita T, Sano T, Yamaguchi K, Seimiya H. In silico chemical screening identifies epidermal growth factor receptor as a therapeutic target of drug-tolerant CD44v9-positive gastric cancer cells. Br J Cancer 2019; 121:846-856. [PMID: 31607750 PMCID: PMC6889183 DOI: 10.1038/s41416-019-0600-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 09/11/2019] [Accepted: 09/19/2019] [Indexed: 01/07/2023] Open
Abstract
Background Tumours consist of heterogeneous cancer cells and are likely to contain drug-tolerant cell subpopulations, causing early relapse. However, treatment strategies to eliminate these cells have not been established. Methods We established gastric cancer patient-derived cells (PDCs) to examine the contribution of CD44 splicing variant 9 (CD44v9)-positive cells in gastric cancer drug tolerance. We performed gene expression signature-based in silico screening using JFCR_LinCAGE, our anticancer compound gene expression database and subsequent validation in BALB/c-nu/nu mouse xenograft to identify agents targeting the drug-tolerant cancer cells. Results CD44v9-positive cancer cells were enriched among residual cancer cells after treatment with SN-38, an active metabolic of irinotecan. CD44v9 protein was responsible for this drug resistance. We identified epidermal growth factor receptor (EGFR) inhibitors as agents that can target CD44v9-positive cell populations in gastric cancer PDCs. CD44v9 promoted cell proliferation, and EGFR inhibition attenuated CD44v9 protein expression through downregulation of the AKT and the ERK signalling pathways, leading to preferential suppression of CD44v9-positive cells. Importantly, EGFR inhibitors significantly reduced the number of residual cancer cells after cytotoxic anticancer drug treatment and enhanced the antitumor effect of irinotecan in vivo. Conclusions EGFR inhibitors could be potential agents to eradicate cytotoxic anticancer drug-tolerant gastric cancer cell populations.
Collapse
Affiliation(s)
- Tetsuo Mashima
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Risa Iwasaki
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Naomi Kawata
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,Gastroenterological Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryuhei Kawakami
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Koshi Kumagai
- Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshiro Migita
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Sano
- Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kensei Yamaguchi
- Gastroenterological Medicine, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroyuki Seimiya
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan. .,Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
223
|
Kemi NA, Eskuri M, Pohjanen V, Karttunen TJ, Kauppila JH. Histological assessment of stromal maturity as a prognostic factor in surgically treated gastric adenocarcinoma. Histopathology 2019; 75:882-889. [DOI: 10.1111/his.13934] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/05/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Niko A Kemi
- Cancer and Translational Medicine Research Unit, Medical Research Center University of Oulu and Oulu University Hospital Oulu Finland
| | - Maarit Eskuri
- Cancer and Translational Medicine Research Unit, Medical Research Center University of Oulu and Oulu University Hospital Oulu Finland
| | - Vesa‐Matti Pohjanen
- Cancer and Translational Medicine Research Unit, Medical Research Center University of Oulu and Oulu University Hospital Oulu Finland
| | - Tuomo J Karttunen
- Cancer and Translational Medicine Research Unit, Medical Research Center University of Oulu and Oulu University Hospital Oulu Finland
| | - Joonas H Kauppila
- Cancer and Translational Medicine Research Unit, Medical Research Center University of Oulu and Oulu University Hospital Oulu Finland
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery Karolinska Institutet, Karolinska University Hospital Stockholm Sweden
| |
Collapse
|
224
|
Nie K, Shi L, Wen Y, Pan J, Li P, Zheng Z, Liu F. Identification of hub genes correlated with the pathogenesis and prognosis of gastric cancer via bioinformatics methods. Minerva Med 2019; 111:213-225. [PMID: 31638362 DOI: 10.23736/s0026-4806.19.06166-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Gastric cancer (GC) is the fourth most common cause of cancer-related deaths in the world and 5-year overall survival (OS) rate is less than 10%. So, it is urgent to identified novel diagnostic and prognostic biomarkers. METHODS Twelve GEO (gene expression omnibus) datasets were obtained from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between GC and normal tissues were screened and integrated using limma and RobustRankAggreg (RRA) packages in R software. Protein-protein interaction (PPI) network, GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) analyses for DEGs were conducted via STRING and DAVID, respectively. Moreover, Cox regression model was used to construct a gene prognosis signature. RESULTS Ten genes (COL1A1, CXCL8, COL3A1, SPP1, COL1A2, TIMP1, CXCL1, BGN, MMP3 and SERPINE1) were identified and might be highly related to GC. Further analysis showed high expression of CXCL8, COL3A1, CXCL1, MMP3 and SERPINE1, were significantly associated with late stage of GC. Lastly, we build a seven-gene prognosis signature (CYP19A1, SERPINE1, CGB5, CALCR, ASGR2, CYTL1 and ABCB5), which can give a good prediction of OS. CONCLUSIONS Our article screened out key genes highly associating with GC's developments and prognosis, and it is useful for researcher to further understand GC's molecular basis and direct the synthesis medicine of GC.
Collapse
Affiliation(s)
- Kechao Nie
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Laner Shi
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yi Wen
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jinglin Pan
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Peiwu Li
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhihua Zheng
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Fengbin Liu
- Department of Gastroenterology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China -
| |
Collapse
|
225
|
Prognostic factors associated with survival in a large cohort of gastric cancer patients resected over a decade at a single Italian center: the Cremona experience. Clin Transl Oncol 2019; 22:1004-1012. [PMID: 31599376 DOI: 10.1007/s12094-019-02220-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/25/2019] [Indexed: 12/18/2022]
Abstract
BACKGROUND Incidence of gastric cancer (GC) shows different distribution in Italy, with higher incidence in the north and center. We retrospectively analyzed the clinical data of patients resected at the Hospital of Cremona between January 2007 and December 2016. Available clinical variables were linked with survival to identify possible prognostic factors. MATERIALS AND METHODS Variables analyzed were age, sex, type of surgery, site, histology, invasion, nodal status, resection margins, grade, HER2 status, Helicobacter pylori infection (neo)adjuvant chemotherapy, adjuvant chemoradiotherapy, neutrophil-to-lymphocyte ratio, number of nodes removed and type of lymphadenectomy. Overall survival (OS) was estimated by the Kaplan-Meier method and differences between groups by the log-rank test. Data on OS were analyzed by Cox regression and the final model was obtained using the step-wise method. RESULTS 379 patients were considered, out of which 195 were operated from 2007 to 2011 and 184 from 2012 to 2016. Median follow-up was 25.5 months, median OS 31.3 months and time to recurrence 23.2 months. D2 resection rate increased from 36% (period 2007-2011) to 74% in 2012-2016 (p = 0.01) with a higher mean number of nodes collected (20.98 for 2007-2011 and 23.53 for 2012-2016, p = 0.040). Only 37% of patients received a postoperative treatment. At multivariate analysis, variables associated with OS were age (p = 0.002), stage (p < 0.001), resection margins status (p < 0.001), adjuvant chemotherapy (p < 0.010) and tumor location (cardia vs non-cardia) (p = 0.029). CONCLUSIONS Our analysis shows that completeness of resection and lower stage are strong predictors of long-term survival in GC, providing the rationale for adjuvant and neoadjuvant approaches (chemotherapy, radiotherapy or combined). Cardial GC has worse prognosis compared to distal cancers. TRIAL REGISTRATION NUMBER Service evaluation number 256, protocol 16821/17, date 05 June 2017.
Collapse
|
226
|
Pinheiro RN, Mucci S, Zanatto RM, Picanço Junior OM, Bottino AAG, Fontoura RP, Lopes Filho GDJ. Quality of life as a fundamental outcome after curative intent gastrectomy for adenocarcinoma: lessons learned from patients. J Gastrointest Oncol 2019; 10:989-998. [PMID: 31602337 DOI: 10.21037/jgo.2019.06.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Gastric cancer has an important epidemiologic impact, and the main curative therapeutic modality for gastric cancer is surgical resection. However, even curative intent therapy can have negative effects on the quality of life (QoL) of these patients, which is undesirable; thus, it is difficult to balance the standardized treatment reported in the literature and treatment response to achieve full patient satisfaction. The purpose of our study was to evaluate the QoL and identify the association of scores on the Functional Assessment of Cancer Therapy-Gastric (FACT-Ga) and Short Form 36 Health Survey version 2 (SF36v2) questionnaires with sociodemographic, clinical and anatomopathological aspects of gastric adenocarcinoma patients undergoing curative surgery. Methods This was a cross-sectional study involving 104 patients from three regions of Brazil. Inferential analyses were used to compare (multiple regression and Mann-Whitney or Kruskal-Wallis tests) the relationships between these scores and variables (Spearman's coefficient). Results In the multiple regression analysis, we found correlations between Helicobacter pylori status and physical well-being (PWB) (P=0.026), between gender and emotional well-being (EWB) (P=0.008), between Lauren's histology and physical functioning (P=0.009), as well as the Short Form 36 Health Survey version 2 (SF-36v2) role-physical (P=0.027), between the tumor site and EWB (P=0.038), between the SF-36v2 mental health and N (the lower the staging, the better the score, P=0.006) and between the SF-36v2 mental health and lymph nodes removed (P=0.029). According to the Mann-Whitney or Kruskal-Wallis test, women had worse FACT-Ga total (P=0.049), PWB (P=0.005), EWB (P=0.007), gastric cancer subscale (GaCS, P=0.011), trial outcome index (TOI, P=0.030) and mental health scores than men (P=0.011). Patients with distal tumors had better scores (FACT-Ga, P=0.018; GaCS, P=0.014; TOI, P=0.020) than patients with proximal tumors. Patients with tumors located in the cardia had better physical functioning than those with proximal tumors (P=0.042). Patients who underwent partial gastrectomy had better FACT-Ga total scores (P=0.011), PWB (P=0.033), GaCS scores (P=0.006) and TOI scores (P=0.008) than those who underwent total gastrectomy. Patients who did not receive adjuvant therapy had worse bodily pain as reported on the SF-36v2 than those who received therapy (P=0.048). According to Spearman's coefficient, a higher lymph node stage corresponded to worse FACT-Ga total (s=-0.200, P=0.034), GaCS (s=-0.206, P=0.037), TOI (s=-0.216; P=0.028) and vitality (s=-0.215, P=0.029) scores. A longer time after treatment corresponded to a better SF-36v2 role-physical domain score (s=0.223; P=0.023). Conclusions The type of treatment instituted, postoperative time and sociodemographic and anatomopathological factors influence the QoL.
Collapse
Affiliation(s)
- Rodrigo Nascimento Pinheiro
- Postgraduate Program in Interdisciplinary Surgical Science, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil.,Federal District Surgical Oncology Unity, Base Hospital Institute, Brasília, Brazil.,Academic League of Oncology, Base Hospital Institute, Brasília, Brazil
| | - Samantha Mucci
- Postgraduate Program in Interdisciplinary Surgical Science, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil.,Psychiatry Department, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Renato Morato Zanatto
- Postgraduate Program in Interdisciplinary Surgical Science, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil.,Amaral Carvalho Cancer Hospital, Jaú, São Paulo, Brazil
| | - Olavo Magalhães Picanço Junior
- Postgraduate Program in Interdisciplinary Surgical Science, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil.,High Complexity Unity in Oncology, Alberto Lima Hospital, Federal University of Amapá, Macapá, Brazil
| | | | | | - Gaspar de Jesus Lopes Filho
- Postgraduate Program in Interdisciplinary Surgical Science, Paulista School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
227
|
Yang JC, Chang N, Wu DC, Cheng WC, Chung WM, Chang WC, Lei FJ, Liu CJ, Wu IC, Lai HC, Ma WL. Preclinical evaluation of exemestane as a novel chemotherapy for gastric cancer. J Cell Mol Med 2019; 23:7417-7426. [PMID: 31557413 PMCID: PMC6815818 DOI: 10.1111/jcmm.14605] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/18/2019] [Accepted: 07/23/2019] [Indexed: 12/14/2022] Open
Abstract
CYP19A1/aromatase (Ar) is a prognostic biomarker of gastric cancer (GCa). Ar is a critical enzyme for converting androstenedione to oestradiol in the steroidogenesis cascade. For decades, Ar has been targeted with Ar inhibitors (ARIs) in gynaecologic malignancies; however, it is unexplored in GCa. A single‐cohort tissue microarray examination was conducted to study the association between Ar expression and disease outcome in Asian patients with GCa. The results revealed that Ar was a prognostic promoter. Bioinformatics analyses conducted on a Caucasian‐based cDNA microarray databank showed Ar to be positively associated with GCa prognosis for multiple clinical modalities, including surgery, 5‐Fluorouracil (5‐FU) for adjuvant chemotherapy, or HER2 positivity. These findings imply that targeting Ar expression exhibits a potential for fulfilling unmet medical needs. Hence, Ar‐targeting compounds were tested, and the results showed that exemestane exhibited superior cancer‐suppressing efficacy to other ARIs. In addition, exemestane down‐regulated Ar expression. Ablating Ar abundance with short hairpin (sh)Ar could also suppress GCa cell growth, and adding 5‐FU could facilitate this effect. Notably, adding oestradiol could not prevent exemestane or shAr effects, implicating a nonenzymatic mechanism of Ar in cancer growth. Regarding translational research, treatment with exemestane alone exhibited tumour suppression efficacy in a dose‐dependent manner. Combining subminimal doses of 5‐FU and exemestane exerted an excellent tumour suppression effect without influencing bodyweight. This study validated the therapeutic potentials of exemestane in GCa. Combination of metronomic 5‐FU and exemestane for GCa therapy is recommended.
Collapse
Affiliation(s)
- Juan-Cheng Yang
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan
| | - Ning Chang
- Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Deng-Chyang Wu
- Department of Medicine, Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Wei-Chung Cheng
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Min Chung
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Chun Chang
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Fu-Ju Lei
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Chung-Jung Liu
- Department of Medicine, Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - I-Chen Wu
- Department of Medicine, Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hsueh-Chou Lai
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan
| | - Wen-Lung Ma
- Department of Gastroenterology, Chinese Medicine Research and Development Center, Sex Hormone Research Center, Research Center for Tumor Medicine, China Medical University Hospital, Taichung, Taiwan.,Department of OBS & GYN, China Medical University Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medicine, Graduate Institute of BioMedical Sciences, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Medicine, China Medical University, Taichung, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
228
|
Wang Z, Ni F, Yu F, Cui Z, Zhu X, Chen J. Prognostic significance of mRNA expression of CASPs in gastric cancer. Oncol Lett 2019; 18:4535-4554. [PMID: 31611962 PMCID: PMC6781674 DOI: 10.3892/ol.2019.10816] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 07/16/2019] [Indexed: 12/28/2022] Open
Abstract
Current studies suggest that the cysteinyl aspartate specific proteinase (caspase/CASP) family may be closely associated with apoptosis. Scientists have suggested that caspases may be a key to the development of more effective anti-cancer therapies. However, the prognostic value of CASP expression in gastric cancer (GC) remains unclear. Using a Kaplan-Meier plotter online database, the predictive prognostic significance of the expression of 12 CASPs genes (CASP1, CASP2, CASP3, CASP4, CASP5, CASP6, CASP7, CASP8, CASP9, CASP10, CASP12 and CASP14) to overall survival (OS) in different clinicopathological features, including Lauren classification, pathological stages, therapies employed and differentiation in gastric cancer patients was explored. The present study revealed that higher CASP1, 2, 3, 4, 5, 6, 7 and 8 mRNA expression was associated with better OS, whereas higher expression of CASP9, 10, 12 and 14 showed an unfavorable OS in all GC patients. Moreover, CASP1 to 8 were all associated with favorable OS in intestinal type and diffuse type classified by Lauren classification. Therefore, the results of the present study suggested that the CASP family may function as new prognostic indicators in GC and may be helpful in making treatment decisions.
Collapse
Affiliation(s)
- Zixiang Wang
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Fubiao Ni
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Fangyi Yu
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhonghui Cui
- First School of Clinical Medicine, Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiandong Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Jicai Chen
- Department of General Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
229
|
Xin L, Zhou Q, Yuan YW, Zhou LQ, Liu L, Li SH, Liu C. METase/lncRNA HULC/FoxM1 reduced cisplatin resistance in gastric cancer by suppressing autophagy. J Cancer Res Clin Oncol 2019; 145:2507-2517. [PMID: 31485766 DOI: 10.1007/s00432-019-03015-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 08/28/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Autophagy plays an important role in regulating cisplatin (CDDP) resistance in gastric cancer cells. However, the underlying mechanism of methioninase (METase) in the regulation of autophagy and CDDP resistance of gastric cancer cells is still not clear. MATERIALS AND METHODS Western blot was used to detect the levels of autophagy-related proteins, multidrug-resistant 1 (MDR-1), and FoxM1 protein. LncRNA HULC was detected by qRT-PCR. Cell viability was detected using CCK-8 assay. The interaction between lncRNA HULC and FoxM1 was confirmed by RNA pull-down and RIP assay. RESULTS Lentiviral vector carrying METase (LV-METase) suppressed autophagy and CDDP resistance of drug-resistant gastric cancer cells. LncRNA HULC was significantly downregulated in drug-resistant gastric cancer cells transfected with LV-METase. Besides, we found that lncRNA HULC interacted with FoxM1. In addition, METase suppressed autophagy to reduce CDDP resistance of drug-resistant gastric cancer cells through regulating HULC/FoxM1, and interfering HULC suppressed autophagy to reduce CDDP resistance of drug-resistant gastric cancer cells through regulating FoxM1. Finally, interfering HULC inhibited tumor growth in vivo. CONCLUSION METase suppressed autophagy to reduce CDDP resistance of drug-resistant gastric cancer cells through regulating HULC/FoxM1 pathway.
Collapse
Affiliation(s)
- Lin Xin
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Qi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yi-Wu Yuan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Li-Qiang Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Li Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Shi-Hao Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Chuan Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
230
|
Okazaki M, Oyama K, Kinoshita J, Miyashita T, Tajima H, Takamura H, Ninomiya I, Fushida S, Ohta T. Incidence of and risk factors for totally implantable vascular access device complications in patients with gastric cancer: A retrospective analysis. Mol Clin Oncol 2019; 11:343-348. [PMID: 31475061 PMCID: PMC6713938 DOI: 10.3892/mco.2019.1897] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 06/27/2019] [Indexed: 01/17/2023] Open
Abstract
Totally implantable vascular access devices (TIVADs) are often used to administer chemotherapy by prolonged intravenous infusion. The objective of the present study was to investigate the incidence of long-term complications and identify risk factors associated with TIVAD placement in patients with gastric cancer. A total of 121 patients with gastric cancer who had undergone 150 TIVAD placement procedures for chemotherapy or supportive care were enrolled in the present retrospective cohort study. A number of risk factors were analyzed, including age, sex, hypertension, diabetes mellitus, history of thrombosis, body mass index, disease stage, and site and purpose of TIVAD. In total, 40 TIVADs (26.7%) developed long-term complications, of which 27 (18.0%) were infections, seven (4.7%) were catheter-related deep vein thrombosis (CR-DVT), and six (4.0%) were obstructions. Chemotherapy was associated with an increased rate of infectious adverse events (odds ratio 2.925; 95% CI, 1.104-7.750; P=0.031) according to the multivariate analysis. CR-DVT occurred more frequently in upper arm ports than in chest wall ports; however, this difference was not statistically significant (7.5 vs. 0.0%; P=0.084) according to the univariable analysis. All CR-DVTs developed in the upper arm sites. Chemotherapy and the upper arm site were associated with long-term complications in patients with TIVAD. However, further studies are needed to confirm the findings of the present study and to determine the reasons for the high incidence of long-term complications in these patients.
Collapse
Affiliation(s)
- Mitsuyoshi Okazaki
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Katsunobu Oyama
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Jun Kinoshita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Tomoharu Miyashita
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Hidehiro Tajima
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Hiroyuki Takamura
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Itasu Ninomiya
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Sachio Fushida
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| | - Tetsuo Ohta
- Department of Gastroenterological Surgery, Division of Cancer Medicine, Graduate School of Medical Science, Kanazawa University, Kanazawa, Ishikawa 920-8641, Japan
| |
Collapse
|
231
|
Song L, Wang XY, He XF. A 5-Gene Prognostic Combination for Predicting Survival of Patients with Gastric Cancer. Med Sci Monit 2019; 25:6313-6320. [PMID: 31422414 PMCID: PMC6713029 DOI: 10.12659/msm.914815] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Background The aim of the study was to identify a multigene prognostic factor in patients with gastric cancer (GC). Material/Methods Random survival forest (RSF) was performed to screen survival-related genes and develop a multigene combination based on the cumulative hazard function of each GC patient in TCGA-STAD and GSE15459. Kaplan-Meier curve and univariate and multivariable Cox proportional hazards regression model were applied to evaluate the prognostic performance of the 5-gene combination. C-index was used to compare the prognostic performance of the 5-gene combination and another 9-gene signature in GC. Gene set enrichment analysis (GSEA) was conducted. Results We obtained 19 survival-related genes through univariate Cox proportional hazards analysis in the training set, 5 of which were identified and were used to develop a 5-gene combination through RSF. Patients in the 5-gene combination low-risk group had better overall survival (OS) than those in the 5-gene combination high-risk group, and the 5-gene combination was demonstrated to be an independent prognostic factor in patients with GC. The 5-gene combination outperformed the 9-gene signature in predicting the OS of GC patients, and it might affect the prognosis of GC patients through E2F signaling, MYC signaling, and G2M checkpoint. Conclusions We introduce a 5-gene combination that can predict the survival of GC patients and might be an independent prognostic factor in GC.
Collapse
Affiliation(s)
- Liang Song
- Endoscopy Room, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| | - Xiao-Yan Wang
- Department of Epidemiology and Health Statistics, Basic Medical College of Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, China (mainland)
| | - Xiao-Feng He
- Department of Science and Education, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China (mainland)
| |
Collapse
|
232
|
Yin X, Zhang F, Guo Z, Kong W, Wang Y. Integrative analysis of miRNA and mRNA expression profiles reveals a novel mRNA/miRNA signature to improve risk classification for patients with gastric cancer. Oncol Lett 2019; 18:2330-2339. [PMID: 31402938 PMCID: PMC6676680 DOI: 10.3892/ol.2019.10536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 05/25/2019] [Indexed: 02/03/2023] Open
Abstract
Gastric cancer (GC) is one of the most common types of malignant cancer and is associated with poor prognosis. Although the prognosis of patients with GC is associated with grade, stage and lymph node metastases, these traditional clinical features are inadequate to predict the outcome of GC. Therefore, there has been an increased focus on identifying novel molecular biomarkers for early diagnosis and prognosis, in order to improve outcomes in GC. In the present study, an integrative analysis of microRNA (miRNA) expression profiles, mRNA expression profiles and clinical characteristics was performed in a large cohort of patients with GC in order to identify an integrative prognostic model for improving postoperative risk classification. An integrative mRNA/miRNA signature (IMMIS), comprised of three miRNAs and one mRNA, was identified from a large number of differentially expressed miRNAs and mRNAs using univariate and multivariate Cox regression analysis. The prognostic value of the IMMIS was validated in the discovery cohort, testing cohort and The Cancer Genome Atlas (TCGA) cohort. The present results suggested that the identified signature had a reliable predictive performance and could classify the patients into high- and low-risk groups with significantly different overall survival times. In the discovery cohort, the hazard ratio (HR) was 2.805 with a 95% CI=1.722–4.567 (P<0.001). The median overall survival time as 1.49 vs. 3.85 years. In the testing cohort, the HR was 1.625 with a 95% CI=1.004–2.638 (P=0.039) and the median overall survival time was 2.17 vs. 4.62 years. In the TCGA cohort, the HR was 2.139 with a 95% CI=1.519–3.012 (P<0.001) and the median overall survival time was 1.53 vs. 4.62 years. The IMMIS constituted a reliable independent prognostic factor compared with clinical covariates, including age, sex, grade and stage, as indicated by multivariate and stratified analyses. Furthermore, comparative analysis revealed that the predictive value of the IMMIS was superior to the mRNA-based signature alone. The present results suggested the potential value of the IMMIS as a promising novel biomarker for improving the clinical management of patients with GC.
Collapse
Affiliation(s)
- Xiang Yin
- Department of Minimally Invasive Tumor Surgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163453, P.R. China
| | - Fumin Zhang
- Department of Minimally Invasive Tumor Surgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163453, P.R. China
| | - Zhongwu Guo
- Department of Minimally Invasive Tumor Surgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163453, P.R. China
| | - Weiyuan Kong
- Department of Minimally Invasive Tumor Surgery, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163453, P.R. China
| | - Yuanyuan Wang
- Department of Gastroenterology, Daqing Long Nan Hospital, Daqing, Heilongjiang 163453, P.R. China
| |
Collapse
|
233
|
Yu X, Wei D, Gao Y, Du H, Yu B, Li R, Qian C, Luo X, Yuan S, Wang J, Sun L. Synergistic combination of DT-13 and Topotecan inhibits aerobic glycolysis in human gastric carcinoma BGC-823 cells via NM IIA/EGFR/HK II axis. J Cell Mol Med 2019; 23:6622-6634. [PMID: 31397978 PMCID: PMC6787456 DOI: 10.1111/jcmm.14523] [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] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
DT-13 combined with topotecan (TPT) showed stronger antitumour effects in mice subcutaneous xenograft model compared with their individual effects in our previous research. Here, we further observed the synergistically effect in mice orthotopic xenograft model. Metabolomics analysis showed DT-13 combined with TPT alleviated metabolic disorders induced by tumour and synergistically inhibited the activity of the aerobic glycolysis-related enzymes in vivo and in vitro. Mechanistic studies revealed that the combination treatment promoted epidermal growth factor receptor (EGFR) degradation through non-muscle myosin IIA (NM IIA)-induced endocytosis of EGFR, further inhibited the activity of hexokinase II (HK II), and eventually promoted the aerobic glycolysis inhibition activity more efficiently compared with TPT or DT-13 monotherapy. The combination therapy also inhibited the specific binding of HK II to mitochondria. When using the NM II inhibitor (-)002Dblebbistatin or MYH-9 shRNA, the synergistic inhibition effect of DT-13 and TPT on aerobic glycolysis was eliminated in BGC-823 cells. Immunohistochemical analysis revealed selective up-regulation of NM IIA while specific down-regulation of p-CREB, EGFR, and HK II by the combination therapy. Collectively, these findings suggested that this regimen has significant clinical implications, warranted further investigation.
Collapse
Affiliation(s)
- Xiao‐Wen Yu
- Jiangsu Key Laboratory for Drug ScreeningChina Pharmaceutical UniversityNanjingChina
- Nanjing Key Laboratory of PediatricsChildren's Hospital of Nanjing Medical UniversityNanjingChina
| | - Dandan Wei
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, State Key Laboratory Cultivation Base for TCM Quality and EfficacyNanjing University of Chinese MedicineNanjingChina
| | - Ying‐Sheng Gao
- Jiangsu Center for Pharmacodynamics Research and EvaluationChina Pharmaceutical UniversityNanjingChina
| | - Hong‐Zhi Du
- School of PharmacyHubei University of Chinese MedicineWuhanChina
| | - Bo‐Yang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Department of Complex Prescription of TCMChina Pharmaceutical UniversityNanjingChina
| | - Rui‐Ming Li
- Tasly Research InstituteTianjin Tasly Holding Group Co. Ltd.TianjinChina
| | - Chang‐Min Qian
- Tasly Research InstituteTianjin Tasly Holding Group Co. Ltd.TianjinChina
| | - Xue‐Jun Luo
- Tasly Research InstituteTianjin Tasly Holding Group Co. Ltd.TianjinChina
| | - Sheng‐Tao Yuan
- Jiangsu Center for Pharmacodynamics Research and EvaluationChina Pharmaceutical UniversityNanjingChina
| | - Jun‐Song Wang
- Center for Molecular MetabolismNanjing University of Science & TechnologyNanjingChina
| | - Li Sun
- Jiangsu Key Laboratory for Drug ScreeningChina Pharmaceutical UniversityNanjingChina
| |
Collapse
|
234
|
He H, Wu W, Sun Z, Chai L. MiR-4429 prevented gastric cancer progression through targeting METTL3 to inhibit m 6A-caused stabilization of SEC62. Biochem Biophys Res Commun 2019; 517:581-587. [PMID: 31395342 DOI: 10.1016/j.bbrc.2019.07.058] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/17/2019] [Indexed: 02/06/2023]
Abstract
Gastric cancer (GC) has been recognized as the major reason for global cancer-associated mortality. SEC62 homolog, preprotein translocation factor (SEC62) has been documented to possess carcinogenic functions in cancers, but its influence on GC remains elusive. Present study aimed to uncover the impact and mechanism of SEC62 in GC. We validated the upregulation of SEC62 in GC samples by GEPIA, and revealed its high level in GC cell lines. Functionally, depletion of SEC62 hindered proliferation and encouraged apoptosis in GC cells. Furthermore, we found through Starbase 3.0 and validated that methyltransferase like 3 (METTL3) interacted with SEC62 to induce the m6A on SEC62 mRNA, therefore facilitated the stabilizing effect of IGF2 binding protein 1 (IGF2BP1) on SEC62 mRNA. Moreover, we predicted through miRmap and validated that miR-4429 targeted and inhibited METTL3 to repress SEC62. Rescue assays demonstrated that miR-4429 inhibited GC progression through METTL3/SEC62 axis. Together, our study firstly revealed that miR-4429 prevented gastric cancer progression through targeting METTL3 to inhibit m6A-caused stabilization of SEC62, indicating miR-4429 as a promising target for treatment improvement for GC.
Collapse
Affiliation(s)
- Huabo He
- Department of Gastrointestinal and Hepatobiliary Surgery, The Affiliated Hospital of Hangzhou Normal University, 126 Wenzhou Road, Hangzhou, Zhejiang, 310000, PR China
| | - Wei Wu
- Department of Gastrointestinal and Hepatobiliary Surgery, The Affiliated Hospital of Hangzhou Normal University, 126 Wenzhou Road, Hangzhou, Zhejiang, 310000, PR China
| | - Zhener Sun
- Department of Gastrointestinal and Hepatobiliary Surgery, The Affiliated Hospital of Hangzhou Normal University, 126 Wenzhou Road, Hangzhou, Zhejiang, 310000, PR China
| | - Lixin Chai
- Department of Gastrointestinal and Hepatobiliary Surgery, The Affiliated Hospital of Hangzhou Normal University, 126 Wenzhou Road, Hangzhou, Zhejiang, 310000, PR China.
| |
Collapse
|
235
|
Hu X, Xiang J, Xiao B, Huang Y, Xie J. Wogonoside promotes apoptosis in gastric cancer AGS and SGC-7901 cells through induction of mitochondrial dysfunction and endoplasmic reticulum stress. FEBS Open Bio 2019; 9:1469-1476. [PMID: 31250981 PMCID: PMC6668368 DOI: 10.1002/2211-5463.12693] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 02/20/2019] [Accepted: 06/26/2019] [Indexed: 12/16/2022] Open
Abstract
Wogonoside (Wg), a natural flavonoid, has anticancer effects against several human cancers. The purpose of the present study was to investigate the antitumor effects and underlying mechanisms of Wg on gastric cancer (GC) cell lines. We report that Wg treatment inhibited cell viability and induced apoptosis in human GC cell lines AGS and SGC-7901, and also retarded GC tumor growth in xenograft mice in vivo. We also found that the Wg exerted its antitumor effects against GC cells via induction of reaction oxygen species accumulation, mitochondrial dysfunction, and endoplasmic reticulum stress. Furthermore, C/EBP homologous protein knockdown inhibited apoptosis and increased the viability of Wg-treated GC cells. Our findings may facilitate the development of novel therapeutic agents for the treatment of GC.
Collapse
Affiliation(s)
- Xiao‐Miao Hu
- Department of Gastrointestinal SurgeryThe First Hospital affiliated to Yangtze UniversityJingzhouChina
| | - Jin‐Jian Xiang
- Department of Gastrointestinal SurgeryThe First Hospital affiliated to Yangtze UniversityJingzhouChina
| | - Bao‐Lai Xiao
- Department of Gastrointestinal SurgeryThe First Hospital affiliated to Yangtze UniversityJingzhouChina
| | | | - Jian‐Ping Xie
- Department of Gastrointestinal SurgeryThe First Hospital affiliated to Yangtze UniversityJingzhouChina
| |
Collapse
|
236
|
The potential role of TNFAIP3 in malignant transformation of gastric carcinoma. Pathol Res Pract 2019; 215:152471. [DOI: 10.1016/j.prp.2019.152471] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/14/2019] [Accepted: 05/23/2019] [Indexed: 11/22/2022]
|
237
|
Russi S, Verma HK, Laurino S, Mazzone P, Storto G, Nardelli A, Zoppoli P, Calice G, La Rocca F, Sgambato A, Lucci V, Falco G, Ruggieri V. Adapting and Surviving: Intra and Extra-Cellular Remodeling in Drug-Resistant Gastric Cancer Cells. Int J Mol Sci 2019; 20:ijms20153736. [PMID: 31370155 PMCID: PMC6695752 DOI: 10.3390/ijms20153736] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 07/26/2019] [Accepted: 07/29/2019] [Indexed: 02/06/2023] Open
Abstract
Despite the significant recent advances in clinical practice, gastric cancer (GC) represents a leading cause of cancer-related deaths in the world. In fact, occurrence of chemo-resistance still remains a daunting hindrance to effectiveness of the current approach to GC therapy. There is accumulating evidence that a plethora of cellular and molecular factors is implicated in drug-induced phenotypical switching of GC cells. Among them, epithelial-mesenchymal transition (EMT), autophagy, drug detoxification, DNA damage response and drug target alterations, have been reported as major determinants. Intriguingly, resistant GC phenotype may be the result of GC cell-induced tumor microenvironment (TME) remodeling, which is currently emerging as a key player in promoting drug resistance and overcoming cytotoxic effects of drugs. In this review, we discuss the possible mechanisms of drug resistance and their involvement in determining current GC therapies failure.
Collapse
Affiliation(s)
- Sabino Russi
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Henu Kumar Verma
- Institute of Experimental Endocrinology and Oncology, National Research Council, 80131 Naples, Italy
- Section of Stem Cell and Development, Istituto di Ricerche Genetiche "Gaetano Salvatore" Biogem s.c. a.r.l., 83031 Ariano Irpino, Italy
| | - Simona Laurino
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Pellegrino Mazzone
- Section of Stem Cell and Development, Istituto di Ricerche Genetiche "Gaetano Salvatore" Biogem s.c. a.r.l., 83031 Ariano Irpino, Italy
| | - Giovanni Storto
- Department of Nuclear Medicine, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Anna Nardelli
- Istituto di Biostrutture e Bioimmagini, Consiglio Nazionale delle Ricerche, 80145 Napoli, Italy
| | - Pietro Zoppoli
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Giovanni Calice
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Francesco La Rocca
- Laboratory of Clinical Research and Advanced Diagnostics, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Alessandro Sgambato
- Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy
| | - Valeria Lucci
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy
| | - Geppino Falco
- Section of Stem Cell and Development, Istituto di Ricerche Genetiche "Gaetano Salvatore" Biogem s.c. a.r.l., 83031 Ariano Irpino, Italy.
- Department of Biology, University of Naples Federico II, 80126 Naples, Italy.
| | - Vitalba Ruggieri
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture (PZ), Italy.
| |
Collapse
|
238
|
Lin JX, Lin JP, Desiderio J, Xie JW, Gemini A, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Tu R, Zheng CH, Li P, Parisi A, Huang CM. Difference in the short-term outcomes of laparoscopic gastrectomy for gastric carcinoma between the east and west: a retrospective study from the IMIGASTRIC trial. J Cancer 2019; 10:4106-4113. [PMID: 31417655 PMCID: PMC6692614 DOI: 10.7150/jca.31192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 04/23/2019] [Indexed: 12/23/2022] Open
Abstract
Purpose: To compare the clinicopathologic data and short-term surgical outcomes of laparoscopic gastrectomy (LG) for gastric cancer (GC) between the east and west. Methods: Patient demographics, surgical procedures, pathological information, and postoperative recovery were compared among gastric cancer patients who underwent LG in the clinical trial of IMIGASTRIC (NCT02325453) between 2009 and 2016. Results: More younger males, higher BMI, lower ASA score and less neoadjvant chemotherapy were evident in east patient cohort. Eastern patients had a higher proportion of proximal, differentiated and advanced gastric cancers. More total gastrectomies, larger extent of lymph node (LN) dissection, and higher number of retrieved LNs were found in the eastern patients. However, more Roux-en-Y anastomosis procedures during distal gastrectomy and intra-corporeal anastomosis were performed in the western patients. The west patients showed faster postoperative recovery than the eastern patients. The mortality rates of the western patients were comparable to those of the eastern patients. However, fewer III-IV complications were evident in the eastern centers. Multivariate analyses revealed that an elderly age, higher ASA score, and more blood loss were the significant independent risk factors of postoperative complications for eastern patients. However, for the western patients, the independent risk factors were neoadjuvant therapy, more retrieval LNs, and pT3-4 stage. Conclusions: The selections and short-term surgical outcomes of LG for GC were widely different between East and West. To obtain more objective and accurate results, these differences should be considered in future international prospective studies.
Collapse
Affiliation(s)
- Jian-Xian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jun-Peng Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jacopo Desiderio
- Department of Digestive Surgery, St. Mary's Hospital, University of Perugia, Terni, Italy
| | - Jian-Wei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Alessandro Gemini
- Department of Digestive Surgery, St. Mary's Hospital, University of Perugia, Terni, Italy
| | - Jia-bin Wang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Qi-Yue Chen
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Long-long Cao
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Mi Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ruhong Tu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Chao-Hui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Amilcare Parisi
- Department of Digestive Surgery, St. Mary's Hospital, University of Perugia, Terni, Italy
| | - Chang-Ming Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
239
|
Liu K, Jiao XD, Hao JL, Qin BD, Wu Y, Chen W, Liu J, He X, Zang YS. MTSS1 inhibits metastatic potential and induces G2/M phase cell cycle arrest in gastric cancer. Onco Targets Ther 2019; 12:5143-5152. [PMID: 31303767 PMCID: PMC6612291 DOI: 10.2147/ott.s203165] [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: 01/28/2019] [Accepted: 05/10/2019] [Indexed: 02/06/2023] Open
Abstract
Background: Metastasis suppressor 1 (MTSS1), a potential metastasis suppressor gene associated with tumor progression, may play an important role in cancer development. Our previous study demonstrated that MTSS1 was downregulated significantly when gastric cancer (GC) progressed and metastasized, suggesting that MTSS1 may be involved in the physiopathologic mechanism of GC. Purpose: The objective of this study was to evaluate the effect of MTSS1 expression on the biological behavior of gastric cancer cell both in vitro and in vivo. Materials and methods: The gain-and-loss function of MTSS1 in GC cells were analyzed after transfection with pEGFP-N1-MTSS1 and ShRNA431. Proliferation and invasion abilities were measured by means of plate clone formation assay and transwell assay. To further explore the underlying mechanism of MTSS1-induced tumor restrain, cell cycle distribution was analyzed using flow cytometry. Results: The results revealed that overexpression of MTSS1 significantly reduced proliferation, migration and invasion of GC cells in vivo and in vitro, while downregulation of MTSS1 had the opposite biological manifestations. Moreover, overexpression of MTSS1 induced accumulation of GC cells in G2/M phase, increased phosphorylated Cdc2 expression and decreased Cdc25C and cyclinB1 levels, suggesting MTSS1 could cause G2/M cell cycle arrest. Conclusion: Our data provided insight into an important role for MTSS1 in suppressing tumor cell proliferation, invasion and migration, indicating that MTSS, as a functional tumor suppressor in GC, could be a potential therapeutic target to prevent GC metastasis.
Collapse
Affiliation(s)
- Ke Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Xiao-Dong Jiao
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Jie-Lu Hao
- Department of Nephrology, Changzheng Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Bao-Dong Qin
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Ying Wu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Wei Chen
- Department of Pharmacy, Changzheng Hospital, Naval Medical University Shanghai, People's Republic of China
| | - Jun Liu
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Xi He
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, People's Republic of China
| | - Yuan-Sheng Zang
- Department of Medical Oncology, Changzheng Hospital, Naval Medical University, Shanghai, People's Republic of China
| |
Collapse
|
240
|
Zhu L, Chen W, Li G, Chen H, Liao W, Zhang L, Xiao X. Upregulated RACK1 attenuates gastric cancer cell growth and epithelial-mesenchymal transition via suppressing Wnt/β-catenin signaling. Onco Targets Ther 2019; 12:4795-4805. [PMID: 31417279 PMCID: PMC6592218 DOI: 10.2147/ott.s205869] [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: 02/18/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose: As there have been few studies on the effects of the receptor for activated C kinase 1 (RACK1) on gastric cancer (GC), we aimed to explore such effects and the mechanism that may be involved. Patients and methods: Normal gastric epithelial cells and six GC cell lines were used to detect the mRNA expression of RACK1. Overexpressing RACK1 was transfected in HGC27 and MGC803 cells. The effects of overexpressing RACK1 on cell viability, migration, and invasion were determined by cell counting kit-8, wound scratch, and Transwell assay, respectively. The expressions of epithelial–mesenchymal transition (EMT) and Wnt/β-catenin signaling related genes were detected using quantitative real-time PCR or Western blot. Wnt pathway agonist LiCl was added into RACK1 overexpressing GC cells, and then cell viability, migration, and invasion were also detected. Results: RACK1 was downregulated in GC cell lines. Under the circumstance that overexpressing RACK1 was successfully transfected in the two lowest RACK1-expressing GC cells, significant inhibition of cell viability, migration, and invasion, promotion to the mRNA and protein expression of E-cadherin, as well as a decrease in the N-cadherin and Snail expressions could be observed. Overexpressing RACK1 also enhanced the protein level of phosphorylation-β-catenin/β-catenin and attenuated c-Jun protein expression. Additionally, LiCl could partially reverse the inhibitory effects of cell viability, migration and invasion by overexpressing RACK. Conclusion: We found RACK1 possibly inhibited epithelial–mesenchymal transition of GC cells through limitation of the Wnt/β-catenin pathway, thereby suppressing cell migration and invasion; RACK1 could also suppress cell growth.
Collapse
Affiliation(s)
- Lihui Zhu
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Wen Chen
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Guoqing Li
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Honghui Chen
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Wenqiu Liao
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Li Zhang
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Xiaoli Xiao
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| |
Collapse
|
241
|
Wei X, Gao M, Ahmed Y, Gao M, Liu W, Zhang Y, Xie X, Zhao Q, Wang H, Gu K. MicroRNA-362-5p enhances the cisplatin sensitivity of gastric cancer cells by targeting suppressor of zeste 12 protein. Oncol Lett 2019; 18:1607-1616. [PMID: 31423228 PMCID: PMC6607034 DOI: 10.3892/ol.2019.10496] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/10/2019] [Indexed: 12/27/2022] Open
Abstract
Chemotherapy resistance is a major obstacle to the effective treatment of patients with gastric cancer (GC). Mounting evidence has indicated that the dysregulation of microRNAs (miRNAs) is associated with the sensitivity of cancer cells to chemotherapy. However, the mechanisms underlying miRNA-mediated chemoresistance in GC cells remain to be elucidated. The present study aimed to identify functional miRNAs that may regulate the sensitivity of human GC cells to cisplatin (DDP) treatment. miRNA microarray analysis was used to identify differentially expressed miRNAs between the human cisplatin-sensitive GC cell line SGC7901 and the corresponding cisplatin-resistant cell line SGC7901/DDP. miRNA (miR)-362-5p, which is associated with numerous types of tumors, was identified to be downregulated in the SGC7901/DDP cell line. However, the biological role of miR-362-5p in SGC7901/DDP cells remains to be explored. The expression level of miR-362-5p was demonstrated to be reduced in SGC7901/DDP cells compared with SGC7901 cells by reverse transcription-quantitative PCR. Upregulation of miR-362-5p significantly increased cisplatin sensitivity and cisplatin-induced apoptosis, whereas downregulation of miR-362-5p attenuated these effects. Databases predicted that suppressor of zeste 12 protein (SUZ12) may function as a target of miR-362-5p. In addition, the mRNA and protein expression levels of SUZ12 in SGC7901/DDP cells were significantly higher compared with SGC7901 cells and negatively associated with miR-362-5p expression. MTT and western blot analysis assays confirmed that knockdown of SUZ12 enhanced cisplatin sensitivity and decreased NF-κB/p65 protein levels in SGC7901/DDP cells. In addition, upregulation of miR-362-5p in SGC7901/DDP cells decreased the protein expression level of SUZ12, whereas downregulation of miR-362-5p increased the SUZ12 expression level. The results of the present study suggested that dysregulated miR-362-5p may target SUZ12 to promote the development of cisplatin resistance and attenuate cisplatin-induced apoptosis. Therefore, miR-362-5p upregulation combined with cisplatin treatment may serve as a promising therapeutic strategy for patients with cisplatin-resistant GC.
Collapse
Affiliation(s)
- Xiaoli Wei
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Mengru Gao
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yaser Ahmed
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Min Gao
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Wenbo Liu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Yiyin Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Xiaoque Xie
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Qihong Zhao
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Kangsheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
242
|
He Y, Cao X, Kong Y, Wang S, Xia Y, Bi R, Liu J. Apoptosis-promoting and migration-suppressing effect of alantolactone on gastric cancer cell lines BGC-823 and SGC-7901 via regulating p38MAPK and NF-κB pathways. Hum Exp Toxicol 2019; 38:1132-1144. [PMID: 31203647 DOI: 10.1177/0960327119855128] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Gastric cancer is a malignant tumor with high incidence rate and mortality rate. PURPOSE In this study, we investigated the anti-cancer effect of alantolactone, a sesquiterpene lactone, on gastric cancer cell lines BGC-823 and SGC-7901. METHODS BGC-823 and SGC-7901 cells were treated with different concentrations of alantolactone, Hoechst 33258 staining, flow cytometry, wound healing assay, invasion assay, colony forming assay, quantative polymerase chain reaction, and western blot analysis were used to evaluate the anticancer activity of alantolactone to gastric cancer. RESULTS Alantolactone induced apoptosis of gastric cancer cells by regulating the expression of Bax, Bcl-2, and p53, which related to intrinsic apoptotic pathway, and suppressed colony formation, migration, and invasion by mediating the expression of matrix metalloproteinase (MMP)-2, MMP-7, and MMP-9. Cell signaling pathway analysis showed that alantolactone enhanced the phosphorylation of p38 and decreased the translocation of nucleus p65, suggesting that the apoptosis-promoting and migration-suppressing effect of alantolactone might at least partially rely on regulating p38 mitogen-activated protein kinase (p38MAPK) pathway and nuclear factor-κB (NF-κB) pathway. CONCLUSIONS Alantolactone can be used as a potential therapeutic agent for treating gastric cancer.
Collapse
Affiliation(s)
- Y He
- School of Life Science, Liaoning University, Shenyang, China
| | - X Cao
- School of Life Science, Liaoning University, Shenyang, China
| | - Y Kong
- School of Life Science, Liaoning University, Shenyang, China
| | - S Wang
- School of Life Science, Liaoning University, Shenyang, China
| | - Y Xia
- School of Life Science, Liaoning University, Shenyang, China
| | - R Bi
- School of Life Science, Liaoning University, Shenyang, China
| | - J Liu
- School of Life Science, Liaoning University, Shenyang, China
| |
Collapse
|
243
|
Wu L, Hu X, Xu L, Zhang G. Cod Skin Oligopeptide Inhibits Human Gastric Carcinoma Cell Growth by Inducing Apoptosis. Nutr Cancer 2019; 72:218-225. [PMID: 31163997 DOI: 10.1080/01635581.2019.1622740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Aim: To investigate the effect of cod skin oligopeptide (CSO), a natural hydrolysis product obtained from Pacific cod skin gelatin, on the growth of human gastric cancer cells.Methods: Morphological changes were observed in CSO-treated BGC-823 cells under a microscope. The viability of BGC-823 cells was examined by cell counting kit-8 assay. The efficacy of CSO in vivo was evaluated using a xenograft tumor model. Apoptotic cells were identified by flow cytometry. Pro-apoptotic protein expression was verified by western blotting.Results: CSO significantly inhibited BGC-823 cell growth in a dose- and time-dependent manner in vitro and also inhibited the growth of murine gastric cancer xenografts. Flow cytometric assays showed that treatment of BGC-823 cells with CSO significantly increased the percentage of apoptotic cells. Mechanistically, CSO effectively induced expression of pro-apoptotic cleaved caspase-3 and cleaved caspase-9 and down-regulated anti-apoptotic Bcl-2.Conclusion: CSO inhibits gastric cancer cell proliferation while inducing apoptosis by modifying the expression of pro-apoptotic proteins, suggesting a potential therapeutic role for CSO in gastric cancer treatment.
Collapse
Affiliation(s)
- Liping Wu
- Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China
| | - Xiaofei Hu
- Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China
| | - Liyun Xu
- Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China
| | - Guoqiang Zhang
- Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang, China
| |
Collapse
|
244
|
Wang X, Yao Y, Qian H, Li H, Zhu X. Longer Operating Time During Gastrectomy Has Adverse Effects on Short-Term Surgical Outcomes. J Surg Res 2019; 243:151-159. [PMID: 31176285 DOI: 10.1016/j.jss.2019.05.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Revised: 04/16/2019] [Accepted: 05/08/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Gastric cancer continues to be one of the malignant tumor types with high morbidity and mortality worldwide. Although remarkable improvements have been made to combat gastric cancer, surgery is still the first choice of treatment for gastric cancer. METHODS This was a single-center and retrospective study. A total of 110 patients who underwent radical gastrectomy with D2 lymph node dissection between 2014 and 2017 were included in this study, and all patients were treated by the same medical staff. Based on the median operating time, patients were grouped into a long-time group (>180 min) and a short-time group (≤180 min). Influences of operating time on outcomes of patients in the short-term and long-term groups were analyzed. RESULTS The long-time group showed a higher incidence of postoperative complications compared with the short time group (P < 0.01) with a significant decrease in serum albumin and the prognostic nutritional index value. Moreover, a long operating time was often caused by the operating start time (P < 0.001), excision difficulty caused by lager tumor size (P < 0.001), worse tumor differentiation, and deeper tumor invasion (P < 0.05). However, length of operating time did not significantly influence overall survival of patients who underwent radical gastrectomy. CONCLUSIONS The results suggested that operating time might be an indicator of the incidence of postoperative complication and that several important variables, such as prognostic nutritional index, serum albumin, operating start time, and excision time, could be intervened in the perioperative period to help patients gain a better outcome after gastrectomy.
Collapse
Affiliation(s)
- Xuchao Wang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huan Qian
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hao Li
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinguo Zhu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
245
|
Halicek M, Fabelo H, Ortega S, Callico GM, Fei B. In-Vivo and Ex-Vivo Tissue Analysis through Hyperspectral Imaging Techniques: Revealing the Invisible Features of Cancer. Cancers (Basel) 2019; 11:E756. [PMID: 31151223 PMCID: PMC6627361 DOI: 10.3390/cancers11060756] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/20/2019] [Accepted: 05/24/2019] [Indexed: 12/27/2022] Open
Abstract
In contrast to conventional optical imaging modalities, hyperspectral imaging (HSI) is able to capture much more information from a certain scene, both within and beyond the visual spectral range (from 400 to 700 nm). This imaging modality is based on the principle that each material provides different responses to light reflection, absorption, and scattering across the electromagnetic spectrum. Due to these properties, it is possible to differentiate and identify the different materials/substances presented in a certain scene by their spectral signature. Over the last two decades, HSI has demonstrated potential to become a powerful tool to study and identify several diseases in the medical field, being a non-contact, non-ionizing, and a label-free imaging modality. In this review, the use of HSI as an imaging tool for the analysis and detection of cancer is presented. The basic concepts related to this technology are detailed. The most relevant, state-of-the-art studies that can be found in the literature using HSI for cancer analysis are presented and summarized, both in-vivo and ex-vivo. Lastly, we discuss the current limitations of this technology in the field of cancer detection, together with some insights into possible future steps in the improvement of this technology.
Collapse
Affiliation(s)
- Martin Halicek
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA.
- Department of Biomedical Engineering, Emory University and The Georgia Institute of Technology, 1841 Clifton Road NE, Atlanta, GA 30329, USA.
| | - Himar Fabelo
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA.
- Institute for Applied Microelectronics (IUMA), University of Las Palmas de Gran Canaria (ULPGC), 35017 Las Palmas de Gran Canaria, Spain.
| | - Samuel Ortega
- Institute for Applied Microelectronics (IUMA), University of Las Palmas de Gran Canaria (ULPGC), 35017 Las Palmas de Gran Canaria, Spain.
| | - Gustavo M Callico
- Institute for Applied Microelectronics (IUMA), University of Las Palmas de Gran Canaria (ULPGC), 35017 Las Palmas de Gran Canaria, Spain.
| | - Baowei Fei
- Department of Bioengineering, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA.
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hine Blvd, Dallas, TX 75390, USA.
- Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hine Blvd, Dallas, TX 75390, USA.
| |
Collapse
|
246
|
杨 万, 潘 云, 管 沛, 李 雪, 尤 崇. [Bioinformatics analysis of COL1A1 regulated by miR-129-5p as a potential therapeutic target for gastric cancer]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:540-546. [PMID: 31140417 PMCID: PMC6743932 DOI: 10.12122/j.issn.1673-4254.2019.05.07] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To explore the pathogenesis of gastric cancer through a bioinformatic approach to provide evidence for the prevention and treatment of gastric cancer. METHODS The differentially expressed genes (DEGs) in gastric cancer and normal gastric mucosa in GSE79973 dataset were analyzed using GEO2R online tool. GO analysis and KEGG pathway enrichment analysis of the DEGs in DAVID database were performed. The protein interaction network was constructed using STRING database, and the key genes (Hub genes) were screened and their functional modules were analyzed using Cytoscape software. The GEPIA database was used to validate the Hub genes, and the Target Scan database was used to predict the microRNAs that regulate the target genes; OncomiR was used to analyze the expressions of the microRNAs in gastric cancer tissues and their relationship with the survival outcomes of the patients. RESULTS A total of 181 DEGs were identified in gastric cancer, and 10 hub genes were screened by the protein- protein interaction network. Functional analysis showed that these DEGs were involved mainly in protein digestion and absorption, PI3K-Akt signaling pathway, ECM-receptor interaction and platelet activation signal pathway. GEPIA database validation showed that COL1A1 was highly expressed in gastric cancer tissues and was associated with a poor prognosis of patients with gastric cancer. MiR-129-5p was found to bind to the 3'UTR of COL1A1 mRNA, and compared with that in normal tissues, miR-129-5p expression was obviously down-regulated in gastric cancer tissues, and was correlated with the prognosis of the patients. CONCLUSIONS COL1A1 under regulation by MiR-129-5p is a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- 万霞 杨
- />兰州大学第二医院检验医学中心,甘肃 兰州 730030Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - 云燕 潘
- />兰州大学第二医院检验医学中心,甘肃 兰州 730030Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - 沛文 管
- />兰州大学第二医院检验医学中心,甘肃 兰州 730030Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - 雪 李
- />兰州大学第二医院检验医学中心,甘肃 兰州 730030Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - 崇革 尤
- />兰州大学第二医院检验医学中心,甘肃 兰州 730030Laboratory Medicine Center, Lanzhou University Second Hospital, Lanzhou 730030, China
| |
Collapse
|
247
|
Zhang Z, Dong Y, Hua J, Xue H, Hu J, Jiang T, Shi L, Du J. A five-miRNA signature predicts survival in gastric cancer using bioinformatics analysis. Gene 2019; 699:125-134. [DOI: 10.1016/j.gene.2019.02.058] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 01/14/2019] [Accepted: 02/21/2019] [Indexed: 12/11/2022]
|
248
|
Tung SY, Lin CT, Chen CN, Huang WS. Effect of mitomycin C on X-ray repair cross complementing group 1 expression and consequent cytotoxicity regulation in human gastric cancer cells. J Cell Biochem 2019; 120:8333-8342. [PMID: 30614038 DOI: 10.1002/jcb.28116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 10/31/2018] [Indexed: 02/07/2023]
Abstract
Gastric cancer is the fourth most common cancer and ranks as the second leading cause of cancer-related deaths across the world. The combination therapy of surgery with chemotherapeutic drugs, that is, mitomycin C (MMC), is becoming a major strategy for patients with advanced gastric cancer. However, drug resistance is a major factor that limits the effectiveness of chemotherapy, which ultimately leads to the failure of cancer chemotherapy. X-ray repair cross complementing group 1 (XRCC1), a scaffold protein of the base excision repair process, has been implicated in the development of tumor chemoresistance. Thus, this study aimed to explore whether XRCC1 expression could be regulated, its role in gastric AGS cancer cells treated with MMC, and the underlying mechanism. The results of this study demonstrate that XRCC1 expression could be upregulated in AGS cells treated with MMC, and this upregulation could subsequently reduce the cytotoxicity of MMC in AGS cells. Furthermore, MMC-upregulated XRCC1 expression was regulated by MAPK signaling through activating the transcription factor Sp1. These results indicate the role of XRCC1 in the development of drug resistance to MMC in gastric AGS cells. Elucidating the mechanism concerning the MAPKs and transcription factor Sp1 may provide another notion for the development of a clinical chemotherapy strategy for gastric cancers in the future.
Collapse
Affiliation(s)
- Shui-Yi Tung
- Department of Hepato-Gastroenterology, Chang Gung Memorial Hospital Chiayi Branch, Chiayi, Taiwan, ROC.,Chang Gung University College of Medicine, Taoyuan, Taiwan, ROC
| | - Chien-Tsong Lin
- Center for General Education, National Formosa University, Yunlin, Taiwan, ROC.,Department of Wood Based Materials and Design, National Chiayi University, Chiayi, Taiwan, ROC
| | - Cheng-Nan Chen
- Department of Biochemical Science and Technology, National Chiayi University, Chiayi, Taiwan, ROC
| | - Wen-Shih Huang
- Division of Colon and Rectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Chiayi, Taiwan, ROC
| |
Collapse
|
249
|
Kahroba H, Hejazi MS, Samadi N. Exosomes: from carcinogenesis and metastasis to diagnosis and treatment of gastric cancer. Cell Mol Life Sci 2019; 76:1747-1758. [PMID: 30734835 PMCID: PMC11105779 DOI: 10.1007/s00018-019-03035-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 01/24/2019] [Accepted: 01/29/2019] [Indexed: 02/07/2023]
Abstract
Exosomes represent an important group of extracellular vesicles with a defined size between 40 and 150 nm and cup-shaped construction which have a pivotal role in elimination of intracellular debris and intercellular signaling networks. A line of evidence revealed the impact of different types of exosomes in initiation, progression, and metastasis of gastric cancer (GC). These bioactive vesicles mediate tumor and stromal communication network through modulation of cell signaling for carcinogenesis and pre-metastatic niche formation in distant organs. Exosomes contain various cargos including DNAs (mitochondrial and genomic), proteins, transposable elements, and RNAs (coding and noncoding) with different compositions related to functional status of origin cells. In this review, we summarize the main roles of key exosomal cargos in induction of exosome-mediated signaling in cancer cells. Body fluids are employed frequently as the source of exosomes released by tumor cells with a potential role in early diagnosis of GC and chemoresistance. These vesicles as non-toxic and non-immunogenic carriers are also found to be applied for novel drug delivery systems.
Collapse
Affiliation(s)
- Houman Kahroba
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Saeid Hejazi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
250
|
Diao N, Li Y, Yang J, Jin C, Meng X, Jiao W, Feng J, Liu Z, Lu N. High expression of HMBOX1 contributes to poor prognosis of gastric cancer by promoting cell proliferation and migration. Biomed Pharmacother 2019; 115:108867. [PMID: 31005794 DOI: 10.1016/j.biopha.2019.108867] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 03/24/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
Homeobox-containing 1 (HMBOX1) has been reported to be associated with biological characteristics of some tumors, but its roles in gastric cancer have never been reported. In the present study, we found that HMBOX1 expression was significantly upregulated in gastric cancer tissues and cell lines and correlated with the TNM stage, lymph-node metastatic and the overall survival (OS) of patients of gastric cancer. The overexpression of HMBOX1 in gastric cancer cells enhanced cell proliferation by accelerating cell cycle, induced cell migration. In contrast, silencing HMBOX1 inhibited these processes. And the expression of HMBOX1 was related with the expression of vascular endothelial growth factor receptor (VEGFR), transforming growth factor-β (TGF-β) and CD133. What's more, we found that the expression of CD133 had a significantly positive correlation with HMBOX1 in gastric cancer tissues, and the co-expression of HMBOX1 and CD133 was significantly correlated with poor prognosis of gastric cancer patients, especially for patients at III and IV stage. In conclusion, HMBOX1 was upregulated in gastric cancer and correlated with gastric cancer cell proliferation and migration. Moreover, HMBOX1 combined CD133 might be useful to predict survival of patients with advanced gastric cancer.
Collapse
Affiliation(s)
- Nannan Diao
- Institute of Diagnostics, School of Medicine, Shandong University, Ji'nan, Shandong, China; Department of Clinical Laboratory, Shanghai Skin Disease Hospital, Shanghai, China
| | - Yuzheng Li
- Institute of Yantai, China Agricultural University, Beijing, China
| | - Jinling Yang
- Institute of Diagnostics, School of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Chengjuan Jin
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China; Key Laboratory of Gynecologic Oncology of Shandong Province, Qilu Hospital, Shandong University, Ji'nan, Shandong, China
| | - Xiaohui Meng
- Institute of Diagnostics, School of Medicine, Shandong University, Ji'nan, Shandong, China
| | - Wenlin Jiao
- National Research Center for Assisted Reproductive Technology and Reproduction Genetics, Ji'nan, Shandong, China
| | - Jinbo Feng
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China; Key Laboratory of Gynecologic Oncology of Shandong Province, Qilu Hospital, Shandong University, Ji'nan, Shandong, China
| | - Zhenping Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China; Key Laboratory of Gynecologic Oncology of Shandong Province, Qilu Hospital, Shandong University, Ji'nan, Shandong, China
| | - Nan Lu
- Institute of Diagnostics, School of Medicine, Shandong University, Ji'nan, Shandong, China.
| |
Collapse
|