101
|
Hill DG, Yu L, Gao H, Balic JJ, West A, Oshima H, McLeod L, Oshima M, Gallimore A, D'Costa K, Bhathal PS, Sievert W, Ferrero RL, Jenkins BJ, Jones GW. Hyperactive gp130/STAT3-driven gastric tumourigenesis promotes submucosal tertiary lymphoid structure development. Int J Cancer 2018; 143:167-178. [PMID: 29417587 PMCID: PMC5969244 DOI: 10.1002/ijc.31298] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/23/2018] [Accepted: 01/30/2018] [Indexed: 01/27/2023]
Abstract
Tertiary lymphoid structures (TLSs) display phenotypic and functional characteristics of secondary lymphoid organs, and often develop in tissues affected by chronic inflammation, as well as in certain inflammation-associated cancers where they are prognostic of improved patient survival. However, the mechanisms that govern the development of tumour-associated TLSs remain ill-defined. Here, we observed tumour-associated TLSs in a preclinical mouse model (gp130F/F ) of gastric cancer, where tumourigenesis is dependent on hyperactive STAT3 signalling through the common IL-6 family signalling receptor, gp130. Gastric tumourigenesis was associated with the development of B and T cell-rich submucosal lymphoid aggregates, containing CD21+ cellular networks and high endothelial venules. Temporally, TLS formation coincided with the development of gastric adenomas and induction of homeostatic chemokines including Cxcl13, Ccl19 and Ccl21. Reflecting the requirement of gp130-driven STAT3 signalling for gastric tumourigenesis, submucosal TLS development was also STAT3-dependent, but independent of the cytokine IL-17 which has been linked with lymphoid neogenesis in chronic inflammation and autoimmunity. Interestingly, upregulated lymphoid chemokine expression and TLS formation were also observed in a chronic gastritis model induced by Helicobacter felis infection. Tumour-associated TLSs were also observed in patients with intestinal-type gastric cancer, and a gene signature linked with TLS development in gp130F/F mice was associated with advanced clinical disease, but was not prognostic of patient survival. Collectively, our in vivo data reveal that hyperactive gp130-STAT3 signalling closely links gastric tumourigenesis with lymphoid neogenesis, and while a TLS gene signature was associated with advanced gastric cancer in patients, it did not indicate a favourable prognosis.
Collapse
Affiliation(s)
- David G. Hill
- Division of Infection and ImmunitySystems Immunity Research Institute, School of Medicine, Cardiff UniversityCardiffWalesUnited Kingdom
| | - Liang Yu
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health SciencesMonash UniversityClaytonVICAustralia
| | - Hugh Gao
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health SciencesMonash UniversityClaytonVICAustralia
| | - Jesse J. Balic
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health SciencesMonash UniversityClaytonVICAustralia
| | - Alison West
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health SciencesMonash UniversityClaytonVICAustralia
| | - Hiroko Oshima
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
| | - Louise McLeod
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
| | - Masanobu Oshima
- Division of GeneticsCancer Research Institute, Kanazawa UniversityKanazawaJapan
| | - Awen Gallimore
- Division of Infection and ImmunitySystems Immunity Research Institute, School of Medicine, Cardiff UniversityCardiffWalesUnited Kingdom
| | - Kimberley D'Costa
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health SciencesMonash UniversityClaytonVICAustralia
| | - Prithi S. Bhathal
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health SciencesMonash UniversityClaytonVICAustralia
| | - William Sievert
- Department of MedicineMonash Medical Centre, Monash UniversityClaytonVICAustralia
| | - Richard L. Ferrero
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Biomedicine Discovery Institute, Department of MicrobiologyMonash UniversityClaytonVICAustralia
| | - Brendan J. Jenkins
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health SciencesMonash UniversityClaytonVICAustralia
| | - Gareth W. Jones
- Division of Infection and ImmunitySystems Immunity Research Institute, School of Medicine, Cardiff UniversityCardiffWalesUnited Kingdom
| |
Collapse
|
102
|
Optimizing miRNA-module diagnostic biomarkers of gastric carcinoma via integrated network analysis. PLoS One 2018; 13:e0198445. [PMID: 29879180 PMCID: PMC5991748 DOI: 10.1371/journal.pone.0198445] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 05/18/2018] [Indexed: 12/17/2022] Open
Abstract
Several microRNAs (miRNAs) have been suggested as novel biomarkers for diagnosing gastric cancer (GC) at an early stage, but the single-marker strategy may ignore the co-regulatory relationships and lead to low diagnostic specificity. Thus, multi-target modular diagnostic biomarkers are urgently needed. In this study, a Zsummary and NetSVM-based method was used to identify GC-related hub miRNAs and activated modules from clinical miRNA co-expression networks. The NetSVM-based sub-network consisting of the top 20 hub miRNAs reached a high sensitivity and specificity of 0.94 and 0.82. The Zsummary algorithm identified an activated module (miR-486, miR-451, miR-185, and miR-600) which might serve as diagnostic biomarker of GC. Three members of this module were previously suggested as biomarkers of GC and its 24 target genes were significantly enriched in pathways directly related to cancer. The weighted diagnostic ROC AUC of this module was 0.838, and an optimized module unit (miR-451 and miR-185) obtained a higher value of 0.904, both of which were higher than that of individual miRNAs. These hub miRNAs and module have the potential to become robust biomarkers for early diagnosis of GC with further validations. Moreover, such modular analysis may offer valuable insights into multi-target approaches to cancer diagnosis and treatment.
Collapse
|
103
|
Multiplex serology of Helicobacter pylori antigens in detection of current infection and atrophic gastritis - A simple and cost-efficient method. Microb Pathog 2018; 119:137-144. [DOI: 10.1016/j.micpath.2018.04.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/10/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023]
|
104
|
Wang Y, Zhang H, Ge S, Fan Q, Zhou L, Li H, Bai M, Ning T, Liu R, Wang X, Deng T, Zhang L, Ying G, Ba Y. Effects of miR‑138‑5p and miR‑204‑5p on the migration and proliferation of gastric cancer cells by targeting EGFR. Oncol Rep 2018; 39:2624-2634. [PMID: 29693184 PMCID: PMC5983934 DOI: 10.3892/or.2018.6389] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 03/29/2018] [Indexed: 12/17/2022] Open
Abstract
GC (gastric cancer) remains one of the most lethal malignancies worldwide. EGFR (epidermal growth factor receptor) plays an important role in the malignant process of GC, therefore, the present study addressed the relationship between EGFR and its potential regulators and examined their regulatory mechanisms in GC. We examined differences in the expression levels of EGFR in GC and adjacent non‑cancerous tissues. Bioinformatics analyses and dual luciferase reporter assays were used to confirm the putative relationship between miR‑138 or miR‑204 and EGFR, and their relationship was further detected using western blotting, RT‑PCR, and a series of cell studies. EGFR proteins were abundantly expressed in GC tissues, however EGFR mRNA levels remained indistinctive. Consequently, EGFR was revealed as a putative target of miR‑138 and miR‑204 which bound to the 3'UTR of EGFR mRNA. Further analysis revealed that miR‑138 and miR‑204 were significantly downregulated in GC tissues and the overexpression of miR‑138 and miR‑204 in GC cell lines resulted in the significant inhibition of EGFR protein levels and GC cell proliferation and metastasis. Rescue experiments confirmed that the roles of the two microRNAs were specific to EGFR. EGFR is a pivotal oncogene in GC progression that may be regulated by miR‑138 and miR‑204.
Collapse
Affiliation(s)
- Yi Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Haiyang Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Shaohua Ge
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Qian Fan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Likun Zhou
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Hongli Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Ming Bai
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Tao Ning
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Rui Liu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Xia Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Ting Deng
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Le Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Guoguang Ying
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| | - Yi Ba
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China
| |
Collapse
|
105
|
Wang L, Li Y, Li L, Wu Z, Wu Y, Ma H, Yu H, Yang D, Wang D. Role of Kruppel-like factor 4 in regulating inhibitor of apoptosis-stimulating protein of p53 in the progression of gastric cancer. Oncol Lett 2018; 15:6865-6872. [PMID: 29849786 PMCID: PMC5962871 DOI: 10.3892/ol.2018.8203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 12/08/2017] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer (GC) remains one of the leading causes of cancer-associated mortality. The overexpression of inhibitor of apoptosis-stimulating protein of p53 (iASPP) has been detected in GC tissues but the function of iASPP in the viability of GC cells and its underlying molecular mechanism remains unknown. Kruppel-like factor 4 (KLF4) is a tumor suppressor gene in GC and it may interact with p53. iASPP is an evolutionarily conserved inhibitor of p53, whereas KLF4 may be negatively associated with iASPP in GC. However, whether KLF4 has regulatory effects on iASPP remains to be investigated. The objective of the present study was to examine the function of iASPP and KLF4 in the proliferation of GC cells and to determine whether KLF4 has regulatory effects on iASPP. It was demonstrated that iASPP was upregulated and KLF4 was downregulated in GC cell lines. Downregulation of iASPP inhibited the proliferation and colony formation ability, and promoted the apoptosis of GC cells. Additionally, upregulation of KLF4 inhibited the proliferation and colony formation ability, and promoted apoptosis of GC cells. Furthermore, upregulation of KLF4 inhibited the expression of iASPP. Upregulation of iASPP following overexpression of KLF4 reversed the KLF4-mediated effects in GC cells. In vivo upregulation of KLF4 or downregulation of iASPP inhibited the growth of tumors, whereas upregulation of iASPP promoted the growth of tumors. In conclusion, iASPP may act as an oncogene that promotes the proliferation of GC cells. The results demonstrated that KLF4 was a negative regulatory factor of iASPP and that overexpression of iASPP inhibited the effects of KLF4. Thus, downregulation of KLF4 in GC may lead to overexpression of iASPP and promote the development of cancer.
Collapse
Affiliation(s)
- Lulu Wang
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Yan Li
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Luchun Li
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Zhijuan Wu
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Yongzhong Wu
- Department of Radiotherapy, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Huiwen Ma
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Huiqing Yu
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Dan Yang
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| | - Donglin Wang
- Department of Oncology, Chongqing Cancer Institute, Chongqing 400030, P.R. China
| |
Collapse
|
106
|
O’Neill L, Moran J, Guinan EM, Reynolds JV, Hussey J. Physical decline and its implications in the management of oesophageal and gastric cancer: a systematic review. J Cancer Surviv 2018; 12:601-618. [DOI: 10.1007/s11764-018-0696-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 05/08/2018] [Indexed: 12/14/2022]
|
107
|
Xu T, Kong Z, Zhao H. Relationship Between Tumor Necrosis Factor-α rs361525 Polymorphism and Gastric Cancer Risk: A Meta-Analysis. Front Physiol 2018; 9:469. [PMID: 29867530 PMCID: PMC5962813 DOI: 10.3389/fphys.2018.00469] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/13/2018] [Indexed: 12/17/2022] Open
Abstract
Tumor necrosis factor (TNF)-α, a major part in inflammatory, infectious and tumor processes, and is pivotal at the early stages of gastric cancer. Relationship between its risk and TNF-α rs361525 polymorphism has been demonstrated, but remains conflicting and controversial. Therefore, a meta-analysis was conducted to more precisely estimate this relationship. PubMed, Web of Science, EMBASE and CNKI were comprehensively searched to find out relevant articles through October 5, 2017. The strength of the relationship was assessed using pooled odds ratios and 95% confidence intervals. Totally 20 articles were included involving 4,084 cases and 7,010 controls. No significant relationship between TNF-α rs361525 polymorphism and increased GC risk was found in the whole populations. Subgroup analyses uncovered TNF-α rs361525 polymorphism intensified the risk of GC among Asians under five models, but decreased the risk of GC among Caucasiansin the allelic and dominant models. Subgroup analysis by genotyping methods revealed increased risk for other methods. In conclusion, this meta-analysis suggests TNF-α rs361525 polymorphism is related to the risk of GC, especially for Asians.
Collapse
Affiliation(s)
- Tianshu Xu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhijun Kong
- Department of General Surgery, Affiliated Hospital of Nanjing Medical University, Changzhou Second People's Hospital, Changzhou, China
| | - Hui Zhao
- Department of General Surgery, Wuxi Third People's Hospital, Wuxi, China
| |
Collapse
|
108
|
Zhu Y, Zhang Y, Sui Z, Zhang Y, Liu M, Tang H. USP14 de-ubiquitinates vimentin and miR-320a modulates USP14 and vimentin to contribute to malignancy in gastric cancer cells. Oncotarget 2018; 8:48725-48736. [PMID: 27448976 PMCID: PMC5564720 DOI: 10.18632/oncotarget.10706] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 06/01/2016] [Indexed: 02/07/2023] Open
Abstract
Vimentin plays important roles in the epithelial-to-mesenchymal transition (EMT). In this study, we found that vimentin was highly expressed in human gastric cancer (GC) tissues and cell lines and significantly promoted cell growth, migration and invasion. Ubiquitin-specific protease 14 (USP14) interacted with the vimentin protein, which led to its de-ubiquitination. miR-320a was found to bind to the 3′UTR of both vimentin and USP14 transcripts and downregulate the expression of both proteins. The downregulation of miR-320a upregulates vimentin expression by directly binding to the 3′UTR of vimentin to derepress expression and indirectly by augmenting USP14 to increase vimentin stability in GC cells. Taken together, these results provide new insight into malignancy in gastric cancers.
Collapse
Affiliation(s)
- Ying Zhu
- Tianjin Life Science Research Center, Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yan Zhang
- Tianjin Life Science Research Center, Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Zhenhua Sui
- Tianjin Life Science Research Center, Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yi Zhang
- Tianjin Life Science Research Center, Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Min Liu
- Tianjin Life Science Research Center, Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Hua Tang
- Tianjin Life Science Research Center, Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
109
|
Liu Z, Yan Y, Cao S, Chen Y. Long non-coding RNA SNHG14 contributes to gastric cancer development through targeting miR-145/SOX9 axis. J Cell Biochem 2018; 119:6905-6913. [PMID: 29667771 DOI: 10.1002/jcb.26889] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 03/21/2018] [Indexed: 12/11/2022]
Abstract
This study aimed to elucidate the roles of long non-coding RNA SNHG14 in gastric cancer development. LncRNA SNHG14 was markedly up-regulated in gastric cancer tissues and cells. Knockdown of SNHG14 significantly inhibited SGC-7901 cell viability, migration, invasion, and promoted cell apoptosis. In addition, miR-145 was negatively regulated by SNHG14 and the effects of SNHG14 knockdown on cell viability, apoptosis, migration, invasion, and the expression of apoptosis-related proteins and EMT-markers were reversed by inhibition of miR-145 at the same time. Furthermore, SOX9 was verified as a functional target of miR-145, and miR-145 regulated tumor malignant behaviors through regulating SOX9. Besides, knockdown of SNHG14 inhibited the expression of p-PI3 K, p-AKT, and p-mTOR and promoted PTEN expression, where miR-145 inhibition had opposite effects. Moreover, the activated PI3 K/AKT/mTOR pathway caused by miR-145 inhibition was counteracted after knockdown of SOX9. Our findings indicate that up-regulation of lncRNA SNHG14 may contribute to gastric cancer development via targeting miR-145/SOX9 axis and involving in PI3 K/AKT/mTOR pathway. SNHG14-miR-145/SOX9 axis may be a promising therapeutic strategy for gastric cancer treatment.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Surgery, Xi'an Chest Hospital, Xi'an TB&Thoracic Tumor Hospital, Xi'an, Shanxi, China
| | - Yan Yan
- Institute of Cancer Research, School of Basic Medical Science of Xi'an Jiao Tong University, Xi'an, Shaanxi, China.,Department of Pathology, The First Hospital of Xi'an, Xi'an, Shaanxi, China
| | - Sizhe Cao
- Department of Medicine, Xi'an Chest Hospital, Xi'an TB&Thoracic Tumor Hospital, Xi'an, Shanxi, China
| | - Yi Chen
- Department of General Surgery, The Changan District Hospital of the First Affiliated Hospital, Xi'an Jiaotong University, Shanxi, China
| |
Collapse
|
110
|
Peng XC, Zeng Z, Huang YN, Deng YC, Fu GH. Clinical significance of TM4SF1 as a tumor suppressor gene in gastric cancer. Cancer Med 2018; 7:2592-2600. [PMID: 29665316 PMCID: PMC6010756 DOI: 10.1002/cam4.1494] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/13/2018] [Accepted: 03/20/2018] [Indexed: 12/14/2022] Open
Abstract
Transmembrane‐4‐L‐six‐family member‐1 (TM4SF1), a tumor‐associated antigen, is overexpressed in most epithelial cell carcinomas and a potential target for antibody‐mediated therapy. However, the role of TM4SF1 in gastric cancer has not been elucidated. The aim of this study was to investigate the clinical significance of TM4SF1 expression in gastric carcinoma (GC) tissues using 152 GC tissue samples and matched adjacent nontumor tissue samples analyzed by immunohistochemistry, and 13 fresh GC tissue samples analyzed by Western blotting. The results showed that TM4SF1 was heterogeneously expressed in normal gastric mucosa, with a high expression rate in fundus mucosa. Higher levels and strong expression rate of TM4SF1 were associated with GC tissues of higher‐grade differentiation. TM4SF1 levels were lower in gastric cancer tissues than gastric noncancerous tissues. Expression of TM4SF1 was not correlated with USP10 (P = 0.157), S100A12 (P = 0.479), p53 (P = 0.249), or Ki67 (P = 0.166) in GC. The expression of TM4SF1 was significantly and negatively correlated with depth of invasion (P = 0.031), nodal metastasis (P = 0.042), TNM stage (P = 0.030), and Lauren classification (P = 0.026). There was no significant correlation between TM4SF1 expression and age, gender, tumor size, or distant metastasis (P > 0.05). The expression of TM4SF1 was associated with well overall survival (P = 0.0164). The 5‐year survival rate for patients with GC showing TM4SF1 positive was 58.82% (10/17), and the median survival time was 78 months, higher than that (12.90%, 12/93) of patients who were TM4SF1 negative, whose median survival time was 62 months. These data suggested that low expression of TM4SF1 is associated with carcinogenesis and development, tumor progression and invasion of gastric cancer, and poor overall survival of patients with GC. TM4SF1 is a tumor suppressor for GC and a novel prognostic marker for patients with GC.
Collapse
Affiliation(s)
- Xing-Chun Peng
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Number 280, South Chong-Qing Road, Shanghai, 200025, China.,School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei Province, China
| | - Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, No.99, Ziyang Road, Wuchang District, Wuhan, 430060, Hubei Province, China
| | - Yu-Ning Huang
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Number 280, South Chong-Qing Road, Shanghai, 200025, China
| | - Yun-Chao Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No.99, Ziyang Road, Wuchang District, Wuhan, 430060, Hubei Province, China
| | - Guo-Hui Fu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Number 280, South Chong-Qing Road, Shanghai, 200025, China
| |
Collapse
|
111
|
Arun K, Arunkumar G, Bennet D, Chandramohan SM, Murugan AK, Munirajan AK. Comprehensive analysis of aberrantly expressed lncRNAs and construction of ceRNA network in gastric cancer. Oncotarget 2018; 9:18386-18399. [PMID: 29719612 PMCID: PMC5915079 DOI: 10.18632/oncotarget.24841] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 02/28/2018] [Indexed: 12/19/2022] Open
Abstract
Gastric cancer remains fifth most common cancer often diagnosed at an advanced stage and is the second leading cause of cancer-related death worldwide. Long non-coding RNAs (lncRNAs) involved in various cellular pathways are essential for tumor occurrence and progression and they have high potential to promote or suppress the expression of many genes. In this study, we profiled 19 selected cancer-associated lncRNAs in thirty gastric adenocarcinomas and matching normal tissues by qRT-PCR. Our results showed that most of the lncRNAs were significantly upregulated (12/19). Further, we performed bioinformatic screening of miRNAs that share common miRNA response elements (MREs) with lncRNAs and their downstream mRNA targets. The prediction identified three microRNAs (miR-21, miR-145 and miR-148a) and five gastric cancer-specific target genes (EGFR, KLF4, DNMT1 and AGO4) which also showed strong correlation with lncRNAs in regression analysis. Finally, we constructed an integrated lncRNA-miRNA-mRNA interaction network of the candidate genes to understand the post-transcriptional gene regulation. The ceRNA network analysis revealed that the differentially regulated miR-21 and miR-148a were playing as central candidates coordinating sponging activity of the lncRNAs analyzed (H19, TUG1 and MALAT1) in this study and the overexpression of H19 and miR-21 could be a signature event of gastric tumorigenesis that could serve as prognostic indicators and therapeutic targets.
Collapse
Affiliation(s)
- Kanagaraj Arun
- Department of Genetics, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai - 600 113, India
| | - Ganesan Arunkumar
- Department of Genetics, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai - 600 113, India
| | - Duraisamy Bennet
- Institute of Surgical Gastroenterology, Rajiv Gandhi Government General Hospital and Madras Medical College, Chennai - 600 001, India
| | - Servarayan Murugesan Chandramohan
- Institute of Surgical Gastroenterology, Rajiv Gandhi Government General Hospital and Madras Medical College, Chennai - 600 001, India
| | - Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh-11211, Saudi Arabia
| | - Arasambattu Kannan Munirajan
- Department of Genetics, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai - 600 113, India
| |
Collapse
|
112
|
The molecular mechanism of anticancer action of novel octahydropyrazino[2,1-a:5,4-a']diisoquinoline derivatives in human gastric cancer cells. Invest New Drugs 2018; 36:970-984. [PMID: 29549610 PMCID: PMC6244973 DOI: 10.1007/s10637-018-0584-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 03/01/2018] [Indexed: 02/08/2023]
Abstract
Objective The aim of the current study was to examine the anticancer activity and the detailed mechanism of novel diisoquinoline derivatives in human gastric cancer cells (AGS). Methods The viability of AGS cells was measured by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay. Cell cycle analysis and apoptosis assay were performed by standard flow cytometric method. Confocal microscopy bioimaging was used to demonstrate the expression of pivotal proteins engaged in apoptosis (caspase-8, caspase-3, p53) and cell signaling (AKT, ERK1/2). Results All compounds decreased the number of viable cells in a dose-dependent manner after 24 and 48 h of incubation, although compound 2 was a more cytotoxic agent, with IC50 values of 21 ± 2 and 6 ± 2 μM, compared to 80 ± 2 and 45 ± 2 μM for etoposide. The cytotoxic and antiproliferative effects of novel compounds were associated with the induction of apoptosis. The highest percentage of early and late apoptotic cells was observed after 48 h of incubation with compound 2 (89.9%). The value was higher compared to compound 1 (20.4%) and etoposide (24.1%). The novel diisoquinoline derivatives decreased the expression of AKT and ERK1/2. Their mechanism was associated with p53-mediated apoptosis, accumulation of cells in the G2/M phase of cell cycle and inhibition of topoisomerase II. Conclusion These data strongly support compound 2 as a promising molecule for treatment of gastric cancer.
Collapse
|
113
|
Soares CD, Rocha BA, Paranaiba LMR, de Melo-Filho MR, Jorge J, de Carvalho MGF, de Almeida OP. A challenging diagnosis: Case report of oral metastasis from gastric adenocarcinoma mimicking pyogenic granuloma. Medicine (Baltimore) 2018; 97:e9934. [PMID: 29595701 PMCID: PMC5895424 DOI: 10.1097/md.0000000000009934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
RATIONALE Oral metastases occur more commonly in bone, but can also manifest in soft tissues and eventually resemble a reactive lesion. Few cases of oral metastases mimicking reactive lesions in soft tissues have been reported to date. PATIENT CONCERNS We report a metastasis of gastric carcinoma (GC) to the oral mucosa without bone involvement in a 43 yom clinically and microscopically mimicking a reactive lesion. The patient related that the lesion had 1 month of evolution, and the ulcerated area suggested the lesion was related to trauma. DIAGNOSES The histopathological examination of the lesion revealed an exuberant granulation tissue with few neoplastic cells, and the initial diagnosis of pyogenic granuloma was considered. In a second analysis, clusters of clear cells morphologically similar to degenerating mucous cells or macrophages, positive for Cytokeratin (CK)-20, and CDX2 were found. At the moment, it was confirmed the presence of a primary GC in the patient. INTERVENTIONS A palliative radiotherapy/chemotherapy was started. OUTCOMES However, the patient died 3 months after the diagnosis of oral metastasis. LESSONS This report highlights the importance of careful clinical and microscopic examinations in cases of oral metastasis that may mimic a reactive lesion.
Collapse
Affiliation(s)
| | - Breno Amaral Rocha
- Oncologic Dentistry Service, Santa Casa Hospital, Montes Claros, Minas Gerais
| | - Lívia Máris Ribeiro Paranaiba
- Dental School, University of Montes Claros, Montes Claros, Minas Gerais
- Health Science Program, State University of Montes Claros, Montes Claros, Minas Gerais
| | - Mário Rodrigues de Melo-Filho
- Oncologic Dentistry Service, Santa Casa Hospital, Montes Claros, Minas Gerais
- Dental School, University of Montes Claros, Montes Claros, Minas Gerais
| | - Jacks Jorge
- Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo
| | | | | |
Collapse
|
114
|
Chen Y, Liu J, Wang W, Xiang L, Wang J, Liu S, Zhou H, Guo Z. High expression of hnRNPA1 promotes cell invasion by inducing EMT in gastric cancer. Oncol Rep 2018; 39:1693-1701. [PMID: 29484423 PMCID: PMC5868405 DOI: 10.3892/or.2018.6273] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/09/2018] [Indexed: 12/20/2022] Open
Abstract
Advanced gastric cancer (GC) has a poor prognosis and its treatment strategies are not very efficient. Heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) has emerged as a plausible GC marker, however the role and molecular mechanism of hnRNPA1 in cell invasion and migration remains unknown. In the present study, the gene expression across normal and tumor tissue (GENT) database was used to evaluate the mRNA expression of hnRNPA1 in various types of cancer. Western blot analysis (WB) and immunohistochemistry (IHC) were performed to detect the protein expression of hnRNPA1 in GC tissues and adjacent non-tumor tissues. The expression of multiple oncogenes was detected by western blot analysis and quantitative RT-PCR in hnRNPA1 overexpressing GC cells. Soft agar colony formation, EdU incorporation, wound healing and invasion assays were applied to verify the role of hnRNPA1 in anchorage-independent cell growth, migration and invasion in GC cells. Epithelial-to-mesenchymal transition (EMT) markers were detected by immunofluorescence, western blot analysis and IHC in vitro. A nude mice model of metastasis carcinoma was established to confirm the role of hnRNPA1 during EMT in vivo. Our results revealed that hnRNPA1 was significantly upregulated in GC tissue. HnRNPA1 overexpression significantly induced cell growth, migration and invasion ability in GC cells. In addition, hnRNPA1 promoted EMT of GC cells in vitro and in vivo. These findings indicated that hnRNPA1 is highly expressed in GC and promoted invasion by inducing EMT transition in GC cells. Thus, hnRNPA1 may be a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Yahua Chen
- Department of Gastroenterology, Longgang People's Hospital, Shenzhen, Guangdong 518172, P.R. China
| | - Jun Liu
- Department of Gastroenterology, Longgang People's Hospital, Shenzhen, Guangdong 518172, P.R. China
| | - Wei Wang
- Department of Gastroenterology, Longgang People's Hospital, Shenzhen, Guangdong 518172, P.R. China
| | - Li Xiang
- Department of Gastroenterology, Longgang People's Hospital, Shenzhen, Guangdong 518172, P.R. China
| | - Jide Wang
- Department of Gastroenterology, Longgang People's Hospital, Shenzhen, Guangdong 518172, P.R. China
| | - Side Liu
- Department of Gastroenterology, Longgang People's Hospital, Shenzhen, Guangdong 518172, P.R. China
| | - Hongyan Zhou
- Department of Pathology, The First People's Hospital of Xinxiang City, Xinxiang, Henan 453100, P.R. China
| | - Zheng Guo
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
115
|
Carnosol suppresses patient-derived gastric tumor growth by targeting RSK2. Oncotarget 2018; 9:34200-34212. [PMID: 30344937 PMCID: PMC6188138 DOI: 10.18632/oncotarget.24409] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/09/2018] [Indexed: 11/25/2022] Open
Abstract
Carnosol is a phenolic diterpene that is isolated from rosemary, sage, and oregano. It has been reported to possess anti-oxidant, anti-inflammatory, and anti-cancer properties. However, the molecular mechanism of carnosol's activity against gastric cancer has not been investigated. Herein, we report that carnosol is an RSK2 inhibitor that attenuates gastric cancer growth. Carnosol reduced anchorage-dependent and -independent gastric cancer growth by inhibiting the RSKs-CREB signaling pathway. The results of in vitro screening and cell-based assays indicated that carnosol represses RSK2 activity and its downstream signaling. Carnosol increased the G2/M phase and decreased S phase cell cycle and also induced apoptosis through the activation of caspases 9 and 7 and inhibition of Bcl-xL expression. Notably, oral administration of carnosol suppressed patient-derived gastric tumor growth in an in vivo mouse model. Our findings suggest that carnosol is an RSK2 inhibitor that could be useful for treating gastric cancer.
Collapse
|
116
|
Wang D, Sun Y, Li W, Ye F, Zhang Y, Guo Y, Zhang DY, Suo J. Antiproliferative effects of the CDK6 inhibitor PD0332991 and its effect on signaling networks in gastric cancer cells. Int J Mol Med 2018; 41:2473-2484. [PMID: 29436583 PMCID: PMC5846637 DOI: 10.3892/ijmm.2018.3460] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 12/20/2017] [Indexed: 12/31/2022] Open
Abstract
PD0332991 (palbociclib/Ibrance®) is a cyclin-dependent kinase (CDK)4/6 inhibitor that has recently been approved for the treatment of estrogen receptor-positive advanced breast cancer. The present study investigated the antiproliferative effects of PD0332991 on gastric cancer (GC) cells and the underlying molecular mechanisms. The activity of PD0332991 was tested in several GC cell lines, including AGS, KATO-III, NCI-N87 and HS746T. Growth inhibitory activity of PD0332991, alone or in combination with fluorouracil (5-FU), was measured by MTT assay. The effects of PD0332991 on cell cycle progression were analyzed by flow cytometry and western blotting. Protein pathway array and Ingenuity Pathway Analysis were used to identify signaling pathways that may mediate the antiproliferative effects of PD0332991. PD0332991 inhibited proliferation in a dose-dependent manner and enhanced the activity of 5-FU in all GC cell lines tested. Cells treated with PD0332991 exhibited cell cycle arrest in G1 phase of the cell cycle, whereas the number of cells in G2/M phase was decreased. PD0332991 also inhibited CDK6-specific phosphorylation of retinoblastoma on Ser780, reduced the expression of cyclin D1, and induced expression of p53 and p27. Furthermore, 31 proteins were identified, the expression of which was significantly altered following treatment with PD0332991 in at least three cell lines. Pathway analysis indicated that the altered proteins were frequently associated with cell death, cell cycle and the molecular mechanism of cancer. The results of the present study indicated that PD0332991 may inhibit cell proliferation via modulation of the cell cycle, and may affect numerous oncogenic signaling pathways. Therefore, PD0332991 may be considered effective for the treatment of GC.
Collapse
Affiliation(s)
- Daguang Wang
- Department of Gastric and Colorectal Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yabin Sun
- Department of Ophthalmology, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei Li
- Department of Gastric and Colorectal Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fei Ye
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yang Zhang
- Department of Gastric and Colorectal Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yuchen Guo
- Department of Gastric and Colorectal Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - David Y Zhang
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jian Suo
- Department of Gastric and Colorectal Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
117
|
Binato R, Santos EC, Boroni M, Demachki S, Assumpção P, Abdelhay E. A common molecular signature of intestinal-type gastric carcinoma indicates processes related to gastric carcinogenesis. Oncotarget 2018; 9:7359-7371. [PMID: 29484116 PMCID: PMC5800908 DOI: 10.18632/oncotarget.23670] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 12/11/2017] [Indexed: 12/11/2022] Open
Abstract
Gastric carcinoma (GC) is one of the most aggressive cancers and the second leading cause of cancer death in the world. According to the Lauren classification, this adenocarcinoma is divided into two subtypes, intestinal and diffuse, which differ in their clinical, epidemiological and molecular features. Several studies have attempted to delineate the molecular signature of gastric cancer to develop new and non-invasive screening tests that improve diagnosis and lead to new treatment strategies. However, a consensus signature has not yet been identified for each condition. Thus, this work aimed to analyze the gene expression profile of Brazilian intestinal-type GC tissues using microarrays and compare the results to those of non-tumor tissue samples. Moreover, we compared our intestinal-type gastric carcinoma profile with those obtained from populations worldwide to assess their similarity. The results identified a molecular signature for intestinal-type GC and revealed that 38 genes differentially expressed in Brazilian intestinal-type gastric carcinoma samples can successfully distinguish gastric tumors from non-tumor tissue in the global population. These differentially expressed genes participate in biological processes important to cell homeostasis. Furthermore, Kaplan-Meier analysis suggested that 7 of these genes could individually be able to predict overall survival in intestinal-type gastric cancer patients.
Collapse
Affiliation(s)
- Renata Binato
- Laboratório de Célula tronco, Centro de Transplante de Medula Óssea (CEMO), Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Ciência e Tecnologia Para o Controle do Câncer (INCT), Rio de Janeiro, RJ, Brazil
| | - Everton Cruz Santos
- Laboratório de Célula tronco, Centro de Transplante de Medula Óssea (CEMO), Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Ciência e Tecnologia Para o Controle do Câncer (INCT), Rio de Janeiro, RJ, Brazil
| | - Mariana Boroni
- Laboratório de Bioinformática e Biologia Computacional, Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil
| | - Samia Demachki
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará (UFPA), Belém, PA, Brazil
| | - Paulo Assumpção
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará (UFPA), Belém, PA, Brazil
| | - Eliana Abdelhay
- Laboratório de Célula tronco, Centro de Transplante de Medula Óssea (CEMO), Instituto Nacional de Câncer (INCA), Rio de Janeiro, RJ, Brazil
- Instituto Nacional de Ciência e Tecnologia Para o Controle do Câncer (INCT), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
118
|
Mokhtari E, Mokhtari H, Moslemi E. HER-3 Knocking Down Induces G2/M Arrest in Gastric Cancer Cells. Avicenna J Med Biotechnol 2018; 10:227-232. [PMID: 30555655 PMCID: PMC6252031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
BACKGROUND The Human Epidermal growth factor Receptor-3 (HER-3) is a member of ErbB receptor family and has deficient kinase activity. HER-3 should heterodimerize with other members of ErbB receptor family, especially with HER-2, to transduce downstream signaling pathways. HER-3 co-expresses with other ErbB receptors in different cancers and overexpresses while the oncogenic signaling pathways such as Jak/Stat, MAPK, and PI3K/Akt are activated and promoted. Here, the expression level of HER-3 was evaluated in Iranian gastric adenocarcinoma's patients and the effects of HER-3 knocking down was investigated on cell cycle and cell viability of human gastric adenocarcinoma cell line of MKN45. METHODS In this study, 38 paraffin-embedded surgical adenocarcinoma specimens and their marginal non-tumor tissue samples were collected. Total RNAs were extracted and cDNAs were synthesized. Finally, the expression level of HER-3 was evaluated by real time PCR approach. Moreover, the human adenocarcinoma cell line of MKN45 was transfected with siRNA against HER-3 and the effects of its down-regulation were evaluated using MTT assay and cell-cycle analysis. RESULTS The data obtained from this study revealed HER-3 is significantly overexpressed in gastric tumors rather than non-tumor marginal tissues. Also, it was found that the expression level of HER-3 is elevated with tumor depth of invasion. Moreover, HER-3 knocking down promotes cell accumulation in G2/M phase of cell cycle and decreases cell viability in MKN45 cells which suggests a potential role for HER-3 in gastric adenocarcinoma tumorigenesis. CONCLUSION Taken together, these results emphasize the importance of HER-3 receptor in diagnosis and prognosis of gastric adenocarcinoma.
Collapse
Affiliation(s)
- Ehsan Mokhtari
- Department of Biology, Faculty of Biological Sciences, Islamic Azad University, East Tehran Branch, Tehran, Iran
| | - Hesamodin Mokhtari
- International Campus, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elham Moslemi
- Department of Biology, Faculty of Biological Sciences, Islamic Azad University, East Tehran Branch, Tehran, Iran,Corresponding author: Elham Moslemi, Ph.D., Department of Biology, Faculty of Biological Sciences, Islamic Azad University, East Tehran Branch, Tehran, Iran, Tel/Fax: +98 9123355872, +98 21 88194061, E-mail:
| |
Collapse
|
119
|
Digital Image Analysis of HER2 Immunostained Gastric and Gastroesophageal Junction Adenocarcinomas. Appl Immunohistochem Mol Morphol 2017; 25:320-328. [PMID: 27801737 PMCID: PMC5447782 DOI: 10.1097/pai.0000000000000463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Manual assessment of human epidermal growth factor receptor 2 (HER2) protein expression by immunohistochemistry (IHC) in gastric and gastroesophageal junction (GGEJ) adenocarcinomas is prone to interobserver variability and hampered by tumor heterogeneity and different scoring criteria. Equivocal cases are frequent, requiring additional in situ hybridization analysis. This study aimed to evaluate the accuracy of digital image analysis for the assessment of HER2 protein expression. In total, 110 GGEJ adenocarcinomas were included in tissue microarrays with 3 tissue cores per case. Two immunoassays, PATHWAY and HercepTest, and fluorescent in situ hybridization analysis were performed. The Visiopharm HER2-CONNECT Analysis Protocol Package was applied through the ONCOtopix digital image analysis software platform. HER2 membrane connectivity, calculated by the Analysis Protocol Package, was converted to standard IHC scores applying predetermined cutoff values for breast carcinoma as well as novel cutoff values. Cases with excessive cytoplasmic staining as well as HER2 amplified IHC negative cases were excluded from analysis. Applying HER2-CONNECT with connectivity cutoff values established for breast carcinoma resulted in 72.7% sensitivity and 100% specificity for the identification of HER2 positive gene amplified cases. By application of new cutoff values, the sensitivity increased to 100% without decreased specificity. With the new cutoff values, a 36% to 50% reduction of IHC equivocal cases was obtained. In conclusion, HER2-CONNECT with adjusted cutoff values seem to be an effective tool for standardized assessment of HER2 protein expression in GGEJ adenocarcinomas, decreasing the need for in situ hybridization analyzes.
Collapse
|
120
|
Feng A, Yuan X, Li X. MicroRNA-345 inhibits metastasis and epithelial-mesenchymal transition of gastric cancer by targeting FOXQ1. Oncol Rep 2017; 38:2752-2760. [PMID: 29048674 PMCID: PMC5780028 DOI: 10.3892/or.2017.6001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 04/07/2017] [Indexed: 01/20/2023] Open
Abstract
MicroRNAs (miRNAs) are a group of critical players in gastric cancer (GC). Among numerous cancer-related miRNAs, the expression level and functional role of miR-345 in GC has not been investigated. This study showed that miR-345 expression was decreased in GC. Decreased expression level of miR-345 was associated with occurrence of lymph metastasis and advanced TNM stage of GC patients. Patients with low expression level of miR-345 had reduced overall survival (OS) and disease-free survival (DFS). In vitro experiments showed that miR-345 could inhibit the migration and invasion of GC cells. In vivo experiments showed that miR-345 knockdown could promote lung metastasis of GC cells in nude mice. miR-345 was found to prevent the metastasis by inhibiting epithelial-mesenchymal transition (EMT) of GC cells. Furthermore, FOXQ1 was confirmed to be the downstream target of miR-345 in GC cells. Forced expression of FOXQ1 could reverse the inhibitory effects of miR-345 on GC metastasis, while knockdown of FOXQ1 prevented the promoting effects of miR-345 knockdown on GC metastasis. In summary, this study demonstrates miR-345 is a promising biomarker and therapeutic target in GC.
Collapse
Affiliation(s)
- Aiwen Feng
- Department of Enterochirurgia, Huaian First People's Hospital, Huaian, Jiangsu 223300, P.R. China
| | - Xiaoming Yuan
- Department of Enterochirurgia, Huaian First People's Hospital, Huaian, Jiangsu 223300, P.R. China
| | - Xiangwei Li
- Department of Gastroenterological Surgery, Huaian First People's Hospital, Huaian, Jiangsu 223300, P.R. China
| |
Collapse
|
121
|
Shafaghi A, Mansour- Ghanaei F, Joukar F, Nabavi F, Mansour- Ghanaei A, Esrafilian Soltani A. Stage Association of Preoperative Serum Carcinoembryonic Antigen with Gastric Adenocarcinoma in Iranian Patients. Asian Pac J Cancer Prev 2017; 18:2669-2672. [PMID: 29072067 PMCID: PMC5747387 DOI: 10.22034/apjcp.2017.18.10.2669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Gastric cancer is the second leading cause of cancer-related mortality and the fourth most common cancer globally. Tumor markers are needed for appropriate management and monitoring of treatment to improve quality of life. Recently, carcinoembryonic antigen (CEA) has been widely used as a tumor marker in the diagnosis and follow-up of some malignancies. The aim of this study was to evaluate the significance of CEA detection in the course of disease in gastric cancer patients at different stages. Methods: Seventy six cases of gastric adenocarcinoma from the Rasht Razi Hospital were studied between January 2016 and December 2016, along with a control group of 152 people. Serum CEA was measured by ELISA reader. Statistical analysis was performed using SPSS 14.0 for Windows (SPSS Inc., Chicago, USA). The two groups were also compared by cross-table analysis using Pearson’s chi-square test, with P-values <0.05 considered significant. Results: CEA was positive in 61.8 % of patients versus 2.6% of the control group (P = 0.0001). Some 21% of patients at stages I and II (initial disease) and 40.8% at stages III and IV (advanced disease) demonstrated positive CEA. which was significantly correlated with higher N stage and poor differentiation. Conclusions: Our study showed that a high preoperative CEA level was not prevalent in early stage gastric cancer patients. We recommend to design other prospective studies and meta-analyses for elucidation of claims for diagnostic efficacy.
Collapse
Affiliation(s)
- Afshin Shafaghi
- GI Cancer Screening and Prevention Research Center(GCSPRC), Guilan University of Medical Sciences), Rasht, Iran.
| | | | | | | | | | | |
Collapse
|
122
|
Wippel HH, Santos MDM, Clasen MA, Kurt LU, Nogueira FCS, Carvalho CE, McCormick TM, Neto GPB, Alves LR, da Gloria da Costa Carvalho M, Carvalho PC, Fischer JDSDG. Comparing intestinal versus diffuse gastric cancer using a PEFF-oriented proteomic pipeline. J Proteomics 2017; 171:63-72. [PMID: 29032071 DOI: 10.1016/j.jprot.2017.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 09/29/2017] [Accepted: 10/06/2017] [Indexed: 12/19/2022]
Abstract
Gastric cancer is the fifth most common malignant neoplasia and the third leading cause of cancer death worldwide. Mac-Cormick et al. recently showed the importance of considering the anatomical region of the tumor in proteomic gastric cancer studies; more differences were found between distinct anatomical regions than when comparing healthy versus diseased tissue. Thus, failing to consider the anatomical region could lead to differential proteins that are not disease specific. With this as motivation, we compared the proteomic profiles of intestinal and diffuse adenocarcinoma from the same anatomical region, the corpus. To achieve this, we used isobaric labeling (iTRAQ) of peptides, a 10-step HILIC fractionation, and reversed-phase nano-chromatography coupled online with a Q-Exactive Plus mass spectrometer. We updated PatternLab to take advantage of the new Comet-PEFF search engine that enables identifying post-translational modifications and mutations included in neXtProt's PSI Extended FASTA Format (PEFF) metadata. Our pipeline then uses a text-mining tool that automatically extracts PubMed IDs from the proteomic result metadata and drills down keywords from manuscripts related with the biological processes at hand. Our results disclose important proteins such as apolipoprotein B-100, S100 and 14-3-3 proteins, among many others, highlighting the different pathways enriched by each cancer type. SIGNIFICANCE Gastric cancer is a heterogeneous and multifactorial disease responsible for a significant number of deaths every year. Despite the constant improvement of surgical techniques and multimodal treatments, survival rates are low, mostly due to limited diagnostic techniques and late symptoms. Intestinal and diffuse types of gastric cancer have distinct clinical and pathological characteristics; yet little is known about the molecular mechanisms regulating these two types of gastric tumors. Here we compared the proteomic profile of diffuse and intestinal types of gastric cancer from the same anatomical location, the corpus, from four male patients. This methodological design aimed to eliminate proteomic variations resulting from comparison of tumors from distinct anatomical regions. Our PEFF-tailored proteomic pipeline significantly increased the identifications as when compared to previous versions of PatternLab.
Collapse
Affiliation(s)
- Helisa Helena Wippel
- Computational Mass Spectrometry & Proteomics Group, Carlos Chagas Institute, Fiocruz - Paraná, Brazil
| | | | - Milan Avila Clasen
- Computational Mass Spectrometry & Proteomics Group, Carlos Chagas Institute, Fiocruz - Paraná, Brazil
| | - Louise Ulrich Kurt
- Computational Mass Spectrometry & Proteomics Group, Carlos Chagas Institute, Fiocruz - Paraná, Brazil
| | - Fabio Cesar Sousa Nogueira
- Laboratory of Proteomics, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Laboratory of Protein Chemistry, Proteomic Unit, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Eduardo Carvalho
- Pathology Service of the Clementino Fraga Filho University Hospital (HUCFF-UFRJ), Rio de Janeiro, Brazil
| | | | - Guilherme Pinto Bravo Neto
- Division of Esophageal and Gastric Surgery, General Surgery Service of the HUCFF-UFRJ, Rio de Janeiro, Brazil
| | | | | | - Paulo Costa Carvalho
- Computational Mass Spectrometry & Proteomics Group, Carlos Chagas Institute, Fiocruz - Paraná, Brazil.
| | | |
Collapse
|
123
|
Fehim S, Bouhaous R, Diaf M, Drici AM, Khaled MB. Epidemiological profile of gastric cancer in the northwestern region of Algeria: about 116 cases. J Gastrointest Oncol 2017; 8:659-664. [PMID: 28890816 PMCID: PMC5582047 DOI: 10.21037/jgo.2017.06.02] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 05/04/2017] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) remains a public health issue despite the global incidence decrease over the last decades. GC is the third leading cause of cancer-related death worldwide and stills of poor prognosis due to its late detection. The overall objective of our study was to describe the epidemiological, clinical, and histopathological aspects of stomach cancer. However, the specific purpose was to determine the gastric precancerous lesions among patients with histologically confirmed GC admitted at the gastroenterology department of the University Hospital of Sidi-Bel-Abbes city, Algeria. METHODS We retrospectively analyzed the patients' medical records admitted from 2010 to 2015. Intended for the statistical analysis of the enrolled data, we executed Student t-test and Chi-squared test, significance level was set at 5%. Histologic evidence objectified the biopsies performed during endoscopy; Hematoxylin and Eosin staining as well as the slides interpretation were executed at the level of the pathology department as a routine examination. RESULTS 116 cases of GC were recorded. The mean age of the whole studied cohort was 58.96±14.75 years. We reported a sex ratio of 1.36 with a peak frequency observed in male gender (57.76%). Gastric precancerous lesions, as well as tumor histology, and topography were revealed. In our context, adenocarcinoma was significantly higher among male patients and linitis plastica was expressively related to female gender (P=0.01). CONCLUSIONS Management of gastric precancerous lesions additionally to early diagnosis represent effective preventive and therapeutic methods of GC.
Collapse
Affiliation(s)
- Safia Fehim
- Department of Biology, Department of Biology, Djillali Liabes University, Sidi bel Abbes, Algeria
| | - Racheda Bouhaous
- Department of Biology, Department of Biology, Djillali Liabes University, Sidi bel Abbes, Algeria
| | - Mustapha Diaf
- Department of Biology, Department of Biology, Djillali Liabes University, Sidi bel Abbes, Algeria
- Biotoxicology Laboratory, Department of Biology, Djillali Liabes University, Sidi bel Abbes, Algeria
| | - Amine Mokhtar Drici
- Department of Biology, Department of Biology, Djillali Liabes University, Sidi bel Abbes, Algeria
| | - Meghit Boumediene Khaled
- Department of Biology, Department of Biology, Djillali Liabes University, Sidi bel Abbes, Algeria
- Biotoxicology Laboratory, Department of Biology, Djillali Liabes University, Sidi bel Abbes, Algeria
| |
Collapse
|
124
|
Li C, Liang G, Yang S, Sui J, Yao W, Shen X, Zhang Y, Peng H, Hong W, Xu S, Wu W, Ye Y, Zhang Z, Zhang W, Yin L, Pu Y. Dysregulated lncRNA-UCA1 contributes to the progression of gastric cancer through regulation of the PI3K-Akt-mTOR signaling pathway. Oncotarget 2017; 8:93476-93491. [PMID: 29212166 PMCID: PMC5706812 DOI: 10.18632/oncotarget.19281] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/12/2017] [Indexed: 12/30/2022] Open
Abstract
The long non-coding RNA (lncRNA) urothelial carcinoma-associated 1 (UCA1) has been recently shown to be dysregulated during disease occurrence and to play an important role in the progression of several cancers. However, the biological role and potential regulation mechanism of UCA1 in the carcinogenesis of gastric cancer remain unclear. In the present study, we found that UCA1 was aberrantly upregulated in gastric cancer tissues and gastric cancer cell lines, and was associated with TNM stage and metastasis. UCA1 silencing significantly inhibited gastric cancer BGC-823 cell proliferation and increased its apoptosis. We also found that UCA1 played an important role in the migration and invasion of gastric cancer cells in vitro and in vivo. The molecular mechanism of UCA1 suggested that UCA1 regulates the PI3K-Akt-mTOR signaling proteins and their downstream mediators, to alter gastric cancer progression in vitro and in vivo. Collectively, the results showed a pivotal role of UCA1 in the tumorigenesis of gastric cancer. In addition, the study characterized a novel lncRNA-mRNA regulatory network, which may lead to a better understanding of the pathogenesis of gastric cancer and assist in lncRNA-directed diagnosis and therapy for this malignancy.
Collapse
Affiliation(s)
- Chengyun Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Sheng Yang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Jing Sui
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Wenzhuo Yao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xian Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yanqiu Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Hui Peng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Weiwei Hong
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Siyi Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Wenjuan Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yancheng Ye
- Gansu Wuwei Tumor Hospital, Wuwei, Gansu 733000, P.R. China
| | - Zhiyi Zhang
- Gansu Wuwei Tumor Hospital, Wuwei, Gansu 733000, P.R. China
| | - Wenhua Zhang
- Gansu Wuwei Tumor Hospital, Wuwei, Gansu 733000, P.R. China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
125
|
LINE-1 hypomethylation is not a common event in preneoplastic stages of gastric carcinogenesis. Sci Rep 2017; 7:4828. [PMID: 28684753 PMCID: PMC5500474 DOI: 10.1038/s41598-017-05143-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/16/2017] [Indexed: 02/07/2023] Open
Abstract
LINE-1 hypomethylation is widely accepted as marker for global genomic DNA hypomethylation, which is a frequent event in cancer. The aim of the study was to evaluate LINE-1 methylation status at different stages of gastric carcinogenesis and evaluate its prognostic potential in clinical settings. LINE-1 methylation was analyzed in 267 tissue samples by bisulfite pyrosequencing including primary colorectal cancer tissues (T-CRC) with corresponding adjacent colon mucosa (N-CRC), gastric cancer tissues (T-GC) with corresponding gastric mucosa (N-GC), normal gastric tissues (N), chronic non-atrophic and atrophic gastritis (CG). LINE-1 methylation level was lower in both T-GC and T-CRC when compared to paired adjacent tissues. No difference was observed for LINE-1 methylation status between patients with normal gastric mucosa, CG and N-GC. LINE-1 methylation in T-GC but not N-GC tended to correlate with age. Subgroup stratification analysis did not reveal significant differences in LINE-1 methylation status according to tumor stage, anatomical location, histological subtype, differentiation grade. We observed similar overall survival data between patients with high or low LINE-1 levels. In summary, LINE-1 hypomethylation is a characteristic feature in GC but not very common in early preneoplastic stages of gastric carcinogenesis. Prognostic role of LINE-1 hypomethylation in GC patients could not be confirmed in this cohort.
Collapse
|
126
|
Zhou J, Wu A, Yu X, Zhu J, Dai H. SIRT6 inhibits growth of gastric cancer by inhibiting JAK2/STAT3 pathway. Oncol Rep 2017; 38:1059-1066. [DOI: 10.3892/or.2017.5753] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 06/12/2017] [Indexed: 11/06/2022] Open
|
127
|
Menbari MN, Nasseri S, Menbari N, Mehdiabadi R, Alipur Y, Roshani D. The -160 (C>A) CDH1 Gene Promoter Polymorphism and Its Relationship with Survival of Patients with Gastric Cancer in
Kurdistan. Asian Pac J Cancer Prev 2017; 18:1561-1565. [PMID: 28669168 PMCID: PMC6373814 DOI: 10.22034/apjcp.2017.18.6.1561] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Introduction: Gastric cancer (GC) is the fourth most common type of neoplasm and the second cause of malignancy-related death across much of the world. Complex multi-factorial processes are involved in its genesis, classified in two determinant clusters: non-genetic and genetic. Variation in CDH1 gene expression may play an important role in increasing risk of diffuse and intestinal subtypes of GC. This tumor suppressor gene, located on chromosome 16q22.1, encodes a trans membrane glycoprotein called epithelial cadherin (E-cadherin). Materials and Methods: In this historical cohort study, from June 2004 to Journey 2005 we collected 50 samples from Kurdish patients with stage II pathologically diagnosed gastric cancer that underwent surgery. Tumor tissues were paraffin-embedded along with 54 control samples from non-ulcer dyspepsia (NUD) cases undergoing upper gastrointestinal endoscopy. Three biopsies were captured by endoscopy from each individual’s gastric antrum. Result: The mean age of the patients was 59.5±2 years. Some 23 cases (53.4%) had the CC genotype, 19 AC and 1 AA. H.pylori infection was noted in 30 patients (69%). Survival rates of gastric cancer patients were 90.7% in the first year, 39.5% in the second year and 6.9% in the third year. Female patients had higher survival rates (P=0.004). Conclusion: In this study we found that frequencies of -160(C>A) CDH1 genotypes were not comparable in H.pylori-infected and H.pylori-uninfected subjects in both case and control groups. These findings suggest that -160 (C>A) CDH1 polymorphism is not related with H.pylori infection susceptibility. In addition we found no significant relationship between the CDH1 -160(C/A) promoter polymorphism with predisposition to gastric cancer.
Collapse
Affiliation(s)
- Mohammad Nazir Menbari
- Cellular and Molecular Research Center, Kurdistan University of Medical
Sciences, Sanandaj, Iran.
| | | | | | | | | | | |
Collapse
|
128
|
Bi M, Yu H, Huang B, Tang C. Long non-coding RNA PCAT-1 over-expression promotes proliferation and metastasis in gastric cancer cells through regulating CDKN1A. Gene 2017; 626:337-343. [PMID: 28571676 DOI: 10.1016/j.gene.2017.05.049] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 05/10/2017] [Accepted: 05/24/2017] [Indexed: 02/06/2023]
Abstract
Prostate cancer-associated transcript1 (PCAT-1) is a novel lncRNA that involved in cell apoptosis and proliferation of prostate cancer. However, the role of PCAT-1 on gastric cancer remains unclear. In the present study, we found that PCAT-1 was increased in gastric cancer tissues and cell lines. Over-expression of PCAT-1 was correlated with poor overall survival in gastric cancer patients. PCAT-1 knockdown through shRNA in AGS and MGC-803 cells inhibited cell proliferation, migration and invasion by regulating CDKN1A. Our data suggested that PCAT-1 could play an oncogenic role in gastric cancer progression. Silencing PCAT-1 is a potential novel therapeutic approach for gastric cancer.
Collapse
Affiliation(s)
- Mingjun Bi
- Department of General Surgery, The Affiliated Weihai Second Municipal Hospital of Qingdao University, 51 Guangming Road, Weihai 264200, PR China
| | - Hongmei Yu
- Department of Laboratory Medicine, The Affiliated Weihai Second Municipal Hospital of Qingdao University, 51 Guangming Road, Weihai 264200, PR China
| | - Bin Huang
- Department of General Surgery, The Affiliated Weihai Second Municipal Hospital of Qingdao University, 51 Guangming Road, Weihai 264200, PR China
| | - Cuiyan Tang
- Department of Laboratory Medicine, The Affiliated Weihai Second Municipal Hospital of Qingdao University, 51 Guangming Road, Weihai 264200, PR China.
| |
Collapse
|
129
|
López-Cortés A, Guerrero S, Redal MA, Alvarado AT, Quiñones LA. State of Art of Cancer Pharmacogenomics in Latin American Populations. Int J Mol Sci 2017; 18:E639. [PMID: 28545225 PMCID: PMC5485925 DOI: 10.3390/ijms18060639] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/07/2017] [Accepted: 03/10/2017] [Indexed: 12/22/2022] Open
Abstract
Over the past decades, several studies have shown that tumor-related somatic and germline alterations predicts tumor prognosis, drug response and toxicity. Latin American populations present a vast geno-phenotypic diversity due to the great interethnic and interracial mixing. This genetic flow leads to the appearance of complex characteristics that allow individuals to adapt to endemic environments, such as high altitude or extreme tropical weather. These genetic changes, most of them subtle and unexplored, could establish a mutational profile to develop new pharmacogenomic therapies specific for Latin American populations. In this review, we present the current status of research on somatic and germline alterations in Latin America compared to those found in Caucasian and Asian populations.
Collapse
Affiliation(s)
- Andrés López-Cortés
- Centro de Investigación Genética y Genómica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad Tecnológica Equinoccial, Quito 170527, Ecuador.
| | - Santiago Guerrero
- Gene Regulation, Stem Cells and Cancer Programme, Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Universitat Pompeu Fabra (UPF), Barcelona 08003, Spain.
| | - María Ana Redal
- Instituto de Fisiopatología y Bioquímica Clínica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Centro de Diagnóstico Molecular, MEDgenomica, Buenos Aires 1000-1499, Argentina.
| | - Angel Tito Alvarado
- Unidad de Bioequivalencia y Medicina Personalizada, Facultad de Medicina, Universidad de San Martín de Porres, Lima 12, Peru.
| | - Luis Abel Quiñones
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology, Faculty of Medicine, University of Chile, Santiago 70111, Chile.
| |
Collapse
|
130
|
Jung SJ, Cho JH, Park WJ, Heo YR, Lee JH. Telomere length is correlated with mitochondrial DNA copy number in intestinal, but not diffuse, gastric cancer. Oncol Lett 2017; 14:925-929. [PMID: 28693253 DOI: 10.3892/ol.2017.6197] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/03/2017] [Indexed: 01/07/2023] Open
Abstract
A positive correlation between telomere length and mitochondrial DNA (mtDNA) copy number has previously been observed in healthy individuals, and in patients with psychiatric disorders. In the present study, telomere length and mtDNA copy number were evaluated in gastric cancer (GC) tissue samples. DNA was extracted from 109 GC samples (including 82 intestinal, and 27 diffuse cases), and the telomere length and mtDNA copy number were analyzed using a quantitative-polymerase chain reaction assay. The relative telomere length and mtDNA copy number in tumor tissue, as compared with in normal tissue, (mean ± standard deviation) in all GC samples were 11.48±1.14 and 14.86±1.35, respectively. Telomere length and mtDNA copy number were not identified as exhibiting clinical or prognostic value for GC. However, positive correlations between telomere length and mitochondrial DNA copy number were identified in GC (r=0.408, P<0.001) and in the adjacent normal mucosa (r=0.363; P<0.001). When stratified by Lauren classification, the correlation was identified in intestinal type GC samples (r=0.461; P<0.001), but not in diffuse type GC samples (r=0.225; P=0.260). This result indicated that loss of the correlation of telomeres and mitochondrial function may induce the initiation or progression of GC pathogenesis.
Collapse
Affiliation(s)
- Soo-Jung Jung
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Ji-Hyoung Cho
- Department of General Surgery, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Won-Jin Park
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Yu-Ran Heo
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Jae-Ho Lee
- Department of Anatomy, School of Medicine, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
131
|
Wessler S, Krisch LM, Elmer DP, Aberger F. From inflammation to gastric cancer - the importance of Hedgehog/GLI signaling in Helicobacter pylori-induced chronic inflammatory and neoplastic diseases. Cell Commun Signal 2017; 15:15. [PMID: 28427431 PMCID: PMC5397778 DOI: 10.1186/s12964-017-0171-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/07/2017] [Indexed: 02/07/2023] Open
Abstract
Infections with the human pathogen Helicobacter pylori (H. pylori) are closely associated with the development of inflammatory disorders and neoplastic transformation of the gastric epithelium. Drastic changes in the micromilieu involve a complex network of H. pylori-regulated signal transduction pathways leading to the release of proinflammatory cytokines, gut hormones and a wide range of signaling molecules. Besides controlling embryonic development, the Hedgehog/GLI signaling pathway also plays important roles in epithelial proliferation, differentiation, and regeneration of the gastric physiology, but also in the induction and progression of inflammation and neoplastic transformation in H. pylori infections. Here, we summarize recent findings of H. pylori-associated Hedgehog/GLI signaling in gastric homeostasis, malignant development and the modulation of the gastric tumor microenvironment.
Collapse
Affiliation(s)
- Silja Wessler
- Division of Microbiology, Cancer Cluster Salzburg, Department of Molecular Biology, Paris-Lodron University of Salzburg, Billroth Strasse 11, A-5020, Salzburg, Austria.
| | - Linda M Krisch
- Division of Microbiology, Cancer Cluster Salzburg, Department of Molecular Biology, Paris-Lodron University of Salzburg, Billroth Strasse 11, A-5020, Salzburg, Austria
| | - Dominik P Elmer
- Division of Molecular Tumor Biology, Cancer Cluster Salzburg, Department of Molecular Biology, Paris-Lodron University of Salzburg, Hellbrunner Strasse 34, A-5020, Salzburg, Austria
| | - Fritz Aberger
- Division of Molecular Tumor Biology, Cancer Cluster Salzburg, Department of Molecular Biology, Paris-Lodron University of Salzburg, Hellbrunner Strasse 34, A-5020, Salzburg, Austria.
| |
Collapse
|
132
|
Bahrami A, Shahidsales S, Khazaei M, Ghayour-Mobarhan M, Maftouh M, Hassanian SM, Avan A. C-Met as a potential target for the treatment of gastrointestinal cancer: Current status and future perspectives. J Cell Physiol 2017; 232:2657-2673. [DOI: 10.1002/jcp.25794] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Afsane Bahrami
- Molecular Medicine Group, Department of Modern Sciences and Technology; Mashhad University of Medical Sciences; Mashhad Iran
- Student Research Center, Faculty of Medicine; Mashhad University of Medical Sciences; Mashhad Iran
| | - Soodabeh Shahidsales
- Cancer Research Center; School of Medicine, Mashhad University of Medical Sciences; Mashhad Iran
| | - Majid Khazaei
- Neurogenic Inflammatory Research Center and Department of Physiology; Mashhad University of Medical Sciences; Mashhad Iran
| | - Majid Ghayour-Mobarhan
- Metabolic syndrome Research Center; School of Medicine, Mashhad University of Medical Sciences; Mashhad Iran
| | - Mina Maftouh
- Metabolic syndrome Research Center; School of Medicine, Mashhad University of Medical Sciences; Mashhad Iran
| | - Seyed Mahdi Hassanian
- Metabolic syndrome Research Center; School of Medicine, Mashhad University of Medical Sciences; Mashhad Iran
- Department of Medical Biochemistry; School of Medicine, Mashhad University of Medical Sciences; Mashhad Iran
| | - Amir Avan
- Metabolic syndrome Research Center; School of Medicine, Mashhad University of Medical Sciences; Mashhad Iran
| |
Collapse
|
133
|
Muñoz M, Rosso M, Carranza A, Coveñas R. Increased nuclear localization of substance P in human gastric tumor cells. Acta Histochem 2017; 119:337-342. [PMID: 28325510 DOI: 10.1016/j.acthis.2017.03.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/08/2017] [Accepted: 03/08/2017] [Indexed: 12/21/2022]
Abstract
Gastric cancer (GC) is an aggressive disease that remains the fourth most common type of cancer and is the second leading cause of cancer-related death worldwide. Treatment of advanced or metastatic GC has seen little progress and median overall survival in this group remains <1 year. It is urgent to investigate new mechanisms to understand GC progression. It is known that substance P (SP), after binding to the neurokinin-1 (NK-1) receptor, elicits GC proliferation; that GC cells and samples express NK-1 receptors; that NK-1 receptor antagonists, in a concentration dependent manner, inhibit the proliferation of GC cells and that these cells die by apoptosis. However, the presence of SP in GC and normal gastric cells is unknown. In order to know more on the involvement of the SP/NK-1 receptor system in GC, we studied in thirty human GC and normal gastric samples the immunolocalization of SP after using an immunohistochemical technique. SP was observed in the cytoplasm and in the nucleus of GC and normal gastric cells. The nuclear expression of SP was higher in GC cells than in normal cells. No significant difference was observed when the cytoplasmatic expression of SP in normal and GC cells was compared. The findings suggest that SP plays an important role in both nuclear function and GC.
Collapse
Affiliation(s)
- Miguel Muñoz
- Virgen del Rocío University Hospital, Research Laboratory on Neuropeptides (IBIS), Sevilla, Spain.
| | - Marisa Rosso
- Virgen del Rocío University Hospital, Research Laboratory on Neuropeptides (IBIS), Sevilla, Spain
| | - Andrés Carranza
- Virgen del Rocío University Hospital, Research Laboratory on Neuropeptides (IBIS), Sevilla, Spain
| | - Rafael Coveñas
- Institute of Neurosciences of Castilla y León (INCYL), Laboratory of Neuroanatomy of the Peptidergic Systems (Lab. 14), University of Salamanca, Salamanca, Spain
| |
Collapse
|
134
|
Beheshtizadeh M, Moslemi E. Analysis of G3BP1 and VEZT Expression in Gastric Cancer and Their Possible Correlation with Tumor Clinicopathological Factors. J Gastric Cancer 2017; 17:43-51. [PMID: 28337362 PMCID: PMC5362833 DOI: 10.5230/jgc.2017.17.e5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 03/04/2017] [Accepted: 03/04/2017] [Indexed: 12/21/2022] Open
Abstract
Purpose This study aimed to analyze G3BP1 and VEZT expression profiles in patients with gastric cancer, and examine the possible relationship between the expressions of each gene and clinicopathological factors. Materials and Methods Expression of these genes in formalin-fixed paraffin embedded (FFPE) tissues, collected from 40 patients with gastric cancer and 40 healthy controls, was analyzed. Differences in gene expression among patient and normal samples were identified using the GraphPad Prism 5 software. For the analysis of real-time polymerase chain reaction products, GelQuantNET software was used. Results Our findings demonstrated that both VEZT and G3BP1 mRNA expression levels were downregulated in gastric cancer samples compared with those in the normal controls. No significant relationship was found between the expression of these genes and gender (P-value, 0.4835 vs. 0.6350), but there were significant changes associated with age (P-value, 0.0004 vs. 0.0001) and stage of disease (P-value, 0.0019 vs. 0.0001). In addition, there was a direct relationship between VEZT gene expression and metastasis (P-value, 0.0462), in contrast to G3BP1 that did not demonstrate any significant correlation (P-value, 0.1833). Conclusions The results suggest that expression profiling of VEZT and G3BP1 can be used for diagnosis of gastric cancer, and specifically, VEZT gene could be considered as a biomarker for the detection of gastric cancer progression.
Collapse
Affiliation(s)
| | - Elham Moslemi
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
135
|
Wu X, Zhou J, Wu Z, Chen C, Liu J, Wu G, Zhai J, Liu F, Li G. miR-101-3p Suppresses HOX Transcript Antisense RNA (HOTAIR)-Induced Proliferation and Invasion Through Directly Targeting SRF in Gastric Carcinoma Cells. Oncol Res 2017; 25:1383-1390. [PMID: 28251884 PMCID: PMC7841108 DOI: 10.3727/096504017x14879366402279] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
miR-101-3p has been identified as a tumor suppressor in several cancers, but its exact role in gastric adenocarcinoma is still largely unknown. In this study, we found that, compared with the RGM-1 human normal gastric epithelial cells, miR-101-3p was significantly downregulated in all six human gastric adenocarcinoma cell lines, including BGC-823, MNK-45, MGC-803, SGC-7901, AGS, and HGC-27. Overexpression of miR-101-3p suppressed both the proliferation and invasion of AGS gastric adenocarcinoma cells, and knockdown of miR-101-3p displayed the opposite effect. In addition, miR-101-3p could directly target and suppress the expression of the serum response factor (SRF) gene, which is a transcription factor of HOTAIR, a well-characterized tumor promoter lncRNA. miR-101-3p negatively regulated SRF-mediated transcription of HOTAIR. Moreover, silencing of either SRF or HOTAIR could counteract the promotion of gastric adenocarcinoma cell proliferation and invasion by miR-101-3p inhibition. Our findings indicate that miR-101-3p suppresses HOTAIR-induced proliferation and invasion through directly targeting SRF in gastric carcinoma cells.
Collapse
|
136
|
Wang R, Fang H, Fang Q. Downregulation of FKBP14 by RNA interference inhibits the proliferation, adhesion and invasion of gastric cancer cells. Oncol Lett 2017; 13:2811-2816. [PMID: 28454471 DOI: 10.3892/ol.2017.5781] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 01/19/2017] [Indexed: 01/08/2023] Open
Abstract
FK506 binding protein (FBBP) 14 belongs to the family of FKBPs. Altered expression of FKBPs are observed in several malignancies. The present study aimed to explore the expression and biological function of FKBP14 in gastric cancer. FKBP14 expression levels in 40 gastric cancer samples and matched control samples were evaluated using quantitative polymerase chain reaction. Cell proliferation was evaluated using Cell Counting kit-8 assay. A cell adhesion and a Transwell assay were performed to detect cell adhesion and invasion. Protein expression was determined using western blot analysis. It was found that FKBP14 expression in gastric cancer tissues was elevated compared with normal tissues. Silencing of FKBP14 expression in the gastric cancer MKN-45 and AGS cell lines, which have a higher expression level of FKBP14 compared with four other gastric cancer cell lines, significantly inhibited cellular proliferation, adhesion and invasion. In addition, the protein levels of proliferating cell nuclear antigen, matrix metalloproteinase 2 and the epithelial-mesenchymal-transition (EMT) markers β-catenin, Snail1 and Twist were repressed in gastric cancer cells with FKBP14 silenced. In conclusion, FKBP14 may act as an oncogene by suppressing cellular proliferation, adhesion and invasion and EMT in gastric carcinogenesis. FKBP14 may be a diagnosis marker and potential therapeutic target in gastric cancer.
Collapse
Affiliation(s)
- Ruizhong Wang
- Department of Laboratory, People's Hospital of Pudong New Area of Shanghai, Shanghai 201299, P.R. China
| | - Hua Fang
- Department of Laboratory, People's Hospital of Pudong New Area of Shanghai, Shanghai 201299, P.R. China
| | - Qin Fang
- Department of Hyperbaric Oxygen, People's Hospital of Pudong New Area of Shanghai, Shanghai 201299, P.R. China
| |
Collapse
|
137
|
Azarbarzin S, Feizi MAH, Safaralizadeh R, Kazemzadeh M, Fateh A. The Value of MiR-383, an Intronic MiRNA, as a Diagnostic and Prognostic Biomarker in Intestinal-Type Gastric Cancer. Biochem Genet 2017; 55:244-252. [PMID: 28243881 DOI: 10.1007/s10528-017-9793-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 02/16/2017] [Indexed: 12/13/2022]
Abstract
MicroRNAs, a class of gene expression regulatory non-coding RNAs, participate in the pathogenic mechanisms of gastric cancer which is one of the life-treating cancers. Due to its aberrant expression in some types of human cancer, miR-383 has the value of being investigated in relation to cancer treatment and diagnosis. MiR-383 is placed in intron of SGCZ, a protein-coding gene, which is subject to dysregulation in various diseases. The purpose of the current study was to investigate the contribution of miR-383 to intestinal-type gastric adenocarcinoma tumorigenesis. The expression level of miR-383 was investigated by qRT-PCR in pairs of tumorous and adjacent tumor-free tissues of 40 patients with gastric cancer during endoscopy. Also, the susceptibility of miR-383 as a tumor marker and the relationship between its aberrant expression and clinicopathological features were determined. qRT-PCR data showed that miR-383 was dysregulated during gastric tumorigenesis. MiR-383 was dramatically downregulated up to sevenfold in intestinal-type gastric adenocarcinoma compared with adjacent tumor-free tissues (P < 0.001). Misregulation of miR-383 did not reveal a significant correlation with clinical characteristics. The ROC area of 80% with 76% sensitivity and 84% specificity was determined by P < 0.001. The current study demonstrated downregulation of miR-383 in intestinal-type gastric adenocarcinoma. Downregulation of miR-383 might be used as a potential tumor marker for the diagnosis of gastric cancer or could be a potential target for gene therapy.
Collapse
Affiliation(s)
- Shirin Azarbarzin
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mina Kazemzadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Alavieh Fateh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| |
Collapse
|
138
|
The Interplay of LncRNA-H19 and Its Binding Partners in Physiological Process and Gastric Carcinogenesis. Int J Mol Sci 2017; 18:ijms18020450. [PMID: 28230721 PMCID: PMC5343984 DOI: 10.3390/ijms18020450] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/12/2017] [Accepted: 02/16/2017] [Indexed: 02/07/2023] Open
Abstract
Long non-coding RNA (lncRNA), a novel and effective modulator in carcinogenesis, has become a study hotspot in recent years. The imprinted oncofetal lncRNA H19 is one of the first identified imprinted lncRNAs with a high expression level in embryogenesis but is barely detectable in most tissues after birth. Aberrant alterations of H19 expression have been demonstrated in various tumors, including gastric cancer (GC), implicating a crucial role of H19 in cancer progression. As one of the top malignancies in the world, GC has already become a serious concern to public health with poor prognosis. The regulatory roles of H19 in gastric carcinogenesis have been explored by various research groups, which leads to the development of GC therapy. This review comprehensively summarizes the current knowledge of H19 in tumorigenesis, especially in GC pathogenesis, with emphasis on the underneath molecular mechanisms depicted from its functional partners. Furthermore, the accumulated knowledge of H19 will provide better understanding on targeted therapy of GC.
Collapse
|
139
|
Kamran M, Long ZJ, Xu D, Lv SS, Liu B, Wang CL, Xu J, Lam EWF, Liu Q. Aurora kinase A regulates Survivin stability through targeting FBXL7 in gastric cancer drug resistance and prognosis. Oncogenesis 2017; 6:e298. [PMID: 28218735 PMCID: PMC5337621 DOI: 10.1038/oncsis.2016.80] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/10/2016] [Accepted: 11/04/2016] [Indexed: 12/19/2022] Open
Abstract
Aurora kinase A (AURKA) has been implicated in the regulation of cell cycle progression, mitosis and a key number of oncogenic signaling pathways in various malignancies. However, little is known about its role in gastric cancer prognosis and genotoxic resistance. Here we found that AURKA was highly overexpressed in gastric cancer and inversely correlated with disease prognosis. Overexpression of AURKA exacerbated gastric cancer drug resistance through upregulating the expression of the anti-apoptotic protein Survivin. Conversely, we demonstrated that AURKA depletion caused a decrease in Survivin protein levels by increasing its ubiquitylation and degradation. Mass spectrometric analysis revealed that upon AURKA depletion, Survivin bound to the FBXL7 E3 ubiquitin ligase, which induced ubiquitin-proteasome degradation of Survivin. In addition, we showed that AURKA regulated FBXL7 both at the levels of transcription and translation. Moreover, proteomic analysis of nuclear AURKA-interacting proteins identified Forkhead box protein P1 (FOXP1). We next showed that AURKA was required for FBXL7 transcription and that AURKA negatively regulated FOXP1-mediated FBXL7 expression. The physiological relevance of the regulation of Survivin by AURKA through the FOXP1–FBXL7 axis was further underscored by the significant positive correlations between AURKA and Survivin expression in gastric cancer patient samples. Moreover, the AURKA depletion or kinase inhibition-induced apoptotic cell death could be reversed by Survivin ectopic overexpression, further supporting that AURKA regulated Survivin to enhance drug resistance. In agreement, inhibition of AURKA synergistically enhanced the cytotoxic effect of DNA-damaging agents in cancer cells by suppressing Survivin expression. Taken together, our data suggest that AURKA restricts Survivin ubiquitylation and degradation in gastric cancer to promote drug resistance and hence the AURKA–Survivin axis can be targeted to promote the efficacy of DNA-damaging agents in gastric cancer.
Collapse
Affiliation(s)
- M Kamran
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian/State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Z-J Long
- Department of Hematology, The Third Affiliated Hospital; Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| | - D Xu
- State key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine/Department of Gastric Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - S-S Lv
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian/State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - B Liu
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian/State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - C-L Wang
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian/State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - J Xu
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian/State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - E W-F Lam
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Q Liu
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian/State Key Laboratory of Oncology in South China, Cancer Center, Sun Yat-sen University, Guangzhou, China.,Department of Hematology, The Third Affiliated Hospital; Institute of Hematology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
140
|
Arora N, Alsaied O, Dauer P, Majumder K, Modi S, Giri B, Dudeja V, Banerjee S, Von Hoff D, Saluja A. Downregulation of Sp1 by Minnelide leads to decrease in HSP70 and decrease in tumor burden of gastric cancer. PLoS One 2017; 12:e0171827. [PMID: 28192510 PMCID: PMC5305197 DOI: 10.1371/journal.pone.0171827] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 01/26/2017] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Gastric cancer is the third leading cause of cancer related mortality worldwide with poor survival rates. Even though a number of chemotherapeutic compounds have been used against this disease, stomach cancer has not been particularly sensitive to these drugs. In this study we have evaluated the effect of triptolide, a naturally derived diterpene triepoxide and its water soluble pro-drug Minnelide on several gastric adenocarcinoma cell lines both as monotherapy and in combination with CPT-11. METHODS Gastric cancer cell lines MKN28 and MKN45 were treated with varying doses of triptolide in vitro. Cell viability was measured using MTT based assay kit. Apoptotic cell death was assayed by measuring caspase activity. Effect of the triptolide pro-drug, Minnelide, was evaluated by implanting the gastric cancer cells subcutaneously in athymic nude mice. RESULTS Gastric cancer cell lines MKN28 and MKN45 cells exhibited decreased cell viability and increased apoptosis when treated with varying doses of triptolide in vitro. When implanted in athymic nude mice, treatment with Minnelide reduced tumor burden in both MKN28 derived tumors as well as MKN45 derived tumors. Additionally, we also evaluated Minnelide as a single agent and in combination with CPT-11 in the NCI-N87 human gastric tumor xenograft model. CONCLUSION Our results indicated that the combination of Minnelide with CPT-11 resulted in significantly smaller tumors compared to control. These studies are extremely encouraging as Minnelide is currently undergoing phase 1 clinical trials for gastrointestinal cancers.
Collapse
Affiliation(s)
- Nivedita Arora
- Div. of Basic and Translational Research Dept. of Surgery University of Minnesota, Minneapolis MN, United States of America
| | - Osama Alsaied
- Div. of Basic and Translational Research Dept. of Surgery University of Minnesota, Minneapolis MN, United States of America
| | - Patricia Dauer
- Div. of Basic and Translational Research Dept. of Surgery University of Minnesota, Minneapolis MN, United States of America
- Div. of Surgical Oncology Dept. of Surgery University of Miami, Miami, FL, United States of America
| | - Kaustav Majumder
- Div. of Basic and Translational Research Dept. of Surgery University of Minnesota, Minneapolis MN, United States of America
| | - Shrey Modi
- Div. of Basic and Translational Research Dept. of Surgery University of Minnesota, Minneapolis MN, United States of America
- Div. of Surgical Oncology Dept. of Surgery University of Miami, Miami, FL, United States of America
| | - Bhuwan Giri
- Div. of Basic and Translational Research Dept. of Surgery University of Minnesota, Minneapolis MN, United States of America
- Div. of Surgical Oncology Dept. of Surgery University of Miami, Miami, FL, United States of America
| | - Vikas Dudeja
- Div. of Basic and Translational Research Dept. of Surgery University of Minnesota, Minneapolis MN, United States of America
- Div. of Surgical Oncology Dept. of Surgery University of Miami, Miami, FL, United States of America
| | - Sulagna Banerjee
- Div. of Basic and Translational Research Dept. of Surgery University of Minnesota, Minneapolis MN, United States of America
- Div. of Surgical Oncology Dept. of Surgery University of Miami, Miami, FL, United States of America
| | - Daniel Von Hoff
- TGen/Virginia G. Piper Cancer Ctr, Suite 600, Phoenix, AZ United States of America
| | - Ashok Saluja
- Div. of Basic and Translational Research Dept. of Surgery University of Minnesota, Minneapolis MN, United States of America
- Div. of Surgical Oncology Dept. of Surgery University of Miami, Miami, FL, United States of America
- * E-mail:
| |
Collapse
|
141
|
Xu Y, Cao X, Jiang J, Chen Y, Wang K. TNF-α-308/-238 polymorphisms are associated with gastric cancer: A case-control family study in China. Clin Res Hepatol Gastroenterol 2017; 41:103-109. [PMID: 27373488 DOI: 10.1016/j.clinre.2016.05.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 04/20/2016] [Accepted: 05/18/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND Associations of TNF-α-308 (rs1800629) and -238 (rs361525) with gastric cancer had the inconsistent indication among different populations. METHODS In this case-control family study, 47 families were determined with the probands diagnosed with gastric cancer (case family, n=296), accordingly 47 families without gastric cancer were matched with the case families by multivariate distribution of age, sex, social class, and pedigree size (control family, n=319). Polymerase chain reaction-restriction fragment length polymorphism (RFLP-PCR) was used to identify the TNF genotype. Chi-square test was used to compare the groups regarding genotype and the allele frequencies, HWE test for Hardy-Weinberg equilibrium. RESULTS The frequencies of TNF-α-308 GA and AA genotypes were significantly higher in case family than that in control family. The risk of gastric cancer was increased in GA and AA carriers in the first degree (OR=2.06, 95% CI=1.20-3.51 and OR=4.89, 95% CI=2.74-8.74), however the similar result was not found in the second degree. Helicobacter pylori infection status were significantly associated with risk of gastric cancer in first-degree relatives (OR=1.96, 95% CI=1.26-3.05) while no statistical significance was noted in the second-degree relatives. Haplotypes of TNF-α-308/-238 alleles, GA/GG, AA/GG and AA/GA indicated the susceptibilities to gastric cancer with OR and 95% confident intervals resulting 2.07 (1.34-3.21), 4.49 (2.74-7.33) and 4.98 (1.76-14.01) respectively. CONCLUSIONS TNF-α-G308A (rs1800629) polymorphisms are associated with gastric cancer in Chinese population. Haplotypes of TNF-α-308/-238 GA/GG, AA/GG and AA/GA increase the susceptibilities to gastric cancer. The first-degree relatives are more likely to develop into gastric cancer with TNF-α-G308 polymorphisms and H. pylori positive than the second-degree are.
Collapse
Affiliation(s)
- Yajuan Xu
- Zhengzhou University, College of Public Health, Zhengzhou 450001, Henan, PR China; Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou 450052, PR China
| | - Xiaoqin Cao
- Zhengzhou University, College of Public Health, Zhengzhou 450001, Henan, PR China; Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou 450052, PR China
| | - Jicheng Jiang
- Zhengzhou University, College of Public Health, Zhengzhou 450001, Henan, PR China; Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou 450052, PR China
| | - Yi Chen
- Zhengzhou University, College of Public Health, Zhengzhou 450001, Henan, PR China; Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou 450052, PR China
| | - Kaijuan Wang
- Zhengzhou University, College of Public Health, Zhengzhou 450001, Henan, PR China; Key Laboratory of Tumor Epidemiology of Henan Province, Zhengzhou 450052, PR China.
| |
Collapse
|
142
|
Patel TN, Roy S, Ravi R. Gastric cancer and related epigenetic alterations. Ecancermedicalscience 2017; 11:714. [PMID: 28144288 PMCID: PMC5243136 DOI: 10.3332/ecancer.2017.714] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer, a malignant and highly proliferative condition, has significantly affected a large population around the globe and is known to be caused by various factors including genetic, epigenetic, and environmental influences. Though the global trend of these cancers is declining, an increase in its frequency is still a threat because of changing lifestyles and dietary habits. However, genetic and epigenetic alterations related to gastric cancers also have an equivalent contribution towards carcinogenic development. DNA methylation is one of the major forms of epigenetic modification which plays a significant role in gastric carcinogenesis. Methylation leads to inactivation of some of the most important genes like DNA repair genes, cell cycle regulators, apoptotic genes, transcriptional regulators, and signalling pathway regulators; which subsequently cause uncontrolled proliferation of cells. Mutations in these genes can be used as suitable prognostic markers for early diagnosis of the disease, since late diagnosis of gastric cancers has a huge negative impact on overall patient survival. In this review, we focus on the important epigenetic mutations that contribute to the development of gastric cancer and the molecular pathogenesis underlying each of them. Methylation, acetylation, and histone modifications play an integral role in the onset of genomic instability, one of the many contributory factors to gastric cancer. This article also covers the constraints of incomplete knowledge of epigenetic factors influencing gastric cancer, thus throwing light on our understanding of the disease.
Collapse
Affiliation(s)
- Trupti N Patel
- Department of Medical Biotechnology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Soumyadipta Roy
- Department of Medical Biotechnology, VIT University, Vellore 632014, Tamil Nadu, India
| | - Revathi Ravi
- Department of Medical Biotechnology, VIT University, Vellore 632014, Tamil Nadu, India
| |
Collapse
|
143
|
Wasmer MH, Krebs P. The Role of IL-33-Dependent Inflammation in the Tumor Microenvironment. Front Immunol 2017; 7:682. [PMID: 28119694 PMCID: PMC5220330 DOI: 10.3389/fimmu.2016.00682] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 12/21/2016] [Indexed: 12/20/2022] Open
Abstract
There is compelling evidence that inflammation contributes to tumorigenesis. Inflammatory mediators within the tumor microenvironment can either promote an antitumor immune response or support tumor pathogenesis. Therefore, it is critical to determine the relative contribution of tumor-associated inflammatory pathways to cancer development. Interleukin-33 (IL-33) is a member of the IL-1 family of cytokines that is released upon tissue stress or damage to operate as an alarmin. IL-33 has been primarily implicated in the induction of type-2 immune responses. However, recent findings have shown a role of IL-33 in several cancers where it may exert multiple functions. In this review, we will present the current knowledge on the role of IL-33 in the microenvironment of different tumors. We will highlight which cells produce and which cells are activated by IL-33 in cancer. Furthermore, we will explain how IL-33 modulates the tumor-associated inflammatory microenvironment to restrain or promote tumorigenesis. Finally, we will discuss the issues to be addressed first before potentially targeting the IL-33 pathway for cancer therapy.
Collapse
Affiliation(s)
- Marie-Hélène Wasmer
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Bern , Bern , Switzerland
| |
Collapse
|
144
|
Ryu B, Kim SY, Vo TS, Kim WS, Kim DG, Kim SK. Characterization of the in vitro effects of gallic acid-grafted-chitooligosaccharides in the suppression of AGS human gastric cancer cell proliferation. RSC Adv 2017. [DOI: 10.1039/c7ra02487h] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
G-COS was compared with COS for its influence on the proliferation of AGS human gastric cancer cells, showing an increase in the accumulation of cells in the sub-G1 phase and early apoptosis.
Collapse
Affiliation(s)
- BoMi Ryu
- Department of Marine Life Science
- Jeju National University
- Republic of Korea
| | - So-Yeon Kim
- Marine Bioprocess Research Center
- Pukyong National University
- Busan 608-739
- Republic of Korea
| | - Thanh-Sang Vo
- NTT Institute of Hi-Technology
- Nguyen Tat Thanh University
- Ho Chi Minh City
- Vietnam
| | - Won-Suk Kim
- Major in Pharaceutical Engineering Division of Bio-Industry
- Silla University
- Busan
- Korea
| | - Dong Gyu Kim
- Specialized Graduate School Science and Technology Convergence
- Department of Marine Bio Convergence Science
- Pukyong National University
- Busan 608-737
- Republic of Korea
| | - Se-Kwon Kim
- Marine Bioprocess Research Center
- Pukyong National University
- Busan 608-739
- Republic of Korea
- Specialized Graduate School Science and Technology Convergence
| |
Collapse
|
145
|
Tan L, Yang Y, Shao Y, Zhang H, Guo J. Plasma lncRNA-GACAT2 is a valuable marker for the screening of gastric cancer. Oncol Lett 2016; 12:4845-4849. [PMID: 28105191 DOI: 10.3892/ol.2016.5297] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 09/16/2016] [Indexed: 12/12/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are crucial in contributing to gastric tumorigenesis and development. However, the diagnostic value of the majority of lncRNAs in gastric cancer (GC) are not clear. The present study investigated the diagnostic value of gastric cancer associated transcript 2 (GACAT2), a lncRNA that is aberrantly expressed in GC tissues. A total of 343 plasma samples from 80 healthy individuals, 29 patients with gastric dysplasia (GD) and 117 paired preoperative and postoperative patients with GC were collected. Plasma GACAT2 levels were subsequently measured by reverse transcription-quantitative polymerase chain reaction. Finally, the associations between plasma GACAT2 levels and various clinicopathological features of patients with GC were assessed. The results demonstrated that plasma GACAT2 levels in preoperative patients with GC were significantly higher than those in the postoperative group (P=0.031). Compared with healthy individuals, plasma GACAT2 levels were significantly increased in patients with GD (P<0.001) and preoperative patients with GC (P=0.040). Moreover, the individual relative changes of plasma GACAT2 expression following surgery were significantly associated with lymphatic metastasis (P=0.034), distal metastasis (P=0.035) and perineural invasion (P=0.039). Therefore, the results of the current study suggest that plasma-based GACAT2 may be developed as a tumor marker to screen and predict the prognosis of GC patients.
Collapse
Affiliation(s)
- Lin Tan
- Department of General Surgery, The Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang 315010, P.R. China
| | - Yunben Yang
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, Zhejiang 315211, P.R. China
| | - Yongfu Shao
- Department of General Surgery, The Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang 315010, P.R. China
| | - Haiqiang Zhang
- Department of General Surgery, The Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang 315010, P.R. China
| | - Junming Guo
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Ningbo University School of Medicine, Ningbo, Zhejiang 315211, P.R. China
| |
Collapse
|
146
|
Wang Y, Zeng J, Pan J, Geng X, Liu Y, Wu J, Song P, Wang Y, Jia J, Wang L. MicroRNA-200c is involved in proliferation of gastric cancer by directly repressing p27 Kip1. Biochem Biophys Rep 2016; 8:227-233. [PMID: 28955960 PMCID: PMC5613965 DOI: 10.1016/j.bbrep.2016.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 08/10/2016] [Accepted: 09/06/2016] [Indexed: 12/21/2022] Open
Abstract
P27Kip1, also known as Cyclin-dependent kinase inhibitor 1B, is an important check-point protein in the cell cycle. It has been identified that although as a tumor suppressor, P27Kip1 is expressed in different cancer cell types, which shows the therapeutic potential in tumor genesis. In this study, we examined the upstream regulatory mechanism of P27Kip1 at the microRNA (miRNA) level in gastric carcinogenesis. We used bioinformatics to predict that microRNA-200c (miR-200c) might be a direct upstream regulator of P27Kip1. It was also verified in gastric epithelial-derived cell lines that overexpression of miR-200c significantly inhibited the expression levels of P27Kip1, whereas knockdown of miR-200c promoted P27Kip1 expression in AGS and BGC-823 cells. Furthermore, we identified the direct binding of miR-200c on the P27Kip1 3′ -UTR sequence by luciferase assay. MiR-200c could enhance the colony formation of cells by repressing P27Kip1 expression. In addition, the negative correlation between P27Kip1 and miR-200c in human gastric cancer tissues and matched normal tissues further supported the tumor-promoting action of miR-200c in vivo. Our finding suggested that miR-200c directly regulates the expression of P27Kip1 and promotes cell growth in gastric cancer as an oncogene, which may provide new clues to treatment. miR-200c is involved in the proliferation of gastric cancer cell lines. P27Kip1 is a direct downstream target of miR-200c. miR-200c is determined an oncogene in human gastric cancer tissue species.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Pharmocology, Shandong University School of Medicine, Jinan 250012, PR China
| | - Jiping Zeng
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan 250012, PR China
| | - Jianyong Pan
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Xue Geng
- Department of Pharmocology, Shandong University School of Medicine, Jinan 250012, PR China
| | - Yansong Liu
- Shandong Tumor's Hospital and Institute, Jinan 250117, PR China
| | - Jing Wu
- Department of Pharmocology, Shandong University School of Medicine, Jinan 250012, PR China
| | - Ping Song
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan 250012, PR China
| | - Ying Wang
- Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan 250012, PR China
| | - Jihui Jia
- Department of Microbiology/Key Laboratory for Experimental Teratology of Chinese Ministry of Education, Shandong University School of Medicine, Jinan 250012, PR China
| | - Lixiang Wang
- Department of Pharmocology, Shandong University School of Medicine, Jinan 250012, PR China
| |
Collapse
|
147
|
Kim SM. Cellular and Molecular Mechanisms of 3,3'-Diindolylmethane in Gastrointestinal Cancer. Int J Mol Sci 2016; 17:ijms17071155. [PMID: 27447608 PMCID: PMC4964527 DOI: 10.3390/ijms17071155] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 12/18/2022] Open
Abstract
Studies in humans have shown that 3,3′-diindolylmethane (DIM), which is found in cruciferous vegetables, such as cabbage and broccoli, is effective in the attenuation of gastrointestinal cancers. This review presents the latest findings on the use, targets, and modes of action of DIM for the treatment of human gastrointestinal cancers. DIM acts upon several cellular and molecular processes in gastrointestinal cancer cells, including apoptosis, autophagy, invasion, cell cycle regulation, metastasis, angiogenesis, and endoplasmic reticulum (ER) stress. In addition, DIM increases the efficacy of other drugs or therapeutic chemicals when used in combinatorial treatment for gastrointestinal cancer. The studies to date offer strong evidence to support the use of DIM as an anticancer and therapeutic agent for gastrointestinal cancer. Therefore, this review provides a comprehensive understanding of the preventive and therapeutic properties of DIM in addition to its different perspective on the safety of DIM in clinical applications for the treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Soo Mi Kim
- Department of Physiology, Chonbuk National University Medical School, Jeonju 561-180, Korea.
| |
Collapse
|
148
|
Song XL, Kang HK, Jeong GW, Ahn KY, Jeong YY, Kang YJ, Cho HJ, Moon CM. Intravoxel incoherent motion diffusion-weighted imaging for monitoring chemotherapeutic efficacy in gastric cancer. World J Gastroenterol 2016; 22:5520-5531. [PMID: 27350730 PMCID: PMC4917612 DOI: 10.3748/wjg.v22.i24.5520] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/12/2016] [Accepted: 04/20/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To assess intravoxel incoherent motion diffusion-weighted imaging (IVIM-DWI) for monitoring early efficacy of chemotherapy in a human gastric cancer mouse model.
METHODS: IVIM-DWI was performed with 12 b-values (0-800 s/mm2) in 25 human gastric cancer-bearing nude mice at baseline (day 0), and then they were randomly divided into control and 1-, 3-, 5- and 7-d treatment groups (n = 5 per group). The control group underwent longitudinal MRI scans at days 1, 3, 5 and 7, and the treatment groups underwent subsequent MRI scans after a specified 5-fluorouracil/calcium folinate treatment. Together with tumor volumes (TV), the apparent diffusion coefficient (ADC) and IVIM parameters [true water molecular diffusion coefficient (D), perfusion fraction (f) and pseudo-related diffusion coefficient (D*)] were measured. The differences in those parameters from baseline to each measurement (ΔTV%, ΔADC%, ΔD%, Δf% and ΔD*%) were calculated. After image acquisition, tumor necrosis, microvessel density (MVD) and cellular apoptosis were evaluated by hematoxylin-eosin (HE), CD31 and terminal-deoxynucleotidyl transferase mediated nick end labeling (TUNEL) staining respectively, to confirm the imaging findings. Mann-Whitney test and Spearman's correlation coefficient analysis were performed.
RESULTS: The observed relative volume increase (ΔTV%) in the treatment group were significantly smaller than those in the control group at day 5 (ΔTVtreatment% = 19.63% ± 3.01% and ΔTVcontrol% = 83.60% ± 14.87%, P = 0.008) and day 7 (ΔTVtreatment% = 29.07% ± 10.01% and ΔTVcontrol% = 177.06% ± 63.00%, P = 0.008). The difference in ΔTV% between the treatment and the control groups was not significant at days 1 and 3 after a short duration of treatment. Increases in ADC in the treatment group (ΔADC%treatment, median, 30.10% ± 18.32%, 36.11% ± 21.82%, 45.22% ± 24.36%) were significantly higher compared with the control group (ΔADC%control, median, 4.98% ± 3.39%, 6.26% ± 3.08%, 9.24% ± 6.33%) at days 3, 5 and 7 (P = 0.008, P = 0.016, P = 0.008, respectively). Increases in D in the treatment group (ΔD%treatment, median 17.12% ± 8.20%, 24.16% ± 16.87%, 38.54% ± 19.36%) were higher than those in the control group (ΔD%control, median -0.13% ± 4.23%, 5.89% ± 4.56%, 5.54% ± 4.44%) at days 1, 3, and 5 (P = 0.032, P = 0.008, P = 0.016, respectively). Relative changes in f were significantly lower in the treatment group compared with the control group at days 1, 3, 5 and 7 follow-up (median, -34.13% ± 16.61% vs 1.68% ± 3.40%, P = 0.016; -50.64% ± 6.82% vs 3.01% ± 6.50%, P = 0.008; -49.93% ± 6.05% vs 0.97% ± 4.38%, P = 0.008, and -46.22% ± 7.75% vs 8.14% ± 6.75%, P = 0.008, respectively). D* in the treatment group decreased significantly compared to those in the control group at all time points (median, -32.10% ± 12.22% vs 1.85% ± 5.54%, P = 0.008; -44.14% ± 14.83% vs 2.29% ± 10.38%, P = 0.008; -59.06% ± 19.10% vs 3.86% ± 5.10%, P = 0.008 and -47.20% ± 20.48% vs 7.13% ± 9.88%, P = 0.016, respectively). Furthermore, histopathologic findings showed positive correlations with ADC and D and tumor necrosis (rs = 0.720, P < 0.001; rs = 0.522, P = 0.007, respectively). The cellular apoptosis of the tumor also showed positive correlations with ADC and D (rs = 0.626, P = 0.001; rs = 0.542, P = 0.005, respectively). Perfusion-related parameters (f and D*) were positively correlated to MVD (rs = 0.618, P = 0.001; rs = 0.538, P = 0.006, respectively), and negatively correlated to cellular apoptosis of the tumor (rs = -0.550, P = 0.004; rs = -0.692, P < 0.001, respectively).
CONCLUSION: IVIM-DWI is potentially useful for predicting the early efficacy of chemotherapy in a human gastric cancer mouse model.
Collapse
|
149
|
Comparison of Survival Rates, Tumor Stages, and Localization in between Obese and Nonobese Patients with Gastric Cancer. Gastroenterol Res Pract 2016; 2016:9382750. [PMID: 27418926 PMCID: PMC4932154 DOI: 10.1155/2016/9382750] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 04/27/2016] [Accepted: 05/25/2016] [Indexed: 12/17/2022] Open
Abstract
Purpose. In this study we tried to determine the association between body-mass index (BMI), survival rate, and the stage of tumor at the time of diagnosis in patients with gastric cancer. Methods. A total of 270 gastric cancer patients' hospital records were retrospectively evaluated. Patients were grouped according to their BMI at the time of tumor diagnosis. Tumor stages at admission were compared according to their BMI values. Results. There were no differences in OS among BMI subgroups (p = 0.230). The percent of patients with stage III tumor was significantly higher in nonobese while the percent of stage IV tumor was surprisingly higher in obese patients (p was 0.011 and 0.004, resp.). Percent of patients who did not have any surgical intervention was significantly lower in overweight and obese patients than normal and/or underweight patients. Conclusions. At the time of diagnosis, obese patients had significantly higher percent of stage IV tumor than nonobese patients. Despite of that, there were no differences in survival rates among BMI subgroups. Our study results are consistent with "obesity paradox" in gastric cancer patients. We also did not find any relationship between BMI and localization of gastric tumor.
Collapse
|
150
|
Li K, Guo X, Wang Z, Li X, Bu Y, Bai X, Zheng L, Huang Y. The prognostic roles of ALDH1 isoenzymes in gastric cancer. Onco Targets Ther 2016; 9:3405-14. [PMID: 27354812 PMCID: PMC4907742 DOI: 10.2147/ott.s102314] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Increased aldehyde dehydrogenase 1 (ALDH1) activity has been determined to be present in the stem cells of several kinds of cancers including gastric cancer (GC). Nevertheless, which ones of ALDH1's isoenzymes are leading to ALDH1 activity remains elusive. In this study, we examined the prognostic value and hazard ratio (HR) of individual ALDH1 isoenzymes in patients with GC using "The Kaplan-Meier plotter" database. mRNA high expression level of ALDH1A1 was not found to be significantly correlated with the overall survival (OS) of all patients with GC followed for 20 years, HR =0.86 (95% confidence interval [CI]: 0.7-1.05), P=0.13. mRNA high expression level of ALDH1A2 was also not significantly correlated with OS for all patients with GC, HR =1.13 (95% CI: 0.91-1.41), P=0.25. mRNA high expression level of ALDH1A3 was found to be significantly correlated with worsened OS in either intestinal-type patients, HR =2.24 (95% CI: 1.44-3.49), P=0.00026, or diffuse-type patients, HR =1.91 (95% CI: 1.02-3.59), P=0.04. Interestingly, mRNA high expression level of ALDH1B1 was found to be significantly correlated with better OS for all patients with GC, HR =0.66 (95% CI: 0.53-0.81), P=7.8e-05, and mRNA high expression level of ALDH1L1 was found to be significantly correlated with worsened OS for all patients with GC, HR =1.23 (95% CI: 1-1.51), P=0.048. Furthermore, our results also indicate that ALDH1A3 and ALDH1L1 are potential major contributors to the ALDH1 activity in GC, since mRNA high expression levels of ALDH1A3 and ALDH1L1 were found to be significantly correlated with worsened OS for all patients with GC. Based on our study, ALDH1A3 and ALDH1L1 are potential prognostic markers and therapeutic targets for patients with GC.
Collapse
Affiliation(s)
- Kai Li
- Hepatobiliary Treatment Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China; Department of Medical Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, People's Republic of China
| | - Xiaoguang Guo
- Surgical Department, Baotou Cancer Hospital, Baotou, Inner Mongolia, People's Republic of China
| | - Ziwei Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xiaofeng Li
- Department of Medical Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, People's Republic of China
| | - Youquan Bu
- Department of Biology, Chongqing Medical University, Chongqing, People's Republic of China
| | - Xuefeng Bai
- Department of Pathology, Baotou Cancer Hospital, Baotou, Inner Mongolia, People's Republic of China
| | - Liansheng Zheng
- Surgical Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, People's Republic of China
| | - Ying Huang
- Department of Medical Oncology, Baotou Cancer Hospital, Baotou, Inner Mongolia, People's Republic of China
| |
Collapse
|