1
|
Yang HC, Fu CF, Qiao LJ, Long GH, Yang LF, Yao B. Relationship between Helicobacter pylori infection and programmed death-ligand 1 in gastric cancer: A meta-analysis. World J Clin Oncol 2025; 16:102397. [PMID: 40290698 PMCID: PMC12019281 DOI: 10.5306/wjco.v16.i4.102397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/04/2024] [Accepted: 02/06/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) is one of the most common malignancies worldwide, and Helicobacter pylori (HP) infection is a well-established risk factor for its development. Programmed death-ligand 1 (PD-L1) expression is a crucial biomarker for predicting the efficacy of immune checkpoint inhibitors in cancer treatment. While HP infection and PD-L1 expression in GC may be linked, the relationship between them remains unclear, in part because there have been conflicting results reported from various studies. AIM To perform a meta-analysis to assess the relationship between HP and PD-L1 expression in patients with GC. METHODS A systematic literature review was conducted using PubMed, Embase, Cochrane Library, and Web of Science databases. Observational studies that examined the association between HP infection and PD-L1 expression in patients with GC were included. Odds ratios and 95% confidence intervals were calculated to estimate the association. Heterogeneity was assessed using Cochrane's Q test and I² statistic. A random-effects model was used due to significant heterogeneity across studies. RESULTS Fourteen studies involving a total of 3069 patients with GC were included. The pooled analysis showed a significant association between HP infection and increased PD-L1 expression in GC tissues (odd ratio = 1.69, 95% confidence interval: 1.24-2.29, P < 0.001, I 2 = 59%). Sensitivity analyses confirmed the robustness of these findings. Subgroup analyses did not show significant variation based on geographic region, sample size, or method of PD-L1 assessment. Publication bias was minimal, as shown by funnel plots and Egger's regression test. CONCLUSION HP infection is associated with increased PD-L1 expression in GC, suggesting that HP status may influence the response to programmed cell death protein 1/PD-L1 blockade therapy.
Collapse
Affiliation(s)
- Hong-Chang Yang
- Department of Gastroenterology, Longgang Central Hospital of Shenzhen, Shenzhen 518100, Guangdong Province, China
| | - Cheng-Feng Fu
- Department of Oncology, Tongren People’s Hospital, Tongren 554300, Guizhou Province, China
| | - Li-Jun Qiao
- Department of Basic Medical Sciences, Guizhou Health Vocational College, Tongren 554300, Guizhou Province, China
| | - Gen-He Long
- Department of School of Medicine, Guizhou Vocational and Technical College, Tongren 554300, Guizhou Province, China
| | - Li-Fen Yang
- Department of Oncology, Tongren People’s Hospital, Tongren 554300, Guizhou Province, China
| | - Biao Yao
- Department of Oncology, Tongren People’s Hospital, Tongren 554300, Guizhou Province, China
| |
Collapse
|
2
|
Jia Z, Zheng M, Jiang J, Cao D, Wu Y, Zhang Y, Fu Y, Cao X. Positive H. pylori status predicts better prognosis of non-cardiac gastric cancer patients: results from cohort study and meta-analysis. BMC Cancer 2022; 22:155. [PMID: 35135494 PMCID: PMC8822753 DOI: 10.1186/s12885-022-09222-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 01/21/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Previous researches have associated Helicobacter pylori (H. pylori) with a prognosis of gastric cancer (GC), however, without a concert conclusion. This study aimed to study this issue further by a prospective cohort study and a meta-analysis. METHODS Histologically diagnosed gastric cancer (GC) patients were recruited into the primary prospective cohort study between January 2009 to December 2013. All the patients were followed-up periodically to record information on post-surgery therapy and overall survival status. The pre-surgery status of H. pylori was measured by enzyme-linked immunosorbent assay. A meta-analysis was conducted after retrieving related researches in the databases of PubMed and Embase up to April 2020. Pooled hazard ratios (HRs) and 95% confidence intervals (CIs) were summarized to validate the relationship between H. pylori infection and the survival time of GC patients. I2 statistics and Q test were used to assess the heterogeneity. Sensitivity analyses were performed using Galbraith's plot, leave-one-out analysis, subgroup analyses and meta-regression to explore the sources of heterogeneity and the stability of the summary results. RESULTS A total of 743 GC patients with radical tumorectomy were included prospectively and 516 (69.4%) were positive on H. pylori. H. pylori-positive patients tended to survive longer than -negative ones (HR 0.92, 95%CI: 0.74-1.15), though the tendency was not statistically significant. Cohort studies on the prognosis of GC were retrieved comprehensively by assessing the full-text and 59 published studies, together with the result of our study, were included in the further meta-analysis. The summarized results related the positive status of H. pylori to better overall survival (HR 0.81, 95%CI: 0.72-0.90) and disease-free survival (HR 0.83, 95%CI: 0.67-0.99). Results from subgroup analyses indicated that the pooled magnitude of this association was relatively lower in studies not referring to H. pylori in title and abstract. CONCLUSIONS In conclusion, gastric cancer patients with H. pylori have a better prognosis than patients of H. pylori negative. More stringent surveillance strategies may be necessary for patients with H. pylori negative at cancer diagnosis.
Collapse
Affiliation(s)
- Zhifang Jia
- Division of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Min Zheng
- Division of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Jing Jiang
- Division of Clinical Research, The First Hospital of Jilin University, Changchun, China.,Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Donghui Cao
- Division of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Yanhua Wu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Yuzheng Zhang
- Division of Clinical Research, The First Hospital of Jilin University, Changchun, China.,Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, China
| | - Yingli Fu
- Division of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Xueyuan Cao
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
3
|
Li G, Yu S, Xu J, Zhang X, Ye J, Wang Z, He Y. The prognostic role of Helicobacter pylori in gastric cancer patients: A meta-analysis. Clin Res Hepatol Gastroenterol 2019; 43:216-224. [PMID: 30361060 DOI: 10.1016/j.clinre.2018.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/04/2018] [Accepted: 08/21/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND The prognostic value of Helicobacter pylori (H. pylori) infection in gastric cancer patients has been investigated over many years; however, the results remain inconclusive. Thus, we performed a comprehensive review of currently available evidence via a systemic meta-analysis to evaluate the effects of H. pylori infection on the prognosis of gastric cancer patients. METHODS Studies that evaluated the prognostic value of H. pylori infection in gastric cancer were extracted in March 2016 by searching PubMed, EMBASE, and the Cochrane Central Register of Controlled Trials. We obtained or calculated hazard ratios (HRs) and the associated 95% confidence intervals (CIs) from the identified studies, and conducted random-effects model analyses of overall survival and progression-free survival. Twenty-four studies with a cumulative sample size of 7191 patients were included in our analysis. RESULTS Our meta-analysis revealed that H. pylori infection is an indicator of improved overall survival in gastric cancer patients (HR, 0.79; 95% CI, 0.64-0.99); however, this was only true for European patients. The benefits of H. pylori infection were not detected in Asian gastric cancer patients (HR, 1.01; 95% CI, 0.91-1.12) or those in the United States (HR, 0.88; 95% CI, 0.73-1.05). Subgroup analyses revealed that the prognostic significance of H. pylori infection differed with respect to the year of study publication, number of patients, H. pylori detection method, tumor stage, H. pylori-positive rate, and risk of bias. The prognostic value of H. pylori infection on progression-free survival was unclear (HR, 0.84; 95% CI, 0.70-1.01). CONCLUSIONS These data provide limited, moderate-quality evidence that H. pylori infection is an indicator of good prognosis in European gastric cancer patients. However, this is not necessarily true for other populations.
Collapse
Affiliation(s)
- Guanghua Li
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China.
| | - Shuangjin Yu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China
| | - Jianbo Xu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China
| | - Xinhua Zhang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China
| | - Jinning Ye
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China.
| | - Zhao Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China.
| | - Yulong He
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China.
| |
Collapse
|
4
|
Cui L, Shu C, Liu Z, Tong W, Cui M, Wei C, Tang JJ, Liu X, Hai H, Jiang J, He J, Zhang DY, Ye F, Li Y. Serum protein marker panel for predicting preeclampsia. Pregnancy Hypertens 2018; 14:279-285. [PMID: 29395656 DOI: 10.1016/j.preghy.2018.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 01/23/2018] [Accepted: 01/24/2018] [Indexed: 01/05/2023]
Abstract
BACKGROUND Preeclampsia is a multi-system disorder in pregnancy which has no effective treatment. The diagnosis of preeclampsia is based on clinical presentation and routine laboratory tests. OBJECTIVE This study aimed at identifying serum protein markers for diagnosis of preeclampsia and predicting its severe features. STUDY DESIGN In total, 172 pregnant women were enrolled in this study including 110 subjects with preeclampsia and 62 normotensive subjects. Eleven serum proteins (VEGF, sFlt-1, sEndoglin, PlGF, sEGFR, prolactin, PTX3, PAI-1, NGAL, IL-27, COX-2) were assessed using Luminex multiplex immunoassay and ELISA. RESULTS The levels of seven proteins (sFlt-1, VEGF, sEndoglin, sEGFR, PlGF, NGAL, COX-2) correlated with preeclampsia, and 4 proteins (VEGF, sEndoglin, PlGF, sEGFR) were identified as independent factors associated with preeclampsia. The levels of three proteins (sEndoglin, PTX3, sFlt-1) correlated with severe features of preeclampsia, and three variables (serum creatinine, platelet count and sEndoglin) were identified as independent factors in predicting severe features of preeclampsia. CONCLUSIONS A combination of serum protein markers (VEGF, sEndoglin, PlGF, sEGFR) and clinical variables (serum creatinine, platelet count and sEndoglin) could be used as analytical tool in diagnosis of preeclampsia and its severe features, respectively. Serum sEGFR, a novel biomarker in preeclampsia, may be involved in the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Lifeng Cui
- Department of Pathology, College of Basic Medical Sciences of Jilin University, Changchun, Jilin 130021, China
| | - Chang Shu
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Zitao Liu
- New Hope Fertility Center, New York, NY10019, USA
| | - Weihua Tong
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Miao Cui
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Chengguo Wei
- Department of Medicine Bioinformatics Core, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jian Jenny Tang
- Department of Obstetrics, Gynecology and Reproductive Science, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiufen Liu
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Hujing Hai
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jing Jiang
- Division of Clinical Epidemiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jin He
- Department of Obstetrics and Gynecology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - David Y Zhang
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Fei Ye
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yulin Li
- Department of Pathology, College of Basic Medical Sciences of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
5
|
Liu J, Liu F, Shi Y, Tan H, Zhou L. Identification of key miRNAs and genes associated with stomach adenocarcinoma from The Cancer Genome Atlas database. FEBS Open Bio 2018; 8:279-294. [PMID: 29435418 PMCID: PMC5794471 DOI: 10.1002/2211-5463.12365] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/13/2017] [Accepted: 12/06/2017] [Indexed: 12/14/2022] Open
Abstract
Stomach adenocarcinoma (STAD) is the second leading cause of cancer death and a fuller understanding of its molecular basis is needed to develop new therapeutic targets. miRNA and mRNA data were downloaded from The Cancer Genome Atlas database, and the differentially expressed miRNAs and genes were identified. The target genes of differentially expressed miRNAs were screened by prediction tools. Furthermore, the biological function of these target genes was investigated. Several key miRNAs and their target genes were selected for validation using quantitative real-time polymerase chain reaction (qRT-PCR). The Gene Expression Omnibus (GEO) dataset was used to verify the expression of selected miRNAs and target genes. The diagnostic value of identified miRNAs and genes was accessed by receiver operating characteristic analysis. A total of 1248 differentially expressed genes were identified in STAD. Additionally, nine differentially expressed miRNAs were identified and 160 target genes of these nine miRNAs were identified via target gene detection. Interestingly, they were remarkably enriched in the calcium signaling pathway and bile secretion. qRT-PCR confirmed the expression of several key miRNAs and their target genes. The expression levels of hsa-miR-145-3p, hsa-miR-145-5p, ADAM12,ACAN,HOXC11 and MMP11 in the GEO database were compatible with the bioinformatics results. hsa-miR-139-5p, hsa-miR-145-3p and MMP11 have a potential diagnostic value for STAD. Differential expression of the mature form of miRNAs (hsa-miR-139-5p, hsa-miR-145-3p, hsa-miR-145-5p and hsa-miR-490-3p) and genes including ADAM12,ACAN,HOXC11 and MMP11 and calcium and bile secretion signaling pathways may play important roles in the development of STAD.
Collapse
Affiliation(s)
- Jixi Liu
- Department of GastroenterologyChina‐Japan Friendship HospitalBeijingChina
| | - Fang Liu
- Department of GastroenterologyChina‐Japan Friendship HospitalBeijingChina
| | - Yanfen Shi
- Department of PathologyChina‐Japan Friendship HospitalBeijingChina
| | - Huangying Tan
- Department of Integrative OncologyChina‐Japan Friendship HospitalBeijingChina
| | - Lei Zhou
- Department of General SurgeryChina‐Japan Friendship HospitalBeijingChina
| |
Collapse
|
6
|
Fang X, Liu K, Cai J, Luo F, Yuan F, Chen P. Positive Helicobacter pylori status is associated with better overall survival for gastric cancer patients: evidence from case-cohort studies. Oncotarget 2017; 8:79604-79617. [PMID: 29108340 PMCID: PMC5668073 DOI: 10.18632/oncotarget.18758] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/15/2017] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori (H. pylori) infection increases the gastric cancer risk; however, the influences of H. pylori infection status on the outcomes for gastric cancer patients have not yet clearly defined. Herein, we systematically assessed the epidemiological studies regarding the associations between the H.pylori infection status at diagnosis and the prognosis for gastric cancer patients with the meta-analysis methods. Thirty-three eligibility studies with 8,199 participants that had determined the H.pylori infection status and the outcomes for gastric cancer patients were identified through searching the PubMed and MEDLINE databases updated to March 1st, 2017. The random-effects model suggested that positive H. pylori infection was associated with better overall survival with the pooled hazard ratio (HR) was 0.79 [95% confidence interval (CI) = 0.66-0.93; Q = 134.86, df = 32, P-heterogeneity < 0.001; I2 = 76.3%] compared to negative patients. The association was found to be more prominent in studies with higher quality, longer following-up time and more sensitive detection methods. An inverse but not statistically significant association between the H.pylori status and the disease-free survival of the patients (pooled HR = 0.84, 95% CI = 0.61-1.05;Q = 30.48, df = 11, P-heterogeneity = 0.001; I2 = 63.9%) was found, while no significant association was noticed in any subgroup analyses. These results suggested that gastric cancer patients with positive H.pylori infection status at diagnosis have better overall survival compared to negative; however, more studies are warranted to confirm the results and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Xuqian Fang
- Translational Medicine Research Center, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China.,Department of Pathology, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Kun Liu
- Department of Surgery, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Jialin Cai
- Translational Medicine Research Center, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Fangxiu Luo
- Department of Pathology, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Peizhan Chen
- Translational Medicine Research Center, Ruijin Hospital North, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| |
Collapse
|
7
|
Bernardini G, Figura N, Ponzetto A, Marzocchi B, Santucci A. Application of proteomics to the study of Helicobacter pylori and implications for the clinic. Expert Rev Proteomics 2017; 14:477-490. [PMID: 28513226 DOI: 10.1080/14789450.2017.1331739] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Helicobacter pylori (H. pylori) is a gram-negative bacterium that colonizes the gastric epithelium and mucous layer of more than half the world's population. H. pylori is a primary human pathogen, responsible for the development of chronic gastritis, peptic ulceration and gastric cancer. Proteomics is impacting several aspects of medical research: understanding the molecular basis of infection and disease manifestation, identification of therapeutic targets and discovery of clinically relevant biomarkers. Areas covered: The main aim of the present review is to provide a comprehensive overview of the contribution of proteomics to the study of H. pylori infection pathophysiology. In particular, we focused on the role of the bacterium and its most important virulence factor, CagA, in the progression of gastric cells transformation and cancer progression. We also discussed the proteomic approaches aimed at the investigation of the host response to bacterial infection. Expert commentary: In the field of proteomics of H. pylori, comprehensive analysis of clinically relevant proteins (functional proteomics) rather than entire proteomes will result in important medical outcomes. Finally, we provided an outlook on the potential development of proteomics in H. pylori research.
Collapse
Affiliation(s)
- Giulia Bernardini
- a Dipartimento di Biotecnologie , Chimica e Farmacia, Università degli Studi di Siena , Siena , Italy
| | - Natale Figura
- a Dipartimento di Biotecnologie , Chimica e Farmacia, Università degli Studi di Siena , Siena , Italy
| | - Antonio Ponzetto
- b Dipartimento di Scienze Mediche , Università degli Studi di Torino , Torino , Italy
| | - Barbara Marzocchi
- a Dipartimento di Biotecnologie , Chimica e Farmacia, Università degli Studi di Siena , Siena , Italy
| | - Annalisa Santucci
- a Dipartimento di Biotecnologie , Chimica e Farmacia, Università degli Studi di Siena , Siena , Italy
| |
Collapse
|
8
|
Mohri Y, Toiyama Y, Kusunoki M. Progress and prospects for the discovery of biomarkers for gastric cancer: a focus on proteomics. Expert Rev Proteomics 2016; 13:1131-1139. [PMID: 27744719 DOI: 10.1080/14789450.2016.1249469] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Patient outcomes from gastric cancer vary due to the complexity of stomach carcinogenesis. Recent research using proteomic technologies has targeted components of all of these systems in order to develop biomarkers to aid the early diagnosis of gastric cancer and to assist in prognostic stratification. Areas covered: This review is comprised of evidence obtained from literature searches from PubMed. It covers the evidence of diagnostic, prognostic, and predictive biomarkers for gastric cancer using proteomic technologies, and provides up-to-date references. Expert commentary: The proteomic technologies have not only enabled the screening of a large number of samples, but also enabled the identification of diagnostic, prognostic and predictive biomarkers for gastric cancer. While major challenges still remain, to date, proteomic studies in gastric cancer have provided a wealth of information in revealing proteome alterations associated with the disease.
Collapse
Affiliation(s)
- Yasuhiko Mohri
- a Department of Gastrointestinal and Pediatric Surgery , Mie University Graduate School of Medicine , Mie , Japan
| | - Yuji Toiyama
- a Department of Gastrointestinal and Pediatric Surgery , Mie University Graduate School of Medicine , Mie , Japan
| | - Masato Kusunoki
- a Department of Gastrointestinal and Pediatric Surgery , Mie University Graduate School of Medicine , Mie , Japan
| |
Collapse
|
9
|
Harada K, Mizrak Kaya D, Shimodaira Y, Song S, Baba H, Ajani JA. Proteomics approach to identify biomarkers for upper gastrointestinal cancer. Expert Rev Proteomics 2016; 13:1041-1053. [PMID: 27718753 DOI: 10.1080/14789450.2016.1246189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The prognosis for patients with upper gastrointestinal cancers remains dismal despite the development of multimodality therapies that incorporate surgery, chemotherapy, and radiotherapy. Early diagnosis and personalized treatment should lead to better prognosis. Given the advances in proteomic technologies over the past decades, proteomics promises to be the most effective technique to identify novel diagnostics and therapeutic targets. Areas covered: For this review, keywords were searched in combination with 'proteomics' and 'gastric cancer' or 'esophageal cancer' in PubMed. Studies that evaluated proteomics associated with upper gastrointestinal cancer were identified through reading, with several studies quoted at second hand. We summarize the proteomics involved in upper gastrointestinal cancer and discuss potential biomarkers and therapeutic targets. Expert commentary: In particular, the development of mass spectrometry has enabled detection of multiple proteins and peptides in more biological samples over a shorter time period and at lower cost than was previously possible. In addition, more sophisticated protein databases have allowed a wider variety of proteins in samples to be quantified. Novel biomarkers that have been identified by new proteomic technologies should be applied in a clinical setting.
Collapse
Affiliation(s)
- Kazuto Harada
- a Department of Gastrointestinal Medical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA.,b Department of Gastroenterological Surgery, Graduate School of Medical Science , Kumamoto University , Kumamoto , Japan
| | - Dilsa Mizrak Kaya
- a Department of Gastrointestinal Medical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Yusuke Shimodaira
- a Department of Gastrointestinal Medical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Shumei Song
- a Department of Gastrointestinal Medical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| | - Hideo Baba
- b Department of Gastroenterological Surgery, Graduate School of Medical Science , Kumamoto University , Kumamoto , Japan
| | - Jaffer A Ajani
- a Department of Gastrointestinal Medical Oncology , The University of Texas MD Anderson Cancer Center , Houston , TX , USA
| |
Collapse
|
10
|
Wei S, Wang L, Zhang L, Li B, Li Z, Zhang Q, Wang J, Chen L, Sun G, Li Q, Xu H, Zhang D, Xu Z. ZNF143 enhances metastasis of gastric cancer by promoting the process of EMT through PI3K/AKT signaling pathway. Tumour Biol 2016; 37:12813-12821. [PMID: 27449034 DOI: 10.1007/s13277-016-5239-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/15/2016] [Indexed: 12/27/2022] Open
Abstract
The zinc finger protein 143 (ZNF143) is a transcription factor, which regulates many cell cycle-associated genes. ZNF143 expressed strongly in multiple solid tumors. However, the influence of ZNF143 on gastric cancer (GC) remains largely unknown. In this study, we investigated the ZNF143 mRNA level in GC tissues and cells by quantitative real-time PCR (qRT-PCR). The protein expression of ZNF143 in GC cells, and the signaling pathway proteins were detected by Western blotting. Transwell assay and wound healing assay were performed to explore the effects of ZNF143 for the migration ability of GC cells in vitro. We also performed the tail vein injection in nude mice with GC cells to explore the impact of ZNF143 on GC metastasis in vivo. ZNF143 was overexpressed in specimens of GC compared with adjacent normal tissues and increased more significantly in GC tissues of patients who had lymph node metastasis. Ectopic overexpression of ZNF143 enhanced GC migration, whereas ZNF143 knockdown suppressed this effect in vitro. In vivo, ZNF143 knockdown reduced distant metastasis of GC cells in nude mice. In addition, overexpression of ZNF143 reduced the expression of epithelial cell marker (E-cadherin) and induced the expression of mesenchymal cell marker (N-cadherin,Vimentin), Snail and Slug. We also found that ZNF143 enhanced GC cell migration by promoting the process of EMT through PI3K/AKT signaling pathway. In general, our findings show that ZNF143 expressed strongly in GC and enhanced migration of GC cells in vitro and in vivo. It is conceivable that ZNF143 could be a therapeutic genetic target for GC treatment.
Collapse
Affiliation(s)
- Song Wei
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Linjun Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bowen Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zheng Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qun Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiwei Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Liang Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guangli Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qing Li
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hao Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Diancai Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zekuan Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
11
|
He L, Ye F, Qu L, Wang D, Cui M, Wei C, Xing Y, Lee P, Suo J, Zhang DY. Protein profiling of alpha-fetoprotein producing gastric adenocarcinoma. Oncotarget 2016; 7:28448-28459. [PMID: 27057629 PMCID: PMC5053738 DOI: 10.18632/oncotarget.8571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/28/2016] [Indexed: 12/14/2022] Open
Abstract
Alpha-fetoprotein (AFP) producing gastric adenocarcinoma is considered as a rare subtype of gastric adenocarcinoma. Compared with AFP non-producing gastric adenocarcinoma, our study and other previous studies showed that AFP producing gastric adenocarcinoma is more aggressive and prone to liver metastasis. Using the Protein Pathway Array, 11 of out of 286 proteins tested were found to be differentially expressed between AFP producing (n=32) and AFP non-producing (n=45) gastric adenocarcinoma tissues. In addition, the high level expression of XIAP and IGF-Irβ in gastric adenocarcinoma tissues was independent factors for poor prognosis in AFP producing gastric adenocarcinoma patients. A risk model based on the XIAP and IGF-Irβ expression levels can separate AFP producing gastric adenocarcinoma patients into 2 subgroups and each subgroup had a distinct set of signaling pathways involved. In conclusion, AFP producing gastric adenocarcinoma is a heterogeneous cancer with different clinical outcomes, biological behaviors and underlying molecular alterations.
Collapse
Affiliation(s)
- Liang He
- Department of Gastrointestinal Surgery, First Hospital of Jilin University, Changchun, China
| | - Fei Ye
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Linlin Qu
- Department of Clinical Laboratory, First Hospital of Jilin University, Changchun, China
| | - Daguang Wang
- Department of Gastrointestinal Surgery, First Hospital of Jilin University, Changchun, China
| | - Miao Cui
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chengguo Wei
- Department of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yanpeng Xing
- Department of Gastrointestinal Surgery, First Hospital of Jilin University, Changchun, China
| | - Peng Lee
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Jian Suo
- Department of Gastrointestinal Surgery, First Hospital of Jilin University, Changchun, China
| | - David Y. Zhang
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
12
|
Coghlin C, Murray GI. Progress in the development of protein biomarkers of oesophageal and gastric cancers. Proteomics Clin Appl 2016; 10:532-545. [PMID: 26582241 DOI: 10.1002/prca.201500079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/10/2015] [Accepted: 11/12/2015] [Indexed: 01/03/2025]
Abstract
Upper gastrointestinal cancers originating in the oesophagus and stomach often present late and have a very poor prognosis. Treatment options include surgery for localised disease but, increasingly, neoadjuvant chemotherapy and radiotherapy are being employed to improve outcome. There is often a variable response to neoadjuvant treatment between individual patients and side effects are relatively common. There is an urgent need for novel biomarkers of upper gastrointestinal cancer, not only to improve screening and early diagnosis of the oesophageal and gastric cancers when treatment options are potentially more effective, but also to accurately guide therapy in more advanced disease. The development of predictive biomarkers will also help to more effectively identify those patients that will benefit from targeted therapies. Although many promising results have been derived from these studies there remains a lack of validated clinically applicable biomarkers available for translation into routine clinical use. This review will provide an overview of the recent proteomic research on upper gastrointestinal cancer protein biomarker identification and validation. The challenges faced in the development of validated, clinically acceptable and accurate protein biomarkers will also be discussed, along with possible areas of future progress.
Collapse
Affiliation(s)
- Caroline Coghlin
- Department of Cellular Pathology, Craigavon Area Hospital, Portadown, UK
| | - Graeme I Murray
- Pathology, Division of Applied Medicine, School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
13
|
Chen J, Ge L, Liu A, Yuan Y, Ye J, Zhong J, Liu L, Chen X. Identification of pathways related to FAF1/H. pylori-associated gastric carcinogenesis through an integrated approach based on iTRAQ quantification and literature review. J Proteomics 2016; 131:163-176. [PMID: 26597625 DOI: 10.1016/j.jprot.2015.10.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/18/2015] [Accepted: 10/22/2015] [Indexed: 02/06/2023]
Abstract
UNLABELLED Previously we showed that down-regulation of tumor suppressor FAF1 mRNA, potentially caused by H. pylori, correlated with increasing tumor differentiation and distant metastasis in gastric cancer. To identify molecular details about how FAF1 and H. pylori contribute to gastric carcinogenesis, we used the iTRAQ labeling approach involving LC-MS/MS to perform proteomic analysis of HGC-27 gastric cancer cells stably transfected with an FAF1 transgene and/or infected with H. pylori. Of the 2926 proteins examined, proteomics identified 157 for which the expression was altered as a result of FAF1 expression, 500 with altered expression as a result of H. pylori infection, and 246 with altered expression as a combined result of FAF1 expression and H. pylori infection. A literature review identified 21 proteins as being differentially expressed in H. pylori-associated gastric cancer in at least two studies. These two complementary analyses were combined in Ingenuity Pathway software, which predicted that FAF1/H. pylori-associated gastric carcinogenesis alters primarily biochemical pathways related to mitochondrial dysfunction, oxidative phosphorylation, integrin signaling, cholesterol/leucine metabolism and G2/M checkpoint regulation. Differential expression of key proteins in several of these pathways was validated by immunoblotting in HGC-27 cells. This integrated approach combining proteomics and literature searching may prove fruitful for elucidating how FAF1 expression and H. pylori infection affect gastric carcinogenesis. BIOLOGICAL SIGNIFICANCE We established, for the first time, the proteomics databases of gastric cancer cell HGC-27 overexpressing FAF1 and infected with H. pylori through an integrated approach based on iTRAQ quantification and literature review, this strategy responded to the call for greater focus on data integration in primary/previous proteomic studies; and provided an integrated picture of the reference pathways and networks behind FAF1/H. pylori-associated gastric carcinogenesis, particularly pathways of mitochondrial dysfunction, oxidative phosphorylation, integrin signaling, cholesterol/leucine metabolism and G2/M checkpoint regulation.
Collapse
Affiliation(s)
- Jiawei Chen
- Department of Endoscopy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Lianying Ge
- Department of Endoscopy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Aiqun Liu
- Department of Endoscopy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Yanling Yuan
- Department of Endoscopy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jiaxiang Ye
- Department of Endoscopy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jianhong Zhong
- Department of Hepatobiliary Surgery, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Li Liu
- Department of Endoscopy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Xiaoni Chen
- Department of Endoscopy, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| |
Collapse
|
14
|
Furman D, Davis MM. New approaches to understanding the immune response to vaccination and infection. Vaccine 2015; 33:5271-81. [PMID: 26232539 DOI: 10.1016/j.vaccine.2015.06.117] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 04/26/2015] [Accepted: 06/29/2015] [Indexed: 02/06/2023]
Abstract
The immune system is a network of specialized cell types and tissues that communicates via cytokines and direct contact, to orchestrate specific types of defensive responses. Until recently, we could only study immune responses in a piecemeal, highly focused fashion, on major components like antibodies to the pathogen. But recent advances in technology and in our understanding of the many components of the system, innate and adaptive, have made possible a broader approach, where both the multiple responding cells and cytokines in the blood are measured. This systems immunology approach to a vaccine response or an infection gives us a more holistic picture of the different parts of the immune system that are mobilized and should allow us a much better understanding of the pathways and mechanisms of such responses, as well as to predict vaccine efficacy in different populations well in advance of efficacy studies. Here we summarize the different technologies and methods and discuss how they can inform us about the differences between diseases and vaccines, and how they can greatly accelerate vaccine development.
Collapse
Affiliation(s)
- David Furman
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, United States; Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, United States
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, United States; Department of Microbiology and Immunology, School of Medicine, Stanford University, Stanford, CA, United States; Howard Hughes Medical Institute, School of Medicine, Stanford University, Stanford, CA, United States.
| |
Collapse
|
15
|
Kang W, Tong JHM, Lung RWM, Dong Y, Zhao J, Liang Q, Zhang L, Pan Y, Yang W, Pang JCS, Cheng ASL, Yu J, To KF. Targeting of YAP1 by microRNA-15a and microRNA-16-1 exerts tumor suppressor function in gastric adenocarcinoma. Mol Cancer 2015; 14:52. [PMID: 25743273 PMCID: PMC4342823 DOI: 10.1186/s12943-015-0323-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 02/16/2015] [Indexed: 12/13/2022] Open
Abstract
Background MicroRNAs (miRNAs) have been reported to play an important role in tumorigenesis. In this study, the role of miR-15a and miR-16-1 in gastric adenocarcinoma (GAC) was investigated. Methods The expression of miR-15a and miR-16-1 in cell lines and primary tumors was examined by miRNA qRT-PCR. Proliferative assays, colony formation, cell invasion and migration, flow cytometry analysis and in vivo study were performed by ectopic expression of miR-15a and miR-16-1. The putative target genes of miR-15a and miR-16-1 were explored by TargetScan and further validated. Results We found that miR-15a and miR-16-1 were down-regulated in GAC cell lines and primary tumor samples compared with normal gastric epithelium. Functional study demonstrated that ectopic expression of miR-15a and miR-16-1 suppressed cell proliferation, monolayer colony formation, invasion and migration, and xenograft formation in vivo. In addition, miR-15a and miR-16-1 induced G0/G1 cell cycle arrest which was further confirmed by Western blot and qRT-PCR of related cell cycle regulators. YAP1 was confirmed to be a functional target of miR-15a and miR-16-1 in GAC. YAP1 re-expression partly abrogated the inhibitory effect of miR-15a and miR-16-1 in GAC cells. In clinical samples, YAP1 protein expression shows negative correlation with miR-15a and miR-16-1 expression. Conclusion In conclusion, targeting YAP1 by tumor suppressor miRNA miR-15a and miR-16-1 plays inhibitory effect and this might have a therapeutic potential in GAC. Electronic supplementary material The online version of this article (doi:10.1186/s12943-015-0323-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China.
| | - Joanna H M Tong
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
| | - Raymond W M Lung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
| | - Yujuan Dong
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
| | - Junhong Zhao
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
| | - Qiaoyi Liang
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
| | - Li Zhang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
| | - Yi Pan
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
| | - Weiqin Yang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, PR China.
| | - Jesse C S Pang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, PR China.
| | - Alfred S L Cheng
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China. .,School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, PR China.
| | - Jun Yu
- Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China. .,Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, PR China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Oncology in South China, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Institute of Digestive Disease, Partner State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Li Ka Shing Institute of Health Science, Sir Y.K. Pao Cancer Center, The Chinese University of Hong Kong, Hong Kong, SAR, PR China. .,Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, PR China.
| |
Collapse
|
16
|
Mizrahi I, Mazeh H, Grinbaum R, Beglaibter N, Wilschanski M, Pavlov V, Adileh M, Stojadinovic A, Avital I, Gure AO, Halle D, Nissan A. Colon Cancer Associated Transcript-1 (CCAT1) Expression in Adenocarcinoma of the Stomach. J Cancer 2015; 6:105-10. [PMID: 25561974 PMCID: PMC4280392 DOI: 10.7150/jca.10568] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 10/15/2014] [Indexed: 12/17/2022] Open
Abstract
Background: Long non-coding RNAs (lncRNAs) have been shown to have functional roles in cancer biology and are dys-regulated in many tumors. Colon Cancer Associated Transcript -1 (CCAT1) is a lncRNA, previously shown to be significantly up-regulated in colon cancer. The aim of this study is to determine expression levels of CCAT1 in gastric carcinoma (GC). Methods: Tissue samples were obtained from patients undergoing resection for gastric carcinoma (n=19). For each patient, tumor tissue and normal appearing gastric mucosa were taken. Normal gastric tissues obtained from morbidly obese patients, undergoing laparoscopic sleeve gastrectomy served as normal controls (n=19). A human gastric carcinoma cell line (AGS) served as positive control. RNA was extracted from all tissue samples and CCAT1 expression was analyzed using quantitative real time-PCR (qRT-PCR). Results: Low expression of CCAT1 was identified in normal gastric mucosa samples obtained from morbidly obese patients [mean Relative Quantity (RQ) = 1.95±0.4]. AGS human gastric carcinoma cell line showed an elevated level of CCAT1 expression (RQ=8.02). Expression levels of CCAT1 were approximately 10.8 fold higher in GC samples than in samples taken from the negative control group (RQ=21.1±5 vs. RQ=1.95±0.4, respectively, p<0.001). Interestingly, CCAT1 expression was significantly overexpressed in adjacent normal tissues when compared to the negative control group (RQ = 15.25±2 vs. RQ=1.95±0.4, respectively, p<0.001). Tissues obtained from recurrent GC cases showed the highest expression levels (RQ = 88.8±31; p<0.001). Expression levels increased with tumor stage (T4- 36.4±15, T3- 16.1±6, T2- 4.7±1), however this did not reach statistical significance (p=0.2). There was no difference in CCAT1 expression between intestinal and diffuse type GC (RQ=22.4±7 vs. 22.4±16, respectively, p=0.9). Within the normal gastric tissue samples, no significant difference in CCAT1 expression was observed in helicobacter pylori negative and positive patients (RQ= 2.4±0.9 vs. 0.93±0.2, respectively, p=0.13). Conclusion: CCAT1 is up-regulated in gastric cancer, and may serve as a potential bio-marker for early detection and surveillance.
Collapse
Affiliation(s)
- Ido Mizrahi
- 1. Department of Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Haggi Mazeh
- 1. Department of Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Ronit Grinbaum
- 1. Department of Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Nahum Beglaibter
- 1. Department of Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Michael Wilschanski
- 2. The Surgical Oncology Laboratory, Department of Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Vera Pavlov
- 2. The Surgical Oncology Laboratory, Department of Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Muchamad Adileh
- 3. Department of Surgery, Hadassah-Hebrew University Medical Center, Ein Kerem, Jerusalem, Israel
| | | | | | - Ali Osmay Gure
- 5. Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - David Halle
- 2. The Surgical Oncology Laboratory, Department of Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel
| | - Aviram Nissan
- 2. The Surgical Oncology Laboratory, Department of Surgery, Hadassah-Hebrew University Medical Center, Mount Scopus, Jerusalem, Israel; ; 3. Department of Surgery, Hadassah-Hebrew University Medical Center, Ein Kerem, Jerusalem, Israel
| |
Collapse
|
17
|
Decreased miR-204 in H. pylori-associated gastric cancer promotes cancer cell proliferation and invasion by targeting SOX4. PLoS One 2014; 9:e101457. [PMID: 24984017 PMCID: PMC4077842 DOI: 10.1371/journal.pone.0101457] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 06/05/2014] [Indexed: 12/14/2022] Open
Abstract
Background The molecular mechanism between Helicobacter pylori (H. pylori) infection and gastric cancer remained largely unknown. In this study, we determined the role of miRNA in H. pylori induced gastric cancer. Methods and Results We found that miR-204 was decreased in H. pylori positive tissues by qRT-PCR. Knockdown of miR-204 enhanced the invasion and proliferation ability of gastric cancer cells in vitro. Luciferase assay revealed that SOX4 was target gene of miR-204, which was found up-regulated in H. pylori positive tissues. Down-regulation of miR-204 and over-expression of SOX4 promoted epithelial-mesenchymal transition process. Conclusion Taken together, our findings demonstrated that miR-204 may act as a tumor suppressor in H. pylori induced gastric cancer by targeting SOX4.
Collapse
|