1
|
Li S, Chen A, Gui J, Zhou H, Zhu L, Mi Y. TLN1: an oncogene associated with tumorigenesis and progression. Discov Oncol 2024; 15:716. [PMID: 39589610 PMCID: PMC11599537 DOI: 10.1007/s12672-024-01593-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
Talin-1 (TLN1), encoded by the TLN1 gene, is a focal adhesion-related protein capable of binding various proteins in the cytoskeleton. It is also expressed at high levels in many cancers wherein it influences cellular adhesion and the activation of integrins. TLN1 is also capable of promoting tumor cell invasivity, proliferation, and metastatic progression, in addition to being a relevant biomarker and therapeutic target in certain cancers. The present review offers a comprehensive overview of current knowledge regarding TLN1 with respect to its structural properties, functions, and role in tumor development.
Collapse
Affiliation(s)
- Sixin Li
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Anjie Chen
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Jiandong Gui
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Hangsheng Zhou
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China.
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
2
|
Zhou X, Lin J, Shao Y, Zheng H, Yang Y, Li S, Fan X, Hong H, Mao Z, Xue P, Zhang S, Sun J. Targeting PLCG2 Suppresses Tumor Progression, Orchestrates the Tumor Immune Microenvironment and Potentiates Immune Checkpoint Blockade Therapy for Colorectal Cancer. Int J Biol Sci 2024; 20:5548-5575. [PMID: 39494327 PMCID: PMC11528457 DOI: 10.7150/ijbs.98200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 10/05/2024] [Indexed: 11/05/2024] Open
Abstract
Background: Tumor progression and limited benefits of immune checkpoint blockade (ICB) therapy have been two major challenges in the clinical management of colorectal cancer (CRC). The objective of our research was to explore the role of PLCG2 in CRC progression, tumor microenvironment, and potentiating ICB therapy. Methods: Based on bioinformatics analysis and a prospective clinical observational study, the expression, prognostic significance, and clinical relevance of PLCG2 in CRC were unveiled. The single-cell and spatial transcriptome revealed the role of PLCG2 in shaping the heterogeneity of the CRC tumor microenvironment. The biological function of PLCG2 was validated by in vivo and in vitro experiments. The underlying mechanisms were elucidated by RNA-seq, western blotting, qRT-PCR, and multicolor immunofluorescence. The multiplex immunohistochemistry and flow cytometry were adopted to clarify the immunomodulatory role of PLCG2 in facilitating CRC immune escape. The translational value of targeting PLCG2 to potentiate the efficacy of ICB therapy and synergistic therapy to improve prognosis was explored in the preclinical animal models. Results: In CRC, PLCG2 exhibited high expression levels and was strongly associated with poor prognosis and advanced clinicopathological characteristics of patients. The single-cell transcriptome shed light on its important role in cell communication and the development and differentiation of immune cells. The spatial transcriptome described the spatial distribution of PLCG2 in CRC tissues. Further mechanistic analysis demonstrated that PLCG2 could promote proliferation, invasion, metastasis, epithelial-mesenchymal transition, and cell cycle regulation and inhibit apoptosis of CRC cells via the Akt-mTOR pathway activation. Furthermore, PLCG2 was found to contribute greatly to the immunosuppressive microenvironment and enhanced immune escape as it significantly suppressed the infiltration and functional activation of CD8+ T cells and promoted the infiltration of Treg cells as well as PD-1 and PD-L1 expression. Meanwhile, knockdown of PLCG2 could potentiate the efficacy of ICB therapy. Conclusion: In summary, we have identified for the first time that PLCG2 could be considered a precise biomarker and promising therapeutic target for predicting CRC prognosis, optimizing individualized treatment, reversing CRC immune escape, and overcoming resistance to ICB therapy.
Collapse
Affiliation(s)
- Xueliang Zhou
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Surgery, Wuxi branch of Ruijin Hospital Shanghai Jiao Tong University School of Medicine, Wuxi, China
| | - Joshua Lin
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanfei Shao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huang Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Yang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuchun Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodong Fan
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hiju Hong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihai Mao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei Xue
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Surgery, Wuxi branch of Ruijin Hospital Shanghai Jiao Tong University School of Medicine, Wuxi, China
| | - Sen Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Surgery, Wuxi branch of Ruijin Hospital Shanghai Jiao Tong University School of Medicine, Wuxi, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Surgery, Wuxi branch of Ruijin Hospital Shanghai Jiao Tong University School of Medicine, Wuxi, China
| |
Collapse
|
3
|
Chen Y, Hu N, Liao L, Yu K, Shu XO, Zheng W, Yuan JM, Koh WP, Qiao YL, Fan JH, Dawsey SM, Freedman ND, Taylor PR, Goldstein AM, Abnet CC. ABO genotypes and the risk of esophageal and gastric cancers. BMC Cancer 2021; 21:589. [PMID: 34022824 PMCID: PMC8141232 DOI: 10.1186/s12885-021-08334-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 05/10/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Blood type has been associated with the risk of gastric cancer, but few studies have examined the association with esophageal squamous cell carcinoma (ESCC). METHODS We conducted a case-control study using genotyping data of Chinese individuals, including cases of 2022 ESCC, 1189 gastric cardia adenocarcinoma, 1161 gastric noncardia adenocarcinoma, and 2696 controls. Genetic blood type was imputed using three single nucleotide polymorphisms. We used logistic regression to examine the association between blood type and the risk of each cancer. RESULTS Compared to blood type O, the risk of ESCC was significantly elevated for blood type B and AB, with the highest risk for type AB (OR, 95%CI: 1.34, 1.07-1.67). Analysis of genotype suggested that the association of ESCC was from carrying the B allele. Similarly, blood type was significantly associated with gastric noncardia adenocarcinoma (P < 0.001) with risk significantly elevated in type A (1.37, 1.14-1.65) and AB (1.44, 1.10-1.89) compared to type O. Blood type was not associated with gastric cardia adenocarcinoma (P = 0.13). CONCLUSIONS This study provides novel insights into the association between blood type and the risk of ESCC and restricted previously observed association to only gastric noncardia cancer, providing important evidence to clarify the pattern of association and suggesting mechanisms of action.
Collapse
Affiliation(s)
- Yingxi Chen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr. 6E3280, Rockville, MD, 20850, USA.
| | - Nan Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr. 6E3280, Rockville, MD, 20850, USA
| | - Linda Liao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr. 6E3280, Rockville, MD, 20850, USA
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr. 6E3280, Rockville, MD, 20850, USA
| | - Xiao-Ou Shu
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zheng
- Department of Medicine, Division of Epidemiology, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jian-Min Yuan
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Woon-Puay Koh
- Health Service and Systems Research, Duke-NUS Medical School, Singapore, 169857, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, 117549, Singapore
| | - You-Lin Qiao
- National Cancer Center, National Center for Cancer Clinical Research, The Cancer Institute, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jin-Hu Fan
- National Cancer Center, National Center for Cancer Clinical Research, The Cancer Institute, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Sanford M Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr. 6E3280, Rockville, MD, 20850, USA
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr. 6E3280, Rockville, MD, 20850, USA
| | - Philip R Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr. 6E3280, Rockville, MD, 20850, USA
| | - Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr. 6E3280, Rockville, MD, 20850, USA
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Dr. 6E3280, Rockville, MD, 20850, USA
| |
Collapse
|
4
|
Cao J, Chen Z, Tian C, Yu J, Zhang H, Yang J, Yang W. A Shared Susceptibility Locus in the p53 Gene for both Gastric and Esophageal Cancers in a Northwestern Chinese Population. Genet Test Mol Biomarkers 2020; 24:804-811. [PMID: 33290139 DOI: 10.1089/gtmb.2020.0192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background: Upper gastrointestinal tract cancers are the leading causes of cancer-related deaths in Northwest China and they share many similarities in terms of histological type, risk factors, and genetic variants. We hypothesized that shared common single-nucleotide polymorphisms (SNPs) in the p53 pathway exist between patients with gastric and esophageal cancer (EC) patients. Materials and Methods: A case-control study to examine genetic variants in the p53 pathway was conducted with subjects from a high-incidence area for upper gastrointestinal cancers of China. Multiple logistic regression analyses were used to estimate the association of genotypes with gastric cancer and EC risks. Median survival was estimated by using the Kaplan-Meier method and compared by using the log-rank test. Results: Compared with the rs1042522 Pro allele, the rs1042522 Arg allele was associated with an increased risk of gastric cancer (1.810×) and an increased risk of EC (2.285×). The rs1042522 Arg allele carriers who also smoked or consumed alcohol had a further increased risk for gastric cancer odds ratios (ORsmoking = 2.422, ORdrinking = 5.152) and EC (ORsmoking = 5.310, ORdrinking = 8.359). No association was found between the rs1042522 genotypes and survival (p > 0.05). Conclusion: The p53 rs1042522 arg allele together with tobacco smoking and alcohol drinking, was associated with an increased risk, for gastric cancer and EC, but not the survival among northwestern Chinese patients. These associations warrant confirmatory studies.
Collapse
Affiliation(s)
- Juan Cao
- Key Laboratory of Environmental Health and Chronic Disease Prevention and Control, School of Public Health, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Zhiqiang Chen
- Department of Radiology, the General Hospital, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Chaoyong Tian
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, PLA Air Force Military Medical University, Xi'an, People's Republic of China
| | - Jia Yu
- Key Laboratory of Fertility Preservation and Maintenance (Ministry of Education), The School of Basic Medicine and General Hospital, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Hongfei Zhang
- Key Laboratory of Fertility Preservation and Maintenance (Ministry of Education), The School of Basic Medicine and General Hospital, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Jingwen Yang
- Key Laboratory of Fertility Preservation and Maintenance (Ministry of Education), The School of Basic Medicine and General Hospital, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Wenjun Yang
- Key Laboratory of Fertility Preservation and Maintenance (Ministry of Education), The School of Basic Medicine and General Hospital, Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| |
Collapse
|
5
|
Wang L, Li C, Tian J, Liu J, Zhao Y, Yi Y, Zhang Y, Han J, Pan C, Liu S, Deng N, Xian Z, Li G, Zhang X, Liang A. Genome-wide transcriptional analysis of Aristolochia manshuriensis induced gastric carcinoma. PHARMACEUTICAL BIOLOGY 2020; 58:98-106. [PMID: 31957525 PMCID: PMC7006638 DOI: 10.1080/13880209.2019.1710219] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Context: Aristolochia manshuriensis Kom (Aristolochiaceae) (AMK) is known for toxicity and mutagenicity.Objective: The tumorigenic role of AMK has yet to be understood.Materials and methods: AMK extracts were extracted from root crude drug. SD (Sprague Dawley) rats underwent gavage with AMK (0.92 g/kg) every other day for 10 (AMK-10) or 20 (AMK-20) weeks. Stomach samples were gathered for histopathological evaluation, microarray and mRNA analysis.Results: The gastric weight to body weight ratio (GW/BW) is 1.7 in the AMK-10 cohort, and 1.8 in AMK-20 cohort compared to control (CTL) cohort. Liver function was damaged in AMK-10 and AMK-20 rats compared to CTL rats. There were no significant changes of CRE (creatinine) in AMK-10 and AMK-20 rats. Histopathological analysis revealed that rats developed dysplasia in the forestomach in AMK-10 rats, and became gastric carcinoma in AMK-20 rats. Genes including Mapk13, Nme1, Gsta4, Gstm1, Jun, Mgst2, Ggt6, Gpx2, Gpx8, Calml3, Rasgrp2, Cd44, Gsr, Dgkb, Rras, and Amt were found to be critical in AMK-10 and AMK-20 rats. Pik3cb, Plcb3, Tp53, Hras, Myc, Src, Akt1, Gnai3, and Fgfr3 worked in AMK-10 rats, and PDE2a and PDE3a played a pivotal role in AMK-20 rats.Discussion and conclusions: AMK induced benign or malignant gastric tumours depends on the period of AMK administration. Genes including Mapk13, Nme1, Gsta4, Gstm1, Jun, Mgst2, Ggt6, Gpx2, Gpx8, Calml3, Rasgrp2, Cd44, Gsr, Dgkb, Rras, Amt, Pik3cb, Plcb3, Tp53, Hras, Myc, Src, Akt1, Gnai3, Fgfr3, PDE2a, and PDE3a were found to be critical in aristolochic acid-induced gastric tumour process.
Collapse
Affiliation(s)
- Lianmei Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Chunying Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Jingzhuo Tian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Jing Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Yong Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Yan Yi
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Yushi Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Jiayin Han
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Chen Pan
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Suyan Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Nuo Deng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Zhong Xian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Guiqin Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| | - Xin Zhang
- Blood Products Engineering Research and Development Center, Shenzhen, China
| | - Aihua Liang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
- CONTACT Aihua Liang Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Beijing, China
| |
Collapse
|
6
|
Huang H, Xue Q, Du X, Cui J, Wang J, Cheng D, Li J, Zheng Y, Huang G, Zhang K, Liu K, Lu J, Zhao J, Chen X, Dong Z, Li X. p21-activated kinase 4 promotes the progression of esophageal squamous cell carcinoma by targeting LASP1. Mol Carcinog 2020; 60:38-50. [PMID: 33289209 PMCID: PMC7756368 DOI: 10.1002/mc.23269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 08/25/2020] [Accepted: 11/19/2020] [Indexed: 12/30/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common malignant tumors of the digestive tract in humans. Several studies have indicated that PAK4 is associated with the risk of ESCC and may be a potential druggable kinase for ESCC treatment. However, the underlying mechanism remains largely unknown. The aim of our study is to identify the functional role of PAK4 in ESCC. To determine the expression of PAK4 in ESCC, Western blot analysis and immunohistochemistry were performed, and the results showed that PAK4 is significantly upregulated in ESCC tissues and cell lines compared with normal controls and normal esophageal epithelial cell line. To further investigate the role of PAK4 in ESCC, cell viability assays, anchorage-independent cell growth assays, wound healing assays, cellular invasion assays, in vivo xenograft mouse models, and metastasis assays were conducted, and the results showed that PAK4 can significantly facilitate ESCC proliferation and metastasis in vitro and in vivo. To determine the potential target of PAK4 in ESCC progression, a pull-down assay was performed, and the results showed that LASP1 may be a potential target of PAK4. An immunoprecipitation assay and confocal microscopy analysis confirmed that PAK4 can bind to and colocalize with LASP1 in vitro and in cells. Notably, rescue experiments further illustrated the mechanistic network of PAK4/LASP1. Our research reveals the oncogenic roles of PAK4 in ESCC and preliminarily elucidates the mechanistic network of PAK4/LASP1 in ESCC.
Collapse
Affiliation(s)
- Hui Huang
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Qianqian Xue
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Department of Public HealthNanshi Hospital of NanyangNanyangHenanChina
| | - Xiaoge Du
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
- Department of NursingHenan Health School of Medicine and PharmacyPingdingshanHenanChina
| | - Jie Cui
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Jing Wang
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
| | - Dan Cheng
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
| | - Jiaqiong Li
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
| | - Yaqiu Zheng
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
| | - Guojing Huang
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
| | - Keke Zhang
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
- Collaborative Innovation Center of Henan Province for Cancer ChemopreventionZhengzhouHenanChina
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou UniversityZhengzhouHenanChina
| | - Jing Lu
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Collaborative Innovation Center of Henan Province for Cancer ChemopreventionZhengzhouHenanChina
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou UniversityZhengzhouHenanChina
| | - Jimin Zhao
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Collaborative Innovation Center of Henan Province for Cancer ChemopreventionZhengzhouHenanChina
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou UniversityZhengzhouHenanChina
| | - Xinhuan Chen
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Collaborative Innovation Center of Henan Province for Cancer ChemopreventionZhengzhouHenanChina
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou UniversityZhengzhouHenanChina
| | - Ziming Dong
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Collaborative Innovation Center of Henan Province for Cancer ChemopreventionZhengzhouHenanChina
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou UniversityZhengzhouHenanChina
| | - Xiang Li
- Department of Pathophysiology, School of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
- Collaborative Innovation Center of Henan Province for Cancer ChemopreventionZhengzhouHenanChina
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
7
|
Long Noncoding RNA Lnc-TLN2-4:1 Suppresses Gastric Cancer Metastasis and Is Associated with Patient Survival. JOURNAL OF ONCOLOGY 2020; 2020:8681361. [PMID: 32256587 PMCID: PMC7086451 DOI: 10.1155/2020/8681361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 01/30/2020] [Accepted: 02/08/2020] [Indexed: 01/23/2023]
Abstract
Gastric cancer (GC) is one of the most common malignancies worldwide, and the tumor metastasis leads to poor outcomes of GC patients. Long noncoding RNAs (lncRNAs) have emerged as new regulatory molecules that play a crucial role in tumor metastasis. However, the biological function and underlying mechanism of numerous lncRNAs in GC metastasis remain largely unclear. Here, we report a novel lncRNA, lnc-TLN2-4:1, whose expression is decreased in GC tissue versus matched normal tissue, and its low expression is involved in the lymph node and distant metastases of GC, as well as poor overall survival rates of GC patients. We further found that lnc-TLN2-4:1 inhibits the ability of GC cells to migrate and invade but does not influence GC cell proliferation and confirmed that lnc-TLN2-4:1 is mainly located in the cytoplasm of GC cells. We then found that lnc-TLN2-4:1 increases the mRNA and protein expression of TLN2 in GC cells and there is a positive correlation between the expression of lnc-TLN2-4:1 and TLN2 mRNA in GC tissue. Collectively, we identified a novel lncRNA, lnc-TLN2-4:1, in GC, where lnc-TLN2-4:1 represses cell migration and invasion. The low expression of lnc-TLN2-4:1 is associated with poor overall survival rates of GC patients. These suggest that lnc-TLN2-4:1 may be a tumor suppressor during GC metastasis.
Collapse
|
8
|
MAPK and ERK polymorphisms are associated with PCOS risk in Chinese women. Oncotarget 2017; 8:100261-100268. [PMID: 29245975 PMCID: PMC5725017 DOI: 10.18632/oncotarget.22153] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 10/02/2017] [Indexed: 12/24/2022] Open
Abstract
In this case-control study, we analyzed the association between eight RegulomeDB-annotated single nucleotide polymorphisms (SNPs) in the MEK1, MEK2, ERK1 and ERK2 genes and polycystic ovarian syndrome (PCOS). Logistic regression analysis demonstrated that MEK1 rs12050732 (OR = 1.29 [95%CI: 1.06-1.58], P = 0.012), ERK2 rs2266966 (OR = 0.81 [95%CI: 0.67-0.99], P = 0.040) and ERK2 rs5999521 (OR = 0.66 [95%CI: 0.51-0.86], P = 0.002) were associated with PCOS risk without adjusting for age and body mass index. Moreover, PCOS risk increased with allele dosage when these three polymorphisms were combined (Ptrend = 0.001). These findings suggest that genetic variants in key MAPK and ERK genes contribute to PCOS risk in Chinese women.
Collapse
|
9
|
Zhang B, Feng L, Guo H, Li H, Wang Y, Zhang K, Yu X, Cheng S. Villi-specific Gene Expression Reveals Novel Prognostic Biomarkers in Multiple Human Cancers. J Cancer 2017; 8:2793-2801. [PMID: 28928868 PMCID: PMC5604211 DOI: 10.7150/jca.19787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/07/2017] [Indexed: 11/26/2022] Open
Abstract
Despite many striking connections, the biological similarities between embryonic development and tumorigenesis have not been well explored. Development of the placental villi is a crucial process involving many cellular activities, including immunity, proliferation, and cell adhesion. In this study, we designed a strategy to identify the gene expression pattern of villi development and to explore the corresponding features in tumors. We discovered villi-specific genes that are highly expressed in the villus as opposed to the mature placenta and then measured the expression levels of these genes in tumors. We found large changes in the expression of villi-specific genes in multiple types of cancer. These villi-specific genes showed distinct expression patterns and were primarily involved in three biological processes: immune-related (5), proliferation-related (6), and focal adhesion-related (8); these genes were extracted from the corresponding enriched Gene Ontology (GO) terms. We observed that these genes were also dysregulated at the transcriptional level across several tumor types. Moreover, the expression of these three gene groups was associated with poor prognosis in a subset of tumors. Based on villi-specific gene expression, this correlation study indicated the existence of common gene expression patterns between embryonic development and tumorigenesis. Therefore, a systematic analysis of villi-specific gene aberrations in various tumors could serve as an indicator for identifying novel prognostic biomarkers.
Collapse
Affiliation(s)
- Botao Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Feng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Honglin Guo
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Hongxia Li
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Yuanjing Wang
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital, Capital Medical University, Beijing, 100038, China
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xuexin Yu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Shujun Cheng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| |
Collapse
|
10
|
Hyland PL, Zhang H, Yang Q, Yang HH, Hu N, Lin SW, Su H, Wang L, Wang C, Ding T, Fan JH, Qiao YL, Sung H, Wheeler W, Giffen C, Burdett L, Wang Z, Lee MP, Chanock SJ, Dawsey SM, Freedman ND, Abnet CC, Goldstein AM, Yu K, Taylor PR. Pathway, in silico and tissue-specific expression quantitative analyses of oesophageal squamous cell carcinoma genome-wide association studies data. Int J Epidemiol 2016; 45:206-20. [PMID: 26635288 PMCID: PMC4881832 DOI: 10.1093/ije/dyv294] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Oesophageal cancer is the fourth leading cause of cancer death in China where essentially all cases are histologically oesophageal squamous cell carcinoma (ESCC). Agnostic pathway-based analyses of genome-wide association study (GWAS) data combined with tissue-specific expression quantitative trait loci (eQTL) analysis and publicly available functional data can identify biological pathways and/or genes enriched with functionally-relevant disease-associated variants. METHOD We used the adaptive multilocus joint test to analyse 1827 pathways containing 6060 genes using GWAS data from 1942 ESCC cases and 2111 controls with Chinese ancestry. We examined the function of risk alleles using in silico and eQTL analyses in oesophageal tissues. RESULTS Associations with ESCC risk were observed for 36 pathways predominantly involved in apoptosis, cell cycle regulation and DNA repair and containing known GWAS-associated genes. After excluding genes with previous GWAS signals, candidate pathways (and genes) for ESCC risk included taste transduction (KEGG_hsa04742; TAS2R13, TAS2R42, TAS2R14, TAS2R46,TAS2R50), long-patch base excision repair (Reactome_pid; POLD2) and the metabolics pathway (KEGG_hsa01100; MTAP, GAPDH, DCTD, POLD2, AMDHD1). We identified and validated CASP8 rs13016963 and IDH2 rs11630814 as eQTLs, and CASP8 rs3769823 and IDH2 rs4561444 as the potential functional variants in high-linkage disequilibrium with these single nucleotide polymorphisms (SNPs), respectively. Further, IDH2 mRNA levels were down-regulated in ESCC (tumour:normal-fold change = 0.69, P = .75E-14). CONCLUSION Agnostic pathway-based analyses and integration of multiple types of functional data provide new evidence for the contribution of genes in taste transduction and metabolism to ESCC susceptibility, and for the functionality of both established and new ESCC risk-related SNPs.
Collapse
Affiliation(s)
| | - Han Zhang
- Division of Cancer Epidemiology and Genetics, and
| | - Qi Yang
- Division of Cancer Epidemiology and Genetics, and
| | | | - Nan Hu
- Division of Cancer Epidemiology and Genetics, and
| | - Shih-Wen Lin
- Division of Cancer Epidemiology and Genetics, and
| | - Hua Su
- Division of Cancer Epidemiology and Genetics, and
| | - Lemin Wang
- Division of Cancer Epidemiology and Genetics, and
| | - Chaoyu Wang
- Division of Cancer Epidemiology and Genetics, and
| | - Ti Ding
- Division of Cancer Epidemiology and Genetics, and
| | - Jin-Hu Fan
- Division of Cancer Epidemiology and Genetics, and
| | - You-Lin Qiao
- Division of Cancer Epidemiology and Genetics, and
| | - Hyuna Sung
- Division of Cancer Epidemiology and Genetics, and
| | | | - Carol Giffen
- Division of Cancer Epidemiology and Genetics, and
| | | | - Zhaoming Wang
- Division of Cancer Epidemiology and Genetics, and Division of Cancer Epidemiology and Genetics, and
| | | | | | | | | | | | | | - Kai Yu
- Division of Cancer Epidemiology and Genetics, and
| | | |
Collapse
|
11
|
Farid SG, Morris-Stiff G. "OMICS" technologies and their role in foregut primary malignancies. Curr Probl Surg 2015; 52:409-41. [PMID: 26527526 DOI: 10.1067/j.cpsurg.2015.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 08/03/2015] [Indexed: 12/18/2022]
|
12
|
Tan J, Yu CY, Wang ZH, Chen HY, Guan J, Chen YX, Fang JY. Genetic variants in the inositol phosphate metabolism pathway and risk of different types of cancer. Sci Rep 2015; 5:8473. [PMID: 25683757 PMCID: PMC4329558 DOI: 10.1038/srep08473] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/21/2015] [Indexed: 12/23/2022] Open
Abstract
Members of the inositol phosphate metabolism pathway regulate cell proliferation, migration and phosphatidylinositol-3-kinase (PI3K)/Akt signaling, and are frequently dysregulated in cancer. Whether germline genetic variants in inositol phosphate metabolism pathway are associated with cancer risk remains to be clarified. We examined the association between inositol phosphate metabolism pathway genes and risk of eight types of cancer using data from genome-wide association studies. Logistic regression models were applied to evaluate SNP-level associations. Gene- and pathway-based associations were tested using the permutation-based adaptive rank-truncated product method. The overall inositol phosphate metabolism pathway was significantly associated with risk of lung cancer (P = 2.00 × 10−4), esophageal squamous cell carcinoma (P = 5.70 × 10−3), gastric cancer (P = 3.03 × 10−2) and renal cell carcinoma (P = 1.26 × 10−2), but not with pancreatic cancer (P = 1.40 × 10−1), breast cancer (P = 3.03 × 10−1), prostate cancer (P = 4.51 × 10−1), and bladder cancer (P = 6.30 × 10−1). Our results provide a link between inherited variation in the overall inositol phosphate metabolism pathway and several individual genes and cancer. Further studies will be needed to validate these positive findings, and to explore its mechanisms.
Collapse
Affiliation(s)
- Juan Tan
- State Key Laboratory of Oncogene and Related Genes, Key Laboratory of Gastroenterology &Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institution of Digestive Disease, 145 Middle Shandong Rd, Shanghai 200001, China
| | - Chen-Yang Yu
- State Key Laboratory of Oncogene and Related Genes, Key Laboratory of Gastroenterology &Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institution of Digestive Disease, 145 Middle Shandong Rd, Shanghai 200001, China
| | - Zhen-Hua Wang
- State Key Laboratory of Oncogene and Related Genes, Key Laboratory of Gastroenterology &Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institution of Digestive Disease, 145 Middle Shandong Rd, Shanghai 200001, China
| | - Hao-Yan Chen
- State Key Laboratory of Oncogene and Related Genes, Key Laboratory of Gastroenterology &Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institution of Digestive Disease, 145 Middle Shandong Rd, Shanghai 200001, China
| | - Jian Guan
- Department of Otolaryngology, The Affiliated Sixth People's Hospital, Otolaryngology Institute of Shanghai Jiao Tong University, Shanghai 200233, China
| | - Ying-Xuan Chen
- State Key Laboratory of Oncogene and Related Genes, Key Laboratory of Gastroenterology &Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institution of Digestive Disease, 145 Middle Shandong Rd, Shanghai 200001, China
| | - Jing-Yuan Fang
- State Key Laboratory of Oncogene and Related Genes, Key Laboratory of Gastroenterology &Hepatology, Ministry of Health, Division of Gastroenterology and Hepatology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Cancer Institute, Shanghai Institution of Digestive Disease, 145 Middle Shandong Rd, Shanghai 200001, China
| |
Collapse
|
13
|
Wang AH, Liu Y, Wang B, He YX, Fang YX, Yan YP. Epidemiological studies of esophageal cancer in the era of genome-wide association studies. World J Gastrointest Pathophysiol 2014; 5:335-343. [PMID: 25133033 PMCID: PMC4133530 DOI: 10.4291/wjgp.v5.i3.335] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/17/2014] [Accepted: 06/03/2014] [Indexed: 02/06/2023] Open
Abstract
Esophageal cancer (EC) caused about 395000 deaths in 2010. China has the most cases of EC and EC is the fourth leading cause of cancer death in China. Esophageal squamous cell carcinoma (ESCC) is the predominant histologic type (90%-95%), while the incidence of esophageal adenocarcinoma (EAC) remains extremely low in China. Traditional epidemiological studies have revealed that environmental carcinogens are risk factors for EC. Molecular epidemiological studies revealed that susceptibility to EC is influenced by both environmental and genetic risk factors. Of all the risk factors for EC, some are associated with the risk of ESCC and others with the risk of EAC. However, the details and mechanisms of risk factors involved in the process for EC are unclear. The advanced methods and techniques used in human genome studies bring a great opportunity for researchers to explore and identify the details of those risk factors or susceptibility genes involved in the process of EC. Human genome epidemiology is a new branch of epidemiology, which leads the epidemiology study from the molecular epidemiology era to the era of genome wide association studies (GWAS). Here we review the epidemiological studies of EC (especially ESCC) in the era of GWAS, and provide an overview of the general risk factors and those genomic variants (genes, SNPs, miRNAs, proteins) involved in the process of ESCC.
Collapse
|
14
|
Li WQ, Hu N, Burton VH, Yang HH, Su H, Conway CM, Wang L, Wang C, Ding T, Xu Y, Giffen C, Abnet CC, Goldstein AM, Hewitt SM, Taylor PR. PLCE1 mRNA and protein expression and survival of patients with esophageal squamous cell carcinoma and gastric adenocarcinoma. Cancer Epidemiol Biomarkers Prev 2014; 23:1579-1588. [PMID: 24867265 DOI: 10.1158/1055-9965.epi-13-1329] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Germline genetic variants in PLCE1 (10q23) have demonstrated consistent associations with risk of esophageal squamous cell carcinoma (ESCC) and gastric cancer among Chinese. We evaluated PLCE1 mRNA and protein expression in paired tumor-normal tissues, and their relationship with survival. METHODS PLCE1 mRNA was profiled using three probes in the Affymetrix GeneChip U133 for paired tumor-normal tissues of ESCC (n = 132), gastric cardia adenocarcinoma (GCA, n = 62), and gastric noncardia adenocarcinoma (GNCA, n = 72). We used immunohistochemistry to detect PLCE1 protein on slides from tissue microarrays in paired tumor-normal tissues of ESCC (n = 303), and tumors of GCA (n = 298) and GNCA (n = 124). RESULTS Compared with normal tissues, PLCE1 mRNA expression was significantly reduced in ESCC tumors (P = 0.03, probe_205112_at), as well as in GCA and GNCA tumors (P < 0.0001, each probe). Protein expression was nonsignificantly reduced in ESCC tumors (P = 0.51). Increased tumor-normal mRNA fold change (probe_205112_at) was associated with longer survival in ESCC (9.6 months for highest vs. lowest quartile; Ptrend = 0.02). Increased mRNA tumor-normal fold change (probe_205111_at) was associated with longer survival for GCA (10.7 months for highest quartile; Ptrend = 0.04), but not for GNCA cases (P = 0.72). Similar to mRNA, elevated tumor-normal fold change for protein in ESCC was also associated with improved survival (8.1 months for highest quartile; Ptrend = 0.04). CONCLUSIONS Dysregulated PLCE1 mRNA expression was observed for both ESCC (one probe only) and GCA tumors, and the altered PLCE1 expression seems to be associated with cancer prognosis. IMPACT A potential role for PLCE1 in the early detection and/or therapy of ESCC and GCA warrants further investigation.
Collapse
Affiliation(s)
- Wen-Qing Li
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD 20852
| | - Nan Hu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD 20852
| | - Victoria H Burton
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD 20852
| | - Howard H Yang
- Office of the Director, Center for Cancer Research, NCI, Bethesda, MD
| | - Hua Su
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD 20852
| | - Catherine M Conway
- Tissue Array Research Program, Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| | - Lemin Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD 20852
| | - Chaoyu Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD 20852
| | - Ti Ding
- Shanxi Cancer Hospital, Taiyuan, Shanxi, PR China
| | - Yi Xu
- Shanxi Cancer Hospital, Taiyuan, Shanxi, PR China
| | - Carol Giffen
- Information Management Services, Inc., Silver Spring, MD
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD 20852
| | - Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD 20852
| | - Stephen M Hewitt
- Tissue Array Research Program, Laboratory of Pathology, National Cancer Institute, Bethesda, MD
| | - Philip R Taylor
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institute of Health, Bethesda, MD 20852
| |
Collapse
|
15
|
Chiurillo MA. Role of gene polymorphisms in gastric cancer and its precursor lesions: Current knowledge and perspectives in Latin American countries. World J Gastroenterol 2014; 20:4503-4515. [PMID: 24782603 PMCID: PMC4000487 DOI: 10.3748/wjg.v20.i16.4503] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/23/2013] [Accepted: 03/13/2014] [Indexed: 02/07/2023] Open
Abstract
Latin America shows one of the highest incidence rates of gastric cancer in the world, with variations in mortality rates among nations or even within countries belonging to this region. Gastric cancer is the result of a multifactorial complex process, for which a multistep model of carcinogenesis is currently accepted. Additionally to the infection with Helicobacter pylori, that plays a major role, environmental factors as well as genetic susceptibility factors are significant players at different stages in the gastric cancer process. The differences in population origin, demographic structure, socio-economic development, and the impact of globalization lifestyles experienced in Latin America in the last decades, all together offer opportunities for studying in this context the influence of genetic polymorphisms in the susceptibility to gastric cancer. The aim of this article is to discuss current trends on gastric cancer in Latin American countries and to review the available published information about studies of association of gene polymorphisms involved in gastric cancer susceptibility from this region of the world. A total of 40 genes or genomic regions and 69 genetic variants, 58% representing markers involved in inflammatory response, have been used in a number of studies in which predominates a low number of individuals (cases and controls) included. Polymorphisms of IL-1B (-511 C/T, 14 studies; -31 T/C, 10 studies) and IL-1RN (variable number of tandem repeats, 17 studies) are the most represented ones in the reviewed studies. Other genetic variants recently evaluated in large meta-analyses and associated with gastric cancer risk were also analyzed in a few studies [e.g., prostate stem cell antigen (PSCA), CDH1, Survivin]. Further and better analysis centered in gene polymorphisms linked to other covariates, epidemiological studies and the information provided by meta-analyses and genome-wide association studies should help to improve our understanding of gastric cancer etiology in order to develop appropriate health programs in Latin America.
Collapse
|
16
|
Abstract
Infection with Helicobacter pylori is established as the major risk factor for gastric cancer development. Damage of the mucosal barrier due to H. pylori-induced inflammation enhances the carcinogenic effect of other risk factors such as salt intake or tobacco smoking. The genetic disposition of both the bacterial strain and the host can increase the potential towards gastric cancer formation. Genetic variance of the bacterial proteins CagA and VacA is associated with a higher gastric cancer risk, as are polymorphisms and epigenetic changes in host gene coding for interleukins (IL1β, IL8), transcription factors (CDX2, RUNX3) and DNA repair enzymes. Application of high-throughput assays for genome-wide assessment of either genetic structural variance or gene expression patterns may lead to a better understanding of the pathobiological background of these processes, including the underlying signaling pathways. Understanding of the stepwise alterations that take place in the transition from chronic atrophic gastritis, via metaplastic changes, to invasive neoplasia is vital to define the 'point of no return' before which eradication of H. pylori has the potential to prevent gastric cancer. Currently, eradication as preventive strategy is only recommended for high-incidence regions in Asia; large population studies with an adequate follow-up are required to demonstrate the effectiveness of such an approach in Western populations.
Collapse
Affiliation(s)
- Jan Bornschein
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto von Guericke University of Magdeburg, Magdeburg, Germany
| | | |
Collapse
|
17
|
Liu F, Wei WQ, Cormier RT, Zhang ST, Qiao YL, Li XQ, Zhu ST, Zhai YC, Peng XX, Yan YX, Wu LJ, He D, He Y. Association of Single Nucleotide Polymorphisms in the Prostaglandin-endoperoxide Synthase 2 (PTGS2) and Phospholipase A2Group IIA (PLA2G2A) Genes with Susceptibility to Esophageal Squamous Cell Carcinoma. Asian Pac J Cancer Prev 2014; 15:1797-802. [DOI: 10.7314/apjcp.2014.15.4.1797] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
18
|
Relationship between EGF, TGFA, and EGFR Gene Polymorphisms and Traditional Chinese Medicine ZHENG in Gastric Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:731071. [PMID: 24454509 PMCID: PMC3876898 DOI: 10.1155/2013/731071] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 10/23/2013] [Accepted: 11/20/2013] [Indexed: 02/06/2023]
Abstract
In traditional Chinese medicine (TCM), correct syndrome differentiation is the most important principle guiding the prescription of Chinese herbal formulae for the treatment of gastric cancer (GC). We aimed to reveal the genetic mechanisms underlying GC syndrome differentiation (ZHENG) in a population of 387 GC patients. Twenty-nine single nucleotide polymorphisms (SNPs) in EGF, TGFA, and EGFR were investigated. Two SNPs, rs11466285 in TGFA and rs884225 in EGFR, were significantly associated with the distribution of ZHENG (P < 0.05). The rs11466285 TT genotype increased the risk of damp heat with toxin (DHT) and deficiency of both Qi and yin (DQY) compared with obstruction of blood stasis (OBS). The rs884225 AA genotype could increase the risk of DQY and deficiency of both Qi and blood (DQB) compared with yin deficiency due to stomach heat (YDSH). Parallel comparison among the SNPs and syndrome types revealed that DQB was distinct from YDSH, disharmony between the liver and stomach, stagnation of phlegm muddiness (SPM), OBS, and other syndromes at several SNP loci (P < 0.05). The rs11466285 TT and rs884225 AA genotypes exhibit increased risk of DQB compared with OBS and SPM (P < 0.05), respectively. In conclusion, the formation of GC ZHENG was related to EGF, TGFA, and EGFR gene polymorphisms.
Collapse
|