1
|
Wang SH, Hsieh YY, Ong KH, Lai HY, Tsai HH, Sun DP, Huang SKH, Tian YF, Wu HC, Chan TC, Joseph K, Chang IW. The clinicopathological significance and prognostic impact of 14-3-3σ/stratifin expression on patients with surgically resectable intrahepatic cholangiocarcinoma. Asian J Surg 2024:S1015-9584(24)01873-6. [PMID: 39232956 DOI: 10.1016/j.asjsur.2024.08.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024] Open
Abstract
INTRODUCTION Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer after hepatocellular carcinoma. Through data mining of publicly available iCCA transcriptomic datasets from the Gene Expression Omnibus, we identified SFN as the most significantly up-regulated gene in iCCA compared to normal tissue, focusing on the Gene Ontology term "cell proliferation" (GO:0008283). SFN encodes the 14-3-3σ protein, also known as stratifin, which plays crucial roles in various cellular processes. MATERIALS AND METHODS Immunohistochemistry was used to assess stratifin expression in 182 patients with localized iCCAs undergoing surgical resection. Patients were divided into low and high expression groups, and the association between stratifin expression and clinicopathological features was analyzed. Univariate and multivariate survival analyses were performed to assess overall survival (OS), disease-specific survival (DSS), local recurrence-free survival (LRFS), and metastasis-free survival (MeFS). RESULTS Elevated stratifin expression in iCCAs was significantly associated with the absence of hepatitis, positive surgical margins, advanced primary tumor stages, and higher histological grades (all p ≤ 0.011). Survival analyses demonstrated a significant negative association between stratifin expression and all prognostic indicators, including OS, DSS, LRFS, and MeFS (all p ≤ 0.0004). Multivariate analysis revealed that stratifin overexpression was significantly correlated with poorer outcomes in terms of DSS, LRFS, and MeFS (all p < 0.001). CONCLUSIONS These findings suggest that stratifin may play a crucial role in iCCA oncogenesis and tumor progression, serving as a potential novel prognostic biomarker.
Collapse
Affiliation(s)
- Su-Hong Wang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Yao-Yu Hsieh
- Division of Hematology and Oncology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan; Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Khaa Hoo Ong
- Department of Surgery, Division of Gastroenterology and General Surgery, Chi Mei Medical Center, Tainan, Taiwan; Department of Medical Technology, Chung Hwa University of Medical Technology, Tainan, Taiwan; Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hong-Yue Lai
- Department of Pharmacology, School of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Hsin-Hwa Tsai
- Department of Laboratory Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ding-Ping Sun
- Department of Surgery, Division of Gastroenterology and General Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Steven Kuan-Hua Huang
- Department of Surgery, Division of Urology, Chi Mei Medical Center, Tainan, Taiwan; Department of Medical Science Industries, College of Health Sciences, Chang Jung Christian University, Tainan, Taiwan
| | - Yu-Feng Tian
- Department of Surgery, Division of Colon and Rectal Surgery, Chi Mei Medical Center, Tainan, Taiwan
| | - Hung-Chang Wu
- Department of Internal Medicine, Division of Hematology and Oncology, Chi Mei Medical Center, Tainan, Taiwan; College of Pharmacy and Science, Chia Nan University, Tainan, Taiwan
| | - Ti-Chun Chan
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | | | - I-Wei Chang
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Clinical Pathology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Pathology, Taipei Medical University Hospital, Taipei, Taiwan; Department of Pathology, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
2
|
Shahraki K, Shahraki K, Ghasemi Boroumand P, Sheervalilou R. Promotor methylation in ocular surface squamous neoplasia development: epigenetics implications in molecular diagnosis. Expert Rev Mol Diagn 2023; 23:753-769. [PMID: 37493058 DOI: 10.1080/14737159.2023.2240238] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 07/20/2023] [Indexed: 07/27/2023]
Abstract
INTRODUCTION Cancer is heavily influenced by epigenetic mechanisms that include DNA methylation, histone modifications, and non-coding RNA. A considerable proportion of human malignancies are believed to be associated with global DNA hypomethylation, with localized hypermethylation at promoters of certain genes. AREA COVERED The present review aims to emphasize on recent investigations on the epigenetic landscape of ocular surface squamous neoplasia, that could be targeted/explored using novel approaches such as personalized medicine. EXPERT OPINION While the former is thought to contribute to genomic instability, promoter-specific hypermethylation might facilitate tumorigenesis by silencing tumor suppressor genes. Ocular surface squamous neoplasia, the most prevalent type of ocular surface malignancy, is suggested to be affected by epigenetic mechanisms, as well. Although the exact role of epigenetics in ocular surface squamous neoplasia has mostly been unexplored, recent findings have greatly contributed to our understanding regarding this pathology of the eye.
Collapse
Affiliation(s)
- Kourosh Shahraki
- Ocular Tissue Engineering Research Center, Ophthalmic Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Ophthalmology, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Kianoush Shahraki
- Department of Ophthalmology, Zahedan University of Medical Sciences, Zahedan, Iran
- Cornea Department, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Paria Ghasemi Boroumand
- ENT, Head and Neck Research Center and Department, Iran University of Medical Science, Tehran, Iran
| | | |
Collapse
|
3
|
Quantitative Expression of SFN, lncRNA CCDC18-AS1, and lncRNA LINC01343 in Human Breast Cancer as the Regulator Biomarkers in a Novel ceRNA Network: Based on Bioinformatics and Experimental Analyses. Genet Res (Camb) 2022; 2022:6787791. [PMID: 36160032 PMCID: PMC9484965 DOI: 10.1155/2022/6787791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/15/2022] [Accepted: 08/05/2022] [Indexed: 11/21/2022] Open
Abstract
Breast cancer (BC) is one of the leading cancers in the world, which has become an increasing serious problem. In this context, reports demonstrate that some long noncoding RNAs (lncRNAs) play significant regulatory roles in breast tumorigenesis and BC progression via various pathways and act as endogenous RNAs. Finding their dysregulation in cancer and evaluating their interaction with other molecules, such as short noncoding RNAs “microRNA (miRNAs)” as well as various genes, are the most important parts in cancer diagnostics. In this study, after performing GSEA and microarray analysis on the GSE71053 dataset, a new ceRNA network of CCDC18-AS1, LINC01343, hsa-miR4462, and SFN in BC was detected by bioinformatics analysis. Therefore, the expression of SFN, CCDC18-AS1, and LINC01343 was quantitatively measured in 24 BC and normal paired tissues using qRT-PCR. CCDC18-AS1, LINC01343, and SFN were expressed higher in BC than in the control (normal paired) tissues based on qRT-PCR data. Furthermore, a significant positive correlation was observed between CCDC18-AS1 and LINC01343 expression in the samples investigated in this study. The investigation of clinicopathological parameters showed that SFN was highly expressed in tumor size of <5 cm and in nonmenopausal ages, while CCDC18-AS1 and LINC01343 indicated a high expression in stages II-III and III of BC, respectively. The overall survival analysis displayed high and low survival in patients with high expression of SFN and CCDC18-AS1, respectively. The ROC curve analysis disclosed that SFN, CCDC18-AS1, and LINC01343 might be suggested as potential biological markers in BC patients. The high expression of CCDC18-AS1, LINC01343, and SFN in BC samples suggests their potential role in BC tumorigenesis and could be considered hallmarks for the diagnosis and prognosis of BC, although this will require further clinical investigations.
Collapse
|
4
|
Mogal MR, Junayed A, Mahmod MR, Sompa SA, Lima SA, Kar N, TasminaTarin, Khatun M, Zubair MA, Sikder MA. A Computational Approach to Justifying Stratifin as a Candidate Diagnostic and Prognostic Biomarker for Pancreatic Cancer. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1617989. [PMID: 35547358 PMCID: PMC9085308 DOI: 10.1155/2022/1617989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/28/2022] [Accepted: 04/12/2022] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer (PC) is considered a silent killer because it does not show specific symptoms at an early stage. Thus, identifying suitable biomarkers is important to avoid the burden of PC. Stratifin (SFN) encodes the 14-3-3σ protein, which is expressed in a tissue-dependent manner and plays a vital role in cell cycle regulation. Thus, SFN could be a promising therapeutic target for several types of cancer. This study was aimed at investigating, using online bioinformatics tools, whether SFN could be used as a diagnostic and prognostic biomarker in PC. SFN expression was explored by utilizing the ONCOMINE, UALCAN, GEPIA2, and GENT2 tools, which revealed that SFN expression is higher in PC than in normal tissues. The clinicopathological analysis using the ULCAN tool showed that the intensity of SFN expression is commensurate with cancer progression. GEPIA2, R2, and OncoLnc revealed a negative correlation between SFN expression and survival probability in PC patients. The ONCOMINE, UCSC Xena, and GEPIA2 tools showed that cofilin 1 is strongly coexpressed with SFN. Moreover, enrichment and network analyses of SFN were performed using the Enrichr and NetworkAnalyst platforms, respectively. Receiver operating characteristic (ROC) curves revealed that tissue-dependent expression of the SFN gene could serve as a diagnostic and prognostic biomarker. However, further wet laboratory studies are necessary to determine the relevance of SFN expression as a biomarker.
Collapse
Affiliation(s)
- Md Roman Mogal
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Asadullah Junayed
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Rashel Mahmod
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Sagarika Adhikary Sompa
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Suzana Afrin Lima
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Newton Kar
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - TasminaTarin
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Marina Khatun
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Abu Zubair
- Department of Food Technology and Nutritional Science, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Md Asaduzzaman Sikder
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| |
Collapse
|
5
|
Chauhan S, Sen S, Chauhan SS, Pushker N, Tandon R, Kashyap S, Vanathi M, Bajaj MS. Stratifin in ocular surface squamous neoplasia and its association with p53. Acta Ophthalmol 2021; 99:e1483-e1491. [PMID: 33769712 DOI: 10.1111/aos.14844] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 02/23/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Sunlight-induced p53 mutations are known to contribute towards increased risk of ocular surface squamous neoplasia (OSSN). Stratifin (14-3-3σ)/HEM (human epithelial marker) is a p53-mediated inhibitor of cell cycle progression and has been shown to be a target of epigenetic deregulation in various carcinomas. In the present study, Stratifin expression, its promoter methylation status as well as expression of mutant p53 in early and advanced AJCC stages (8th edition) of OSSN, was evaluated. METHODS Sixty-four OSSN [20 conjunctival intraepithelial neoplasia (CIN) and 44 squamous cell carcinoma (SCC)] patients were registered for this study, and they were followed up for 36-58 months (mean 48 ± 3.6). Immunoexpression of Stratifin and mutant p53 protein, mRNA expression of Stratifin by reverse transcription polymerase chain reaction (PCR) and methylation status of Stratifin by methylation-specific PCR, was undertaken. RESULTS Hypermethylation of Stratifin promoter in 63% (40/64), loss of Stratifin expression in 75% (48/64) and downregulation of Stratifin mRNA in 61% (39/64) were observed. Stratifin hypermethylation was significantly associated with reduced disease-free survival in both early and advanced T stage SCC cases. Expression of mutant p53 expression was seen in 48% (31/64) OSSN cases. Of the 31 patients with mutant p53 expression, 87% (27/31) also demonstrated loss of Stratifin immunoexpression. A significant association was seen between mutant p53 expression and Stratifin loss (p = 0.01) in advanced T stage SCC cases. CONCLUSIONS Hypermethylation of Stratifin gene and its reduced mRNA expression both are potential biomarkers for identifying high-risk OSSN patients. Aberrant methylation of Stratifin and simultaneous mutant p53 expression implicates involvement of p53-Stratifin mediated signalling pathway in the pathogenesis of OSSN.
Collapse
Affiliation(s)
- Sheetal Chauhan
- Department of Ocular Pathology Dr. Rajendra Prasad Centre for Ophthalmic Sciences All India Institute of Medical Sciences New Delhi India
| | - Seema Sen
- Department of Ocular Pathology Dr. Rajendra Prasad Centre for Ophthalmic Sciences All India Institute of Medical Sciences New Delhi India
| | - Shyam S. Chauhan
- Department of Biochemistry All India Institute of Medical Sciences New Delhi India
| | - Neelam Pushker
- Ophthalmoplasty Service Dr. Rajendra Prasad Centre for Ophthalmic Sciences All India Institute of Medical Sciences New Delhi India
| | - Radhika Tandon
- Cornea and External Disease, Cataract and Refractive Ocular Oncology and Low Vision Services, Dr. Rajendra Prasad Centre for Ophthalmic Sciences All India Institute of Medical Sciences New Delhi India
| | - Seema Kashyap
- Department of Ocular Pathology Dr. Rajendra Prasad Centre for Ophthalmic Sciences All India Institute of Medical Sciences New Delhi India
| | - Murugesan Vanathi
- Cornea & Ocular Surface Cataract & Refractive Services Dr. Rajendra Prasad Centre for Ophthalmic Sciences All India Institute of Medical Sciences New Delhi India
| | - Mandeep S. Bajaj
- Ophthalmoplasty Service Dr. Rajendra Prasad Centre for Ophthalmic Sciences All India Institute of Medical Sciences New Delhi India
| |
Collapse
|
6
|
Li MX, Sun XM, Cheng WG, Ruan HJ, Liu K, Chen P, Xu HJ, Gao SG, Feng XS, Qi YJ. Using a machine learning approach to identify key prognostic molecules for esophageal squamous cell carcinoma. BMC Cancer 2021; 21:906. [PMID: 34372798 PMCID: PMC8351329 DOI: 10.1186/s12885-021-08647-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 07/19/2021] [Indexed: 01/03/2023] Open
Abstract
Background A plethora of prognostic biomarkers for esophageal squamous cell carcinoma (ESCC) that have hitherto been reported are challenged with low reproducibility due to high molecular heterogeneity of ESCC. The purpose of this study was to identify the optimal biomarkers for ESCC using machine learning algorithms. Methods Biomarkers related to clinical survival, recurrence or therapeutic response of patients with ESCC were determined through literature database searching. Forty-eight biomarkers linked to recurrence or prognosis of ESCC were used to construct a molecular interaction network based on NetBox and then to identify the functional modules. Publicably available mRNA transcriptome data of ESCC downloaded from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) datasets included GSE53625 and TCGA-ESCC. Five machine learning algorithms, including logical regression (LR), support vector machine (SVM), artificial neural network (ANN), random forest (RF) and XGBoost, were used to develop classifiers for prognostic classification for feature selection. The area under ROC curve (AUC) was used to evaluate the performance of the prognostic classifiers. The importances of identified molecules were ranked by their occurrence frequencies in the prognostic classifiers. Kaplan-Meier survival analysis and log-rank test were performed to determine the statistical significance of overall survival. Results A total of 48 clinically proven molecules associated with ESCC progression were used to construct a molecular interaction network with 3 functional modules comprising 17 component molecules. The 131,071 prognostic classifiers using these 17 molecules were built for each machine learning algorithm. Using the occurrence frequencies in the prognostic classifiers with AUCs greater than the mean value of all 131,071 AUCs to rank importances of these 17 molecules, stratifin encoded by SFN was identified as the optimal prognostic biomarker for ESCC, whose performance was further validated in another 2 independent cohorts. Conclusion The occurrence frequencies across various feature selection approaches reflect the degree of clinical importance and stratifin is an optimal prognostic biomarker for ESCC.
Collapse
Affiliation(s)
- Meng-Xiang Li
- School of Information Engineering of Henan University of Science and Technology, 263 Kaiyuan Road, Luolong Qu, Luoyang, 471023, P. R. China.,Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment; Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, 24 Jinghua Road, Jianxi Qu, Luoyang, 471003, P. R. China
| | - Xiao-Meng Sun
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment; Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, 24 Jinghua Road, Jianxi Qu, Luoyang, 471003, P. R. China.,The Sixth People's Hospital of Luoyang, Oncology Department, 14 Xiyuan Road, Jianxi Qu, Luoyang, 471003, P. R. China
| | - Wei-Gang Cheng
- Department of Thyroid and Breast Cancer Surgery, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, 24 Jinghua Road, Jianxi Qu, Luoyang, 471003, P. R. China
| | - Hao-Jie Ruan
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment; Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, 24 Jinghua Road, Jianxi Qu, Luoyang, 471003, P. R. China
| | - Ke Liu
- School of Information Engineering of Henan University of Science and Technology, 263 Kaiyuan Road, Luolong Qu, Luoyang, 471023, P. R. China.,Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment; Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, 24 Jinghua Road, Jianxi Qu, Luoyang, 471003, P. R. China
| | - Pan Chen
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment; Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, 24 Jinghua Road, Jianxi Qu, Luoyang, 471003, P. R. China
| | - Hai-Jun Xu
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment; Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, 24 Jinghua Road, Jianxi Qu, Luoyang, 471003, P. R. China
| | - She-Gan Gao
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment; Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, 24 Jinghua Road, Jianxi Qu, Luoyang, 471003, P. R. China
| | - Xiao-Shan Feng
- School of Information Engineering of Henan University of Science and Technology, 263 Kaiyuan Road, Luolong Qu, Luoyang, 471023, P. R. China. .,Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment; Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, 24 Jinghua Road, Jianxi Qu, Luoyang, 471003, P. R. China.
| | - Yi-Jun Qi
- Henan Key Laboratory of Microbiome and Esophageal Cancer Prevention and Treatment; Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, 24 Jinghua Road, Jianxi Qu, Luoyang, 471003, P. R. China.
| |
Collapse
|
7
|
Shiba-Ishii A. Significance of stratifin in early progression of lung adenocarcinoma and its potential therapeutic relevance. Pathol Int 2021; 71:655-665. [PMID: 34324245 DOI: 10.1111/pin.13147] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/06/2021] [Indexed: 12/21/2022]
Abstract
Lung cancer is the most common cause of global cancer-related mortality, and the main histological type is adenocarcinoma, accounting for 50% of non-small cell lung cancer. In 2015, the World Health Organization (WHO) histological classification defined the concepts of "adenocarcinoma in situ" (AIS) and "minimally invasive adenocarcinoma" (MIA), which are considered to be adenocarcinomas at a very early stage. Although AIS and MIA have a very favorable outcome, once they progress to early but invasive adenocarcinoma (eIA), they can sometimes have a fatal outcome. We previously compared the expression profiles of eIA and AIS, and identified stratifin (SFN; 14-3-3 sigma) as a protein showing significantly higher expression in eIA than in AIS. Expression of SFN is controlled epigenetically by DNA demethylation, and its overexpression is significantly correlated with poorer outcome. In vitro and in vivo analyses have shown that SFN facilitates early progression of adenocarcinoma by enhancing cell proliferation. This review summarizes genetic and epigenetic abnormalities that can occur in early-stage lung adenocarcinoma and introduces recent findings regarding the biological significance of SFN overexpression during the course of lung adenocarcinoma progression. Therapeutic strategies for targeting SFN are also discussed.
Collapse
Affiliation(s)
- Aya Shiba-Ishii
- Department of Diagnostic Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba-shi, Ibaraki, Japan
| |
Collapse
|
8
|
Use of DNA methylation profiling in translational oncology. Semin Cancer Biol 2020; 83:523-535. [PMID: 33352265 DOI: 10.1016/j.semcancer.2020.12.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
DNA methylation is a highly regulated process that has a critical role in human development and homeostatic control of the cell. The number of genes affected by anomalous DNA methylation in cancer-associated pathways is swiftly accelerating and with the advancement of molecular technologies, new layers of complexity are opening up and refining our strategies to combat cancer. DNA methylation profiling is an essential facet to understanding malignant transformation and is becoming an increasingly important tool for cancer diagnosis, prognosis and therapy monitoring. In this review, the role of DNA methylation in normal cellular function is discussed, as well as how epigenetic aberrations override normal cellular cues that lead to tumor initiation and propagation. The review also focuses on the latest advancements in DNA methylation profiling as a biomarker for early cancer detection, predicting patient clinical outcomes and responses to treatment and provides new insights into epigenetic-based therapy in clinical oncology.
Collapse
|
9
|
Bajpai U, Sharma R, Kausar T, Dattagupta S, Chattopadhayay T, Ralhan R. Clinical Significance of 14-3-3 Zeta in Human Esophageal Cancer. Int J Biol Markers 2018; 23:231-7. [DOI: 10.1177/172460080802300406] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We recently found 14-3-3 zeta to be overexpressed in esophageal squamous cell carcinomas (ESCCs) by differential display. In the present study we determined the clinical significance of 14-3-3 zeta in esophageal tumorigenesis. Immunohistochemical analysis was carried out in 61 ESCCs, 33 dysplasia samples, 14 hyperplasia samples and 7 matched histologically normal esophageal tissues and correlated with clinicopathological parameters. Cytoplasmic expression of 14-3-3 zeta protein was observed in 95% of ESCCs; 63% of tumors also showed nuclear localization. All hyperplastic and dysplastic tissues distant from ESCCs as well as dysplastic endoscopic biopsies showed cytoplasmic immunopositivity for 14-3-3 zeta, while nuclear localization was observed in 58% of dysplasia and 36% of hyperplasia samples. Matched distant histologically normal epithelia either showed basal cytoplasmic expression of 14-3-3 zeta or no detectable nuclear expression of the protein. Interestingly, immunopositivity observed in normal esophageal tissues and early hyperplasia was confined to cytoplasm only, though significant nuclear expression was detected in dysplasia and ESCC. Immunoblotting and RT-PCR analyses further confirmed 14-3-3 zeta expression in dysplasia and ESCC. To our knowledge, this is the first report demonstrating overexpression of 14-3-3 zeta in esophageal hyperplasia, dysplasia and squamous cell carcinoma, suggesting that alteration in its expression occurs in early stages and is associated with esophageal tumorigenesis.
Collapse
Affiliation(s)
- U. Bajpai
- Department of Biomedical Sciences, Acharaya Narendra Dev College, University of Delhi South Campus, Delhi
| | - R. Sharma
- School of Biotechnology, Guru Gobind Singh Indraprastha University, Kashmere Gate, Delhi
| | - T. Kausar
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi - India
| | - S. Dattagupta
- Department of Pathology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi - India
| | - T.K. Chattopadhayay
- Department of Gastrointestinal Surgery, All India Institute of Medical Sciences, Ansari Nagar, New Delhi - India
| | - R. Ralhan
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi - India
| |
Collapse
|
10
|
Raungrut P, Petjaroen P, Geater SL, Keeratichananont W, Phukaoloun M, Suwiwat S, Thongsuksai P. Methylation of 14-3-3σ gene and prognostic significance of 14-3-3σ expression in non-small cell lung cancer. Oncol Lett 2017; 14:5257-5264. [PMID: 29113161 PMCID: PMC5662907 DOI: 10.3892/ol.2017.6881] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 07/07/2017] [Indexed: 12/31/2022] Open
Abstract
Loss of 14-3-3σ expression through DNA methylation has been associated with carcinogenesis and the prognosis for various cancer types. Detection of methylation of the gene in serum may be useful for diagnostic utility. The present study aimed to investigate the correlation between 14-3-3σ methylation level in 36 paired tumor tissues of non-small cell lung cancer (NSCLC) and matched serum using methylation-specific polymerase chain reaction. The prognostic significance of 14-3-3σ expression in 167 NSCLC was also evaluated using immunohistochemistry. Methylation of the 14-3-3σ gene was identified in all samples. The methylation level in the serum (mean 87.7%, range 64.6–100%) was higher compared with tumor (mean 46.7%, range 25.3–56.3%). However, no significant correlation between methylation levels in tissues and serums was observed (Spearman's correlation, −0.036; P=0.837). In the 167 tumor tissues, the majority of the cases (83.8%) exhibited negative expression. Adenocarcinoma is more likely to exhibit negative expression (91.4%) compared with squamous cell carcinoma (70.2%). No significant difference was identified in the overall survival according to 14-3-3σ expression status and 14-3-3σ expression did not demonstrated independent prognostic significance. In conclusion, NSCLC harbors certain levels of 14-3-3σ methylation in the tumor and the sera of patients. The clinical value of serum 14-3-3σ methylation should be further elucidated. Immunohistochemical expression 14-3-3σ protein has limited value on prognostic significance.
Collapse
Affiliation(s)
- Pritsana Raungrut
- Department of Biomedical Sciences and The Excellent Research Laboratory of Cancer Molecular Biology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Pingpond Petjaroen
- Department of Biomedical Sciences and The Excellent Research Laboratory of Cancer Molecular Biology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Sarayut Lucien Geater
- Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Warangkana Keeratichananont
- Department of Internal Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Monlika Phukaoloun
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Supaporn Suwiwat
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| | - Paramee Thongsuksai
- Department of Pathology, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand
| |
Collapse
|
11
|
Tang YF, Zhang YB, Feng XD, Lin SH, Qiao N, Sun ZY, Zhou WP. Role of 14-3-3 proteins in human diseases. Shijie Huaren Xiaohua Zazhi 2017; 25:509-520. [DOI: 10.11569/wcjd.v25.i6.509] [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] [Indexed: 02/06/2023] Open
Abstract
14-3-3 proteins are a family of highly conserved small proteins. By interacting with target proteins, 14-3-3 proteins are involved in regulating multiple cellular processes, such as signal transduction, cell cycle regulation, apoptosis, cellular metabolism, cytoskeleton organization and malignant transformation. Mounting evidence suggests that 14-3-3 proteins play an important role in a wide variety of human diseases, such as human cancers and nervous system diseases. This review aims to summarize the current knowledge on the expression, regulation and biological function of 14-3-3 to highlight the role of 14-3-3 proteins in human diseases.
Collapse
|
12
|
Liu Z, Yanagisawa K, Griesing S, Iwai M, Kano K, Hotta N, Kajino T, Suzuki M, Takahashi T. TTF-1/NKX2-1 binds to DDB1 and confers replication stress resistance to lung adenocarcinomas. Oncogene 2017; 36:3740-3748. [PMID: 28192407 DOI: 10.1038/onc.2016.524] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 12/17/2016] [Accepted: 12/21/2016] [Indexed: 01/19/2023]
Abstract
TTF-1, also known as NKX2-1, is a transcription factor that has indispensable roles in both lung development and physiology. We and others have reported that TTF-1 frequently exhibits high expression with increased copy number in lung adenocarcinomas, and also has a role as a lineage-survival oncogene through transcriptional activation of crucial target genes including ROR1 and LMO3. In the present study, we employed a global proteomic search for proteins that interact with TTF-1 in order to provide a more comprehensive picture of this still enigmatic lineage-survival oncogene. Our results unexpectedly revealed a function independent of its transcriptional activity, as TTF-1 was found to interact with DDB1 and block its binding to CHK1, which in turn attenuated ubiquitylation and subsequent degradation of CHK1. Furthermore, TTF-1 overexpression conferred resistance to cellular conditions under DNA replication stress (RS) and prevented an increase in consequential DNA double-strand breaks, as reflected by attenuated induction of pCHK2 and γH2AX. Our findings suggest that the novel non-transcriptional function of TTF-1 identified in this study may contribute to lung adenocarcinoma development by conferring tolerance to DNA RS, which is known to be inherently elicited by activation of various oncogenes.
Collapse
Affiliation(s)
- Z Liu
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - K Yanagisawa
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - S Griesing
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - M Iwai
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - K Kano
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - N Hotta
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - T Kajino
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - M Suzuki
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - T Takahashi
- Division of Molecular Carcinogenesis, Center for Neurological Diseases and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
13
|
Yu CC, Yang ST, Huang WW, Peng SF, Huang AC, Tang NY, Liu HC, Yang MD, Lai KC, Chung JG. Bisdemethoxycurcumin induces DNA damage and inhibits DNA repair associated protein expressions in NCI-H460 human lung cancer cells. ENVIRONMENTAL TOXICOLOGY 2016; 31:1859-1868. [PMID: 26332341 DOI: 10.1002/tox.22187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 07/22/2015] [Accepted: 07/26/2015] [Indexed: 06/05/2023]
Abstract
Nonsmall cell lung carcinoma (NSCLC) is a devastating primary lung tumor resistant to conventional therapies. Bisdemethoxycurcumin (BDMC) is one of curcumin derivate from Turmeric and has been shown to induce NSCLC cell death. Although there is one report to show BDMC induced DNA double strand breaks, however, no available information to show BDMC induced DNA damage action with inhibited DNA repair protein in lung cancer cells in detail. In this study, we tested BDMC-induced DNA damage and condensation in NCI-H460 cells by using Comet assay and DAPI staining examinations, respectively and we found BDMC induced DNA damage and condension. Western blotting was used to examine the effects of BDMC on protein expression associated with DNA damage and repair and results indicated that BDMC suppressed the protein levels associated with DNA damage and repair, such as 14-3-3σ (an important checkpoint keeper of DDR), O6-methylguanine-DNA methyltransferase, DNA repair proteins breast cancer 1, early onset, mediator of DNA damage checkpoint 1 but activate phosphorylated p53 and p-H2A.X (phospho Ser140) in NCI-H460 cells. Confocal laser systems microscopy was used for examining the protein translocation and results show that BDMC increased the translocation of p-p53 and p-H2A.X (phospho Ser140) from cytosol to nuclei in NCI-H460 cells. In conclusion, BDMC induced DNA damage and condension and affect DNA repair proteins in NCI-H460 cells in vitro. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1859-1868, 2016.
Collapse
Affiliation(s)
- Chien-Chih Yu
- School of Pharmacy, China Medical University, Taichung, 404, Taiwan
| | - Su-Tso Yang
- Department of Radiology, China Medical University Hospital, Taichung, 404, Taiwan
- School of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
| | - Wen-Wen Huang
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
| | - Shu-Fen Peng
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
| | - An-Cheng Huang
- Department of Nursing, St. Mary's Junior college of Medicine, Nursing and Management, Yilan, 266, Taiwan
| | - Nou-Ying Tang
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
| | - Hsin-Chung Liu
- Department of Biological Science and Technology, China Medical University, Taichung, 404, Taiwan
| | - Mei-Due Yang
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Kuang-Chi Lai
- School of Medicine, China Medical University Hospital, Taichung, 404, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medicine and Life Science, Chung Hwa University of Medical Technology, Tainan, 717, Taiwan
| | - Jing-Gung Chung
- Graduate Institute of Chinese Medicine, China Medical University, Taichung, 404, Taiwan
- Department of Biotechnology, Asia University, Wu Feng, Taichung, 404, Taiwan
| |
Collapse
|
14
|
Dong N, Shi L, Wang DC, Chen C, Wang X. Role of epigenetics in lung cancer heterogeneity and clinical implication. Semin Cell Dev Biol 2016; 64:18-25. [PMID: 27575638 DOI: 10.1016/j.semcdb.2016.08.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 08/24/2016] [Indexed: 12/15/2022]
Abstract
Lung cancer, as a highly heterogeneous disease, can be initiated and progressed through the interaction between permanent genetic mutations and dynamic epigenetic alterations. However, the mediating mechanisms of epigenetics in cancer heterogeneity remain unclear. The evolution of cancer, the existence of cancer stem cells (CSCs) and the phenomenon of epithelial-mesenchymal transition (EMT) have been reported to be involved in lung cancer heterogeneity. In this review, we briefly recap the definition of heterogeneity and concept of epigenetics, highlight the potential roles and mechanisms of epigenetic regulation in heterogeneity of lung cancer, and summarize the diagnostic and therapeutic implications of epigenetic alterations in lung cancer, especially the role of DNA methylation and histone acetylation. Deep understanding of epigenetic regulation in cancer heterogeneity is instrumental to the design of novel therapeutic approaches that target lung cancer.
Collapse
Affiliation(s)
- Nian Dong
- Department of Pulmonary Medicine, The First affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Lin Shi
- Zhongshan Hospital, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics; Zhongshan Hospital Institute of Clinical Science of Fudan University, Shanghai, China
| | - Diane C Wang
- Department of Pulmonary Medicine, The First affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China
| | - Chengshui Chen
- Department of Pulmonary Medicine, The First affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Xiangdong Wang
- Department of Pulmonary Medicine, The First affiliated Hospital, Wenzhou Medical University, Wenzhou, 325000, China; Zhongshan Hospital, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics; Zhongshan Hospital Institute of Clinical Science of Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Peng C, Jia X, Xiong Y, Yin J, Li N, Deng Y, Luo K, Zhang Q, Wang C, Zhang Z, Zheng G, He Z. The 14-3-3σ/GSK3β/β-catenin/ZEB1 regulatory loop modulates chemo-sensitivity in human tongue cancer. Oncotarget 2016; 6:20177-89. [PMID: 26036631 PMCID: PMC4652996 DOI: 10.18632/oncotarget.3896] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 04/25/2015] [Indexed: 12/18/2022] Open
Abstract
Here we demonstrated that chemotherapy induced 14-3-3σ expression in tongue cancer (TC) cells and overexpressed 14-3-3σ sensitized TC cells to chemotherapy especially in multidrug resistant TC (MDR-TC) cells. In agreement, 14-3-3σ knockdown enhanced resistance of TC cells to chemotherapy. Mechanically, we found 14-3-3σ physically bound to GSK3β in protein level and the binding inhibited β-catenin signaling. Coincidentally, chemotherapy as well as 14-3-3σ overexpression led to increase of GSK3β protein level. Increased GSK3β protein sensitized TC cells to chemotherapy. Moreover, deregulation of 14-3-3σ/GSK3β/β-catenin axis led to overexpressed ZEB1 in TC cells, especially in MDR-TC cells. As a negative feedback loop, ZEB1 bond to 14-3-3σ promoter to enhance promoter hypermethylation in TC cells. Promoter hypermethylation resulted into the decrease of 14-3-3σ expression. Importantly, a positive correlation was observed between 14-3-3σ and GSK3β protein expression in TC tissues from patients receiving chemotherapy. High levels of 14-3-3σ and GSK3β were associated with better prognosis in TC patients.
Collapse
Affiliation(s)
- Cong Peng
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Xiaoting Jia
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Yan Xiong
- Department of Pharmacology, Guangzhou Institute of Snake Venom Research, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China
| | - Jiang Yin
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Nan Li
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Yingen Deng
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Kai Luo
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Qiong Zhang
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Chengkun Wang
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Zhijie Zhang
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Guopei Zheng
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| | - Zhimin He
- Cancer Hospital and Cancer Research Institute of Guangzhou Medical University, Guangzhou 510095, Guangdong, China
| |
Collapse
|
16
|
Poon J, Wessel GM, Yajima M. An unregulated regulator: Vasa expression in the development of somatic cells and in tumorigenesis. Dev Biol 2016; 415:24-32. [PMID: 27179696 PMCID: PMC4902722 DOI: 10.1016/j.ydbio.2016.05.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 02/08/2023]
Abstract
Growing evidence in diverse organisms shows that genes originally thought to function uniquely in the germ line may also function in somatic cells, and in some cases even contribute to tumorigenesis. Here we review the somatic functions of Vasa, one of the most conserved "germ line" factors among metazoans. Vasa expression in somatic cells is tightly regulated and often transient during normal development, and appears to play essential roles in regulation of embryonic cells and regenerative tissues. Its dysregulation, however, is believed to be an important element of tumorigenic cell regulation. In this perspectives paper, we propose how some conserved functions of Vasa may be selected for somatic cell regulation, including its potential impact on efficient and localized translational activities and in some cases on cellular malfunctioning and tumorigenesis.
Collapse
Affiliation(s)
- Jessica Poon
- MCB Department, Brown University, 185 Meeting Street, BOX-GL173, Providence, RI 02912, USA
| | - Gary M Wessel
- MCB Department, Brown University, 185 Meeting Street, BOX-GL173, Providence, RI 02912, USA
| | - Mamiko Yajima
- MCB Department, Brown University, 185 Meeting Street, BOX-GL173, Providence, RI 02912, USA.
| |
Collapse
|
17
|
Raychaudhuri K, Chaudhary N, Gurjar M, D'Souza R, Limzerwala J, Maddika S, Dalal SN. 14-3-3σ Gene Loss Leads to Activation of the Epithelial to Mesenchymal Transition Due to the Stabilization of c-Jun Protein. J Biol Chem 2016; 291:16068-81. [PMID: 27261462 DOI: 10.1074/jbc.m116.723767] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Indexed: 12/21/2022] Open
Abstract
Loss of 14-3-3σ has been observed in multiple tumor types; however, the mechanisms by which 14-3-3σ loss leads to tumor progression are not understood. The experiments in this report demonstrate that loss of 14-3-3σ leads to a decrease in the expression of epithelial markers and an increase in the expression of mesenchymal markers, which is indicative of an induction of the epithelial to mesenchymal transition (EMT). The EMT was accompanied by an increase in migration and invasion in the 14-3-3σ(-/-) cells. 14-3-3σ(-/-) cells show increased stabilization of c-Jun, resulting in an increase in the expression of the EMT transcription factor slug. 14-3-3σ induces the ubiquitination and degradation of c-Jun in an FBW7-dependent manner. c-Jun ubiquitination is dependent on the presence of an intact nuclear export pathway as c-Jun is stabilized and localized to the nucleus in the presence of a nuclear export inhibitor. Furthermore, the absence of 14-3-3σ leads to the nuclear accumulation and stabilization of c-Jun, suggesting that 14-3-3σ regulates the subcellular localization of c-Jun. Our results have identified a novel mechanism by which 14-3-3σ maintains the epithelial phenotype by inhibiting EMT and suggest that this property of 14-3-3σ might contribute to its function as a tumor suppressor gene.
Collapse
Affiliation(s)
- Kumarkrishna Raychaudhuri
- From the KS215, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India
| | - Neelam Chaudhary
- Laboratory of Cell Death and Cell Survival, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally, Hyderabad 500001, India, and Graduate Studies, Manipal University, Manipal, Karnataka 576104, India
| | - Mansa Gurjar
- From the KS215, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India
| | - Roseline D'Souza
- From the KS215, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India
| | - Jazeel Limzerwala
- From the KS215, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India
| | - Subbareddy Maddika
- Laboratory of Cell Death and Cell Survival, Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally, Hyderabad 500001, India, and
| | - Sorab N Dalal
- From the KS215, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai 410210, India,
| |
Collapse
|
18
|
Shi G, Wu J, Wang P, Song M, Sun L, Tu YJ, Huang XD. Expression of RhoGDI2 and 14-3-3σ in gastric carcinoma. Shijie Huaren Xiaohua Zazhi 2015; 23:5282-5289. [DOI: 10.11569/wcjd.v23.i33.5282] [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] [Indexed: 02/06/2023] Open
Abstract
AIM: To detect the expression of RhoGDI2 and 14-3-3σ in gastric cancer, analyze their correlation with clinicopathological characteristics of gastric cancer, and assess their correlation.
METHODS: Immunohistochemistry was used to detect the expression of RhoGDI2 and 14-3-3σ in 152 gastric cancer specimens.
RESULTS: The positive rates of RhoGDI2 and 14-3-3σ in gastric cancer were 76.97% and 27.63%, respectively. The expression of RhoGDI2 and 14-3-3σ was related with tumor differentiation, depth of invasion, lymph node metastasis, distant metastasis and TNM stage in gastric cancer. There was a negative correlation between the expression of RhoGDI2 and 14-3-3σ in gastric cancer.
CONCLUSION: There is a correlation between the expression of RhoGDI2 and 14-3-3σ in gastric cancer. 14-3-3σ is very likely regulated by RhoGDI2 and functions as a downstream target gene of RhoGDI2 in tumor invasion and metastasis. They may be both involved in regulating the invasion and metastasis of gastric cancer.
Collapse
|
19
|
Hou Q, Lin J, Huang W, Li M, Feng J, Mao X. CTRP3 Stimulates Proliferation and Anti-Apoptosis of Prostate Cells through PKC Signaling Pathways. PLoS One 2015. [PMID: 26218761 PMCID: PMC4517796 DOI: 10.1371/journal.pone.0134006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
C1q/TNF-related protein-3 (CTRP3) is a novel adipokine with roles in multiple cellular processes. However, little is known about its function in prostate cells. This study investigated the effects and mechanisms of CTRP3 in prostate cells. We first generated and purified CTRP3 protein in HEK 293T cells. Proliferation of RWPE-1 prostate cells was evaluated by MTT analyses under treatment with different concentrations of CTRP3 for various exposure times. The results revealed maximum enhancement of proliferation with 10 μg/mL CTRP3 for 72 h. Cell apoptosis and cell cycle were determined by TUNEL staining and flow cytometry analysis. TUNEL assay showed decreased TUNEL-positive cells in RWPE-1 prostate cells treated with CTRP3, and flow cytometry showed significantly decreased apoptotic cells upon CTRP3 treatment (treated cells, 8.34±1.175 vs. controls, 20.163±0.35) (P < 0.01). Moreover, flow cytometry analysis also showed a significant decrease of cells in the G1 phase and an increase of cells in the S and G2 phase upon CTRP3 treatment (treated cells, 42.85±1.40 vs. control, 52.77±0.90; 28.41±0.57 vs. 23.49±1.13; 27.08±1.97 vs. 22.20±1.32, respectively) (all P < 0.05). Two-dimensional gel electrophoresis and mass spectrometry identified differentially expressed proteins, including cytokeratin-19, GLRX3 and DDAH1, which were upregulated in CTRP3 treated cells, and cytokeratin-17 and 14-3-3 sigma, which were downregulated. GLRX3, DDAH1 and 14-3-3 sigma were confirmed using western blot analysis. A PKC inhibitor, staurosporine, was used to inhibit PKC activity in CTRP3 treated RWPE-1 cells. Staurosporine completely abolished the CTRP3-induced increased phosphorylation of intracellular PKC substrates and CTRP3-stimulated effect by RWPE-1 cells. Our results provide the first evidence for a physiological role of the novel adipokine, CTRP3, in prostate cells. Our findings suggest that CTRP3 could improve proliferation and anti-apoptosis of prostate cells through protein kinase C signaling pathways.
Collapse
Affiliation(s)
- Qi Hou
- Department of Urology, Longgang District Central Hospital, Shenzhen, China
| | - Jinyan Lin
- Health management center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wentao Huang
- Department of Urology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Maoyin Li
- Department of Urology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianhua Feng
- Department of Urology, Longgang District Central Hospital, Shenzhen, China
- * E-mail: (JF); (XM)
| | - Xiangming Mao
- Department of Urology, Peking University Shenzhen Hospital, Shenzhen, China
- * E-mail: (JF); (XM)
| |
Collapse
|
20
|
Noutsios GT, Ghattas P, Bennett S, Floros J. 14-3-3 isoforms bind directly exon B of the 5'-UTR of human surfactant protein A2 mRNA. Am J Physiol Lung Cell Mol Physiol 2015; 309:L147-57. [PMID: 26001776 DOI: 10.1152/ajplung.00088.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/16/2015] [Indexed: 12/26/2022] Open
Abstract
Human surfactant protein (SP) A (SP-A), an innate immunity molecule, is encoded by two genes, SFTPA1 and SFTPA2. The 5'-untranslated splice variant of SP-A2 (ABD), but not SP-A1 (AD), contains exon B (eB). eB is an enhancer for transcription and translation and contains cis-regulatory elements. Specific trans-acting factors, including 14-3-3, bind eB. The 14-3-3 protein family contains seven isoforms that have been found by mass spectrometry in eB electromobility shift assays (Noutsios et al. Am J Physiol Lung Cell Mol Physiol 304: L722-L735, 2013). We used four different approaches to investigate whether 14-3-3 isoforms bind directly to eB. 1) eB RNA pulldown assays showed that 14-3-3 isoforms specifically bind eB. 2) RNA electromobility shift assay complexes were formed using purified 14-3-3 isoforms β, γ, ε, η, σ, and τ, but not isoform ζ, with wild-type eB RNA. 3 and 4) RNA affinity chromatography assays and surface plasmon resonance analysis showed that 14-3-3 isoforms β, γ, ε, η, σ, and τ, but not isoform ζ, specifically and directly bind eB. Inhibition of 14-3-3 isoforms γ, ε, η, and τ/θ with shRNAs in NCI-H441 cells resulted in downregulation of SP-A2 levels but did not affect SP-A1 levels. However, inhibition of 14-3-3 isoform σ was correlated with lower levels of SP-A1 and SP-A2. Inhibition of 14-3-3 isoform ζ/δ, which does not bind eB, had no effect on expression levels of SP-A1 and SP-A2. In conclusion, the 14-3-3 protein family affects differential regulation of SP-A1 and SP-A2 by binding directly to SP-A2 5'-UTR mRNA.
Collapse
Affiliation(s)
- Georgios T Noutsios
- Center for Host Defense, Inflammation, and Lung Disease (CHILD), Research Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, Pennsylvania; and
| | - Paul Ghattas
- Center for Host Defense, Inflammation, and Lung Disease (CHILD), Research Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, Pennsylvania; and
| | - Stephanie Bennett
- Center for Host Defense, Inflammation, and Lung Disease (CHILD), Research Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, Pennsylvania; and
| | - Joanna Floros
- Center for Host Defense, Inflammation, and Lung Disease (CHILD), Research Department of Pediatrics, College of Medicine, The Pennsylvania State University, Hershey, Pennsylvania; and Department of Obstetrics and Gynecology, College of Medicine, The Pennsylvania State University, Hershey, Pennsylvania
| |
Collapse
|
21
|
Sun N, Wu Y, Huang B, Liu Q, Dong Y, Ding J, Liu Y. Decreased expression of 14-3-3 σ, an early event of malignant transformation of respiratory epithelium, also facilitates progression of squamous cell lung cancer. Thorac Cancer 2015; 6:715-21. [PMID: 26557909 PMCID: PMC4632923 DOI: 10.1111/1759-7714.12246] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 01/19/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND It has been shown that 14-3-3 σ serves as a tumor suppressor gene, and is downregulated in various tumor tissues. However, the role of 14-3-3 σ during the initiation and progression of lung squamous cell carcinoma (SqCC) is not well understood. METHODS The expression status of 14-3-3 σ in archival tissue samples from 40 lung SqCC patients (36 with normal bronchia, 19 squamous metaplasia, and 17 dysplasia/carcinoma in situ, in their tissue samples) was examined by immunohistochemical analysis. The proliferation rate and tumor formation ability of the H520 cell transfected with 14-3-3 σ was tested with methyl thiazolyl tetrazolium assay and nude mice subcutaneous injection, respectively. RESULTS In the normal bronchial epithelia, 14-3-3 σ was highly expressed, whereas it was significantly decreased in precancerous and cancerous tissues. Compared with matched invasive cancer tissues, the expression level of 14-3-3 σ in squamous metaplasia was significantly higher (P = 0.049), while that in dysplasia/carcinoma in situ showed no significant changes (P = 0.135). Statistical analysis showed that the expression level of 14-3-3 σ in tumor tissue was associated with the differentiation grade of the tumor (P = 0.001) and the prognosis of the patient (P = 0.003). The overexpression of 14-3-3 σ significantly suppressed the proliferation of H520 cells in vitro and in vivo. CONCLUSION The inactivation of 14-3-3 σ may be a very early event in tumorigenesis and could facilitate the initiation and progression of lung SqCC in a sustainable way.
Collapse
Affiliation(s)
- Nan Sun
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute Shenyang, China
| | - Yongkai Wu
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute Shenyang, China
| | - Bo Huang
- Department of Pathology, Liaoning Cancer Hospital and Institute Shenyang, China
| | - Qian Liu
- Department of Pathology, Liaoning Cancer Hospital and Institute Shenyang, China
| | - Yinan Dong
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute Shenyang, China
| | - Jianqiao Ding
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute Shenyang, China
| | - Yongyu Liu
- Department of Thoracic Surgery, Liaoning Cancer Hospital and Institute Shenyang, China
| |
Collapse
|
22
|
Ko S, Kim JY, Jeong J, Lee JE, Yang WI, Jung WH. The role and regulatory mechanism of 14-3-3 sigma in human breast cancer. J Breast Cancer 2014; 17:207-18. [PMID: 25320618 PMCID: PMC4197350 DOI: 10.4048/jbc.2014.17.3.207] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 09/02/2014] [Indexed: 12/25/2022] Open
Abstract
Purpose 14-3-3 sigma (σ) is considered to be an important tumor suppressor and decreased expression of the same has been reported in many malignant tumors by hypermethylation at its promoter or ubiquitin-mediated proteolysis by estrogen-responsive ring finger protein (Efp). In this study, we investigated the significance of 14-3-3 σ expression in human breast cancer and its regulatory mechanism. Methods Efp was silenced using small interfering RNA (siRNA) in the MCF-7 breast cancer cell line in order to examine its influence on the level of 14-3-3 σ protein. The methylation status of the 14-3-3 σ promoter was also evaluated by methylation-specific polymerase chain reaction (PCR). The expression of Efp and 14-3-3 σ in 220 human breast carcinoma tissues was assessed by immunohistochemistry. Other clinicopathological parameters were also evaluated. Results Silencing Efp in the MCF-7 breast cancer cell line resulted in increased expression of 14-3-3 σ. The Efp-positive human breast cancers were more frequently 14-3-3 σ-negative (60.5% vs. 39.5%). Hypermethylation of 14-3-3 σ was common (64.9%) and had an inverse association with 14-3-3 σ positivity (p=0.072). Positive 14-3-3 σ expression was significantly correlated with poor prognosis: disease-free survival (p=0.008) and disease-specific survival (p=0.009). Conclusion Our data suggests that in human breast cancer, the regulation of 14-3-3 σ may involve two mechanisms: ubiquitin-mediated proteolysis by Efp and downregulation by hypermethylation. However, the inactivation of 14-3-3 σ is probably achieved mainly by hypermethylation. Interestingly, 14-3-3 σ turned out to be a very significant poor prognostic indicator, which is in contrast to its previously known function as a tumor suppressor, suggesting a different role of 14-3-3 σ in breast cancer.
Collapse
Affiliation(s)
- SeungSang Ko
- Department of Surgery, Cheil General Hospital & Women's Health Care Center, Catholic Kwandong University College of Medicine, Seoul, Korea
| | - Ji Young Kim
- Department of Pathology, CHA Gangnam Medical Center, CHA University, Seoul, Korea
| | - Joon Jeong
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Jong Eun Lee
- Department of Anatomy, Yonsei University College of Medicine, Seoul, Korea
| | - Woo Ick Yang
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Woo Hee Jung
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
23
|
Padden J, Megger DA, Bracht T, Reis H, Ahrens M, Kohl M, Eisenacher M, Schlaak JF, Canbay AE, Weber F, Hoffmann AC, Kuhlmann K, Meyer HE, Baba HA, Sitek B. Identification of novel biomarker candidates for the immunohistochemical diagnosis of cholangiocellular carcinoma. Mol Cell Proteomics 2014; 13:2661-72. [PMID: 25034945 PMCID: PMC4188994 DOI: 10.1074/mcp.m113.034942] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The aim of this study was the identification of novel biomarker candidates for the diagnosis of cholangiocellular carcinoma (CCC) and its immunohistochemical differentiation from benign liver and bile duct cells. CCC is a primary cancer that arises from the epithelial cells of bile ducts and is characterized by high mortality rates due to its late clinical presentation and limited treatment options. Tumorous tissue and adjacent non-tumorous liver tissue from eight CCC patients were analyzed by means of two-dimensional differential in-gel electrophoresis and mass-spectrometry-based label-free proteomics. After data analysis and statistical evaluation of the proteins found to be differentially regulated between the two experimental groups (fold change ≥ 1.5; p value ≤ 0.05), 14 candidate proteins were chosen for determination of the cell-type-specific expression profile via immunohistochemistry in a cohort of 14 patients. This confirmed the significant up-regulation of serpin H1, 14-3-3 protein sigma, and stress-induced phosphoprotein 1 in tumorous cholangiocytes relative to normal hepatocytes and non-tumorous cholangiocytes, whereas some proteins were detectable specifically in hepatocytes. Because stress-induced phosphoprotein 1 exhibited both sensitivity and specificity of 100%, an immunohistochemical verification examining tissue sections of 60 CCC patients was performed. This resulted in a specificity of 98% and a sensitivity of 64%. We therefore conclude that this protein should be considered as a potential diagnostic biomarker for CCC in an immunohistochemical application, possibly in combination with other candidates from this study in the form of a biomarker panel. This could improve the differential diagnosis of CCC and benign bile duct diseases, as well as metastatic malignancies in the liver.
Collapse
Affiliation(s)
- Juliet Padden
- From the ‡Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany;
| | - Dominik A Megger
- From the ‡Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Thilo Bracht
- From the ‡Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Henning Reis
- ¶Institut für Pathologie, Universitätsklinikum Essen, Universität Duisburg-Essen, 45141 Essen, Germany
| | - Maike Ahrens
- From the ‡Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Michael Kohl
- From the ‡Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Martin Eisenacher
- From the ‡Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Jörg F Schlaak
- ‖Klinik für Gastroenterologie und Hepatologie, Universitätsklinikum Essen, 45141 Essen, Universität Duisburg-Essen, 45141 Essen, Germany
| | - Ali E Canbay
- ‖Klinik für Gastroenterologie und Hepatologie, Universitätsklinikum Essen, 45141 Essen, Universität Duisburg-Essen, 45141 Essen, Germany
| | - Frank Weber
- **Klinik für Allgemeinchirurgie, Viszeral- und Transplantationschirurgie, Universitätsklinikum Essen, Universität Duisburg-Essen, 45141 Essen, Germany
| | - Andreas-Claudius Hoffmann
- ‡‡Innere Klinik (Tumorforschung), Westdeutsches Tumorzentrum, Universitätsklinikum Essen, Universität Duisburg-Essen, 45141 Essen, Germany
| | - Katja Kuhlmann
- From the ‡Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - Helmut E Meyer
- From the ‡Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany; §§Leibniz Institute for Analytical Sciences - ISAS, 44139 Dortmund, Germany
| | - Hideo A Baba
- ¶Institut für Pathologie, Universitätsklinikum Essen, Universität Duisburg-Essen, 45141 Essen, Germany
| | - Barbara Sitek
- From the ‡Medizinisches Proteom-Center, Ruhr-Universität Bochum, 44801 Bochum, Germany;
| |
Collapse
|
24
|
Qi YJ, Wang M, Liu RM, Wei H, Chao WX, Zhang T, Lou Q, Li XM, Ma J, Zhu H, Yang ZH, Liu HQ, Ma YF. Downregulation of 14-3-3σ correlates with multistage carcinogenesis and poor prognosis of esophageal squamous cell carcinoma. PLoS One 2014; 9:e95386. [PMID: 24743601 PMCID: PMC3990633 DOI: 10.1371/journal.pone.0095386] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/25/2014] [Indexed: 12/30/2022] Open
Abstract
Aims The asymptomatic nature of early-stage esophageal squamous cell carcinoma (ESCC) results in late presentation and consequent dismal prognosis This study characterized 14-3-3σ protein expression in the multi-stage development of ESCC and determined its correlation with clinical features and prognosis. Materials and Methods Western blot was used to examine 14-3-3σ protein expression in normal esophageal epithelium (NEE), low grade intraepithelial neoplasia (LGIN), high grade intraepithelial neoplasia (HGIN), ESCC of TNM I to IV stage and various esophageal epithelial cell lines with different biological behavior. Immunohistochemistry was used to estimate 14-3-3σ protein in 110 biopsy samples of NEE, LGIN or HGIN and in 168 ESCC samples all of whom had follow-up data. Support vector machine (SVM) was used to develop a classifier for prognosis. Results 14-3-3σ decreased progressively from NEE to LGIN, to HGIN, and to ESCC. Chemoresistant sub-lines of EC9706/PTX and EC9706/CDDP showed high expression of 14-3-3σ protein compared with non-chemoresistant ESCC cell lines and immortalized NEC. Furthermore, the downregulation of 14-3-3σ correlated significantly with histological grade (P = 0.000) and worse prognosis (P = 0.004). Multivariate Cox regression analysis indicated that 14-3-3σ protein (P = 0.016) and T stage (P = 0.000) were independent prognostic factors for ESCC. The SVM ESCC classifier comprising sex, age, T stage, histological grade, lymph node metastasis, clinical stage and 14-3-3σ, distinguished significantly lower- and higher-risk ESCC patients (91.67% vs. 3.62%, P = 0.000). Conclusions Downregulation of 14-3-3σ arises early in the development of ESCC and predicts poor survival, suggesting that 14-3-3σ may be a biomarker for early detection of high-risk subjects and diagnosis of ESCC. Our seven-feature SVM classifier for ESCC prognosis may help to inform clinical decisions and tailor individual therapy.
Collapse
Affiliation(s)
- Yi-Jun Qi
- Key Laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan, P. R. China
| | - Ming Wang
- Key Laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan, P. R. China
| | - Rui-Min Liu
- Key Laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan, P. R. China
| | - Hua Wei
- Huaihe Hospital, Henan University, Kaifeng, Henan, P. R. China
| | - Wei-Xia Chao
- Key Laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan, P. R. China
| | - Tian Zhang
- Key Laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan, P. R. China
| | - Qiang Lou
- Key Laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan, P. R. China
| | - Xiu-Min Li
- Xinxiang Medical University, Xinxiang, Henan, China
| | - Jin Ma
- Key Laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan, P. R. China
| | - Han Zhu
- Key Laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan, P. R. China
| | - Zhen-Hua Yang
- Linzhou Cancer Hospital, Linzhou, Henan, P. R. China
| | - Hai-Qing Liu
- Linzhou Cancer Hospital, Linzhou, Henan, P. R. China
| | - Yuan-Fang Ma
- Key Laboratory of Cellular and Molecular Immunology, College of Medicine, Henan University, Kaifeng, Henan, P. R. China
| |
Collapse
|
25
|
Expression of 14-3-3 sigma, cyclin-dependent kinases 2 and 4, p16, and Epstein–Barr nuclear antigen 1 in nasopharyngeal carcinoma. The Journal of Laryngology & Otology 2014; 128:134-41. [PMID: 24460960 DOI: 10.1017/s0022215113003447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AbstractObjective:The protein 14-3-3 sigma plays a role in cell cycle arrest by sequestering cyclin-dependent kinase 1 cyclin B1 complexes, as well as cyclin-dependent kinases 2 and 4, hence its definition as a cyclin-dependent kinase inhibitor. However, the nature of the interaction between these biological markers in nasopharyngeal carcinoma is unknown. This study aimed to investigate whether altered expression of these markers contributes to nasopharyngeal carcinogenesis.Methods:The study population consisted of 30 nasopharyngeal carcinoma patients and 10 patients without nasopharyngeal carcinoma. The nasopharyngeal carcinoma cell lines TW02, TW04 and Hone-1 were also assessed. We analysed levels of messenger RNA and protein for the p16 gene and the 14-3-3 sigma, Epstein–Barr nuclear antigen 1, and cyclin-dependent kinase 2 and 4 proteins, in nasopharyngeal carcinoma tissue specimens and cell lines and in normal nasopharyngeal tissue.Results:Protein and messenger RNA levels for cyclin-dependent kinase 2 and Epstein–Barr nuclear antigen 1 were significantly higher in nasopharyngeal carcinoma compared with normal tissue, while levels of cyclin-dependent kinase 4 generally were not; results for 14-3-3 sigma varied. Nasopharyngeal carcinoma patients had diminished p16 gene expression, compared with normal tissue.Conclusion:Levels of cyclin-dependent kinase 2 and Epstein–Barr nuclear antigen 1 were significantly higher in nasopharyngeal carcinoma than in normal tissue, while p16 gene expression was diminished. These three proteins may contribute to nasopharyngeal carcinogenesis.
Collapse
|
26
|
Nagappan A, Park HS, Park KI, Kim JA, Hong GE, Kang SR, Zhang J, Kim EH, Lee WS, Won CK, Kim GS. Proteomic analysis of differentially expressed proteins in vitamin C-treated AGS cells. BMC BIOCHEMISTRY 2013; 14:24. [PMID: 24067024 PMCID: PMC3848938 DOI: 10.1186/1471-2091-14-24] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/24/2013] [Indexed: 12/20/2022]
Abstract
BACKGROUND Vitamin C (ascorbic acid) is an essential nutrient of most living tissues that readily acts as a strong reducing agent, which is abundant in fruits and vegetables. Although, it inhibits cell growth in many human cancer cells in vitro, treatment in cancer is still controversial. Hence, the purpose of this study was to investigate the molecular mechanism of the inhibitory effect of vitamin C on AGS cell growth, and protein profiles in AGS cells after exposure to vitamin C treatment, by using proteomic tools. RESULTS Vitamin C showed a cytotoxic effect on AGS cells (IC50 300 μg/mL) and, 20 differentially expressed proteins (spot intensities which show ≥2 fold change and statistically significant, p<0.05 between the control and vitamin-C treated group) were successfully identified by assisted laser desorption/ ionization-time of flight/mass spectrometry (MALDI-TOF/MS). Of the 20 proteins, six were up-regulated and fourteen were down-regulated. Specifically, 14-3-3σ, 14-3-3ϵ, 14-3-3δ, tropomyosin alpha-3 chain and tropomyosin alpha-4 chain were down-regulated and peroxiredoxin-4 and thioredoxin domain-containing proteins 5 were up-regulated. The identified proteins are mainly involved in cell mobility, antioxidant and detoxification, signal transduction and protein metabolism. Further, the expressions of 14-3-3 isoforms were verified with immuno-blotting analysis. CONCLUSIONS Our proteome results suggest that the apoptosis related proteins were involved in promoting and regulating cell death of AGS cells, and might be helpful to understand the molecular mechanism of vitamin C on AGS cell growth inhibition.
Collapse
Affiliation(s)
- Arulkumar Nagappan
- Research Institute of Life Science and College of Veterinary Medicine, Gyeongsang National University, 900 Gajwadong, Jinju, Gyeongnam 660-701, Republic of Korea.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
14-3-3 Sigma is a Useful Immunohistochemical Marker for Diagnosing Ovarian Granulosa Cell Tumors and Steroid Cell Tumors. Int J Gynecol Pathol 2013; 32:156-62. [DOI: 10.1097/pgp.0b013e31825a0353] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Suzuki F, Nagase S, Suzuki K, Oba E, Hiroki E, Matsuda Y, Akahira JI, Nishigori H, Sugiyama T, Otsuki T, Yoshinaga K, Takano T, Niikura H, Ito K, Sasano H, Yaegashi N. Decreased Expression of 14-3-3 σ Is Predictive of Poor Prognosis for Patients with Human Uterine Papillary Serous Carcinoma. TOHOKU J EXP MED 2013; 231:193-9. [DOI: 10.1620/tjem.231.193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Fumihiko Suzuki
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Satoru Nagase
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Kichiya Suzuki
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
- Tohoku Medical Megabank Organization (ToMMo), Tohoku University
| | - Etsuko Oba
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Eri Hiroki
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Yukika Matsuda
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Jun-ichi Akahira
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
- Department of Pathology, Tohoku University Graduate School of Medicine
| | - Hidekazu Nishigori
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Takashi Sugiyama
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Takeo Otsuki
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Kousuke Yoshinaga
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Tadao Takano
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Hitoshi Niikura
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
| | - Kiyoshi Ito
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
- International Research Institute of Disaster Science (IRIDeS), Tohoku University
| | - Hironobu Sasano
- Department of Pathology, Tohoku University Graduate School of Medicine
| | - Nobuo Yaegashi
- Department of Obstetrics and Gynecology, Tohoku University Graduate School of Medicine
- Tohoku Medical Megabank Organization (ToMMo), Tohoku University
| |
Collapse
|
29
|
Gheibi A, Kazemi M, Baradaran A, Akbari M, Salehi M. Study of promoter methylation pattern of 14-3-3 sigma gene in normal and cancerous tissue of breast: A potential biomarker for detection of breast cancer in patients. Adv Biomed Res 2012; 1:80. [PMID: 23326810 PMCID: PMC3544103 DOI: 10.4103/2277-9175.102990] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Accepted: 03/12/2012] [Indexed: 12/25/2022] Open
Abstract
Background: In recent years, DNA methylation as a main epigenetic modification in human cancer is found as a promising biomarker in early detection of breast cancer. Possible applications of numerous hypermethylated genes have been reported in diagnosis of breast cancer but there has been a little comprehensive study on the clinical usefulness of these genes in breast cancer. The aim of the present study was to investigate the promoter methylation status of 14-3-3 sigma gene with the goal of developing a diagnostic application in breast cancer. Materials and Methods: Totally 40 cases of cancerous and noncancerous tissues were studied. DNA was extracted from tissue samples, and promoter methylation pattern was determined by using methylation-specific polymerase chain reaction. Results: Methylation pattern of 14-3-3 sigma promoter significantly differed between control and malignant breast tissues (P = 0.001), and there was no remarkable correlation between methylation and age (P > 0.05). Conclusion: The relationship of promoter methylation of 14-3-3 sigma with development of breast cancer found in this study and confirmed the results of previous reports suggests that we can provide the foundation for possible application of 14-3-3 sigma as a potential biomarker for early detection and monitoring disease status.
Collapse
Affiliation(s)
- A Gheibi
- Department of Biomedical Sciences, Division of Genetics, Isfahan, Iran
| | | | | | | | | |
Collapse
|
30
|
Liang S, Shen G, Liu Q, Xu Y, Zhou L, Xiao S, Xu Z, Gong F, You C, Wei Y. Isoform-specific expression and characterization of 14-3-3 proteins in human glioma tissues discovered by stable isotope labeling with amino acids in cell culture-based proteomic analysis. Proteomics Clin Appl 2012; 3:743-53. [PMID: 21136983 DOI: 10.1002/prca.200800198] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Human 14-3-3 proteins have isoform-specific expression and functions in different kinds of normal or tumor cells and tissues. However, the expression profiling of 14-3-3 proteins and isoform-specific biological functions are unclear in human glioma so far. In our study, the expression levels and characterization of 14-3-3 isoforms in human glioma tissues were investigated by a sensitive, accurate stable isotope labeling with amino acids in cell culture-based quantitative proteomic strategy. As a result, except unexpressed 14-3-3σ, the other six isoforms, with different expression levels, were existed in glioma tissues and para-cancerous brain tissues (PBTs). 14-3-3β and η were upregulated, whereas 14-3-3ζ was downregulated in glioma tissues compared with that in PBTs. And the other three isoforms 14-3-3ε, θ, and γ had similar expression levels in human glioma tissues and PBTs. Western blot and immunohistochemistry analysis were both consistent with the quantitative proteomic data. The loss of expression of 14-3-3σ was further discovered due to DNA high methylation in its coding region in glioma by methylation-specific PCR analysis. These results indicated that the four isoforms, including 14-3-3β, η, ζ, and σ, may play important roles in tumorigenesis of human glioma, which is probably used as potential biomarkers for diagnosis and targets for treatment of human gliomas in future.
Collapse
Affiliation(s)
- Shufang Liang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, P.R. China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Campa D, Müller P, Edler L, Knoefel L, Barale R, Heussel CP, Thomas M, Canzian F, Risch A. A comprehensive study of polymorphisms inABCB1, ABCC2andABCG2and lung cancer chemotherapy response and prognosis. Int J Cancer 2012; 131:2920-8. [DOI: 10.1002/ijc.27567] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 02/09/2012] [Indexed: 12/16/2022]
|
32
|
Genome-scale analysis of DNA methylation in lung adenocarcinoma and integration with mRNA expression. Genome Res 2012; 22:1197-211. [PMID: 22613842 PMCID: PMC3396362 DOI: 10.1101/gr.132662.111] [Citation(s) in RCA: 395] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lung cancer is the leading cause of cancer death worldwide, and adenocarcinoma is its most common histological subtype. Clinical and molecular evidence indicates that lung adenocarcinoma is a heterogeneous disease, which has important implications for treatment. Here we performed genome-scale DNA methylation profiling using the Illumina Infinium HumanMethylation27 platform on 59 matched lung adenocarcinoma/non-tumor lung pairs, with genome-scale verification on an independent set of tissues. We identified 766 genes showing altered DNA methylation between tumors and non-tumor lung. By integrating DNA methylation and mRNA expression data, we identified 164 hypermethylated genes showing concurrent down-regulation, and 57 hypomethylated genes showing increased expression. Integrated pathways analysis indicates that these genes are involved in cell differentiation, epithelial to mesenchymal transition, RAS and WNT signaling pathways, and cell cycle regulation, among others. Comparison of DNA methylation profiles between lung adenocarcinomas of current and never-smokers showed modest differences, identifying only LGALS4 as significantly hypermethylated and down-regulated in smokers. LGALS4, encoding a galactoside-binding protein involved in cell–cell and cell–matrix interactions, was recently shown to be a tumor suppressor in colorectal cancer. Unsupervised analysis of the DNA methylation data identified two tumor subgroups, one of which showed increased DNA methylation and was significantly associated with KRAS mutation and to a lesser extent, with smoking. Our analysis lays the groundwork for further molecular studies of lung adenocarcinoma by identifying novel epigenetically deregulated genes potentially involved in lung adenocarcinoma development/progression, and by describing an epigenetic subgroup of lung adenocarcinoma associated with characteristic molecular alterations.
Collapse
|
33
|
Shiba-Ishii A, Noguchi M. Aberrant stratifin overexpression is regulated by tumor-associated CpG demethylation in lung adenocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1653-62. [PMID: 22310466 DOI: 10.1016/j.ajpath.2011.12.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 12/02/2011] [Accepted: 12/09/2011] [Indexed: 01/05/2023]
Abstract
We previously have shown the aberrant overexpression of stratifin (SFN, 14-3-3 ς) in lung adenocarcinoma. Although SFN is known to facilitate tumor cell proliferation, the mechanism that underlies its aberrant expression has remained unclear. SFN, the downstream target of p53, often has been reported to be hypermethylated and subsequently silenced in certain cancers; however, its hypomethylation-linked reactivation has not yet been validated. In this study, we investigated the DNA methylation status of the SFN promoter region using 8 lung cancer cell lines and 32 specimens of adenocarcinoma tissue. Real-time methylation-specific PCR analysis showed that although both normal lung tissue and adenocarcinoma in situ bore a completely methylated SFN promoter, the promoter region in almost all invasive adenocarcinomas was at least partially methylated. The expression of SFN and its level of methylation were correlated strongly. Furthermore, statistical analysis revealed that the level of methylation became reduced with progression of the pathologic stage, although no clear relationship between methylation level and p53 abnormality was found. These results suggest that methylation-related silencing of SFN occurs in both normal lung tissues and adenocarcinoma in situ, and that demethylation of the SFN promoter participates in the aberrant expression of SFN in invasive adenocarcinoma cells, independently of p53 alteration. This novel finding might be informative for clarifying the mechanism that underlies the progression of early lung adenocarcinoma.
Collapse
Affiliation(s)
- Aya Shiba-Ishii
- Department of Pathology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | | |
Collapse
|
34
|
Abstract
BACKGROUND It remains important to understand the biology and identify biomarkers for less studied cancers like testicular cancer. The purpose of this study was to determine the methylation frequency of several cancer-related genes in different histological types of testicular cancer and normal testis tissues (NT). METHODS DNA was isolated from 43 seminomas (SEs), 14 non-SEs (NSEs) and 23 NT, and was assayed for promoter methylation status of 15 genes by quantitative methylation-specific PCR. The methylation status was evaluated for an association with cancer, and between SEs and NSEs. RESULTS We found differential methylation pattern in SEs and NSEs. MGMT, VGF, ER-β and FKBP4 were predominately methylated in NSEs compared with SEs. APC and hMLH1 are shown to be significantly more methylated in both subtypes in comparison with NT. When combining APC, hMLH1, ER-β and FKBP4, it is possible to identify 86% of the NSEs, whereas only 7% of the SEs. CONCLUSIONS Our results indicate that the methylation profile of cancer-associated genes in testicular cancer correlates with histological types and show cancer-specific pattern for certain genes. Further methylation analysis, in a larger cohort is needed to elucidate their role in testicular cancer development and potential for therapy, early detection and disease monitoring.
Collapse
|
35
|
Tan F, Jiang Y, Sun N, Chen Z, Lv Y, Shao K, Li N, Qiu B, Gao Y, Li B, Tan X, Zhou F, Wang Z, Ding D, Wang J, Sun J, Hang J, Shi S, Feng X, He F, He J. Identification of isocitrate dehydrogenase 1 as a potential diagnostic and prognostic biomarker for non-small cell lung cancer by proteomic analysis. Mol Cell Proteomics 2011; 11:M111.008821. [PMID: 22064513 DOI: 10.1074/mcp.m111.008821] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death in the world. To explore tumor biomarkers for clinical application, two-dimensional fluorescence difference gel electrophoresis and subsequent MALDI-TOF/TOF mass spectrometry were performed to identify proteins differentially expressed in 12 pairs of lung squamous cell tumors and their corresponding normal tissues. A total of 28 nonredundant proteins were identified with significant alteration in lung tumors. The up-regulation of isocitrate dehydrogenase 1 (IDH1), superoxide dismutase 2, 14-3-3ε, and receptor of activated protein kinase C1 and the down-regulation of peroxiredoxin 2 in tumors were validated by RT-PCR and Western blot analysis in independent 15 pairs of samples. Increased IDH1 expression was further verified by the immunohistochemical study in extended 73 squamous cell carcinoma and 64 adenocarcinoma clinical samples. A correlation between IDH1 expression and poor overall survival of non-small cell lung cancer (NSCLC) patients was observed. Furthermore, ELISA analysis showed that the plasma level of IDH1 was significantly elevated in NSCLC patients compared with benign lung disease patients and healthy individuals. In addition, knockdown of IDH1 by RNA interference suppressed the proliferation of NSCLC cell line and decreased the growth of xenograft tumors in vivo. These observations suggested that IDH1, as a protein promoting tumor growth, could be used as a plasma biomarker for diagnosis and a histochemical biomarker for prognosis prediction of NSCLC.
Collapse
Affiliation(s)
- Fengwei Tan
- Department of Thoracic Surgery, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Zhao J, Meyerkord CL, Du Y, Khuri FR, Fu H. 14-3-3 proteins as potential therapeutic targets. Semin Cell Dev Biol 2011; 22:705-12. [PMID: 21983031 DOI: 10.1016/j.semcdb.2011.09.012] [Citation(s) in RCA: 132] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 09/23/2011] [Indexed: 02/05/2023]
Abstract
The 14-3-3 family of phosphoserine/phosphothreonine-binding proteins dynamically regulates the activity of client proteins in various signaling pathways that control diverse physiological and pathological processes. In response to environmental cues, 14-3-3 proteins orchestrate the highly regulated flow of signals through complex networks of molecular interactions to achieve well-controlled physiological outputs, such as cell proliferation or differentiation. Accumulating evidence now supports the concept that either an abnormal state of 14-3-3 protein expression, or dysregulation of 14-3-3/client protein interactions, contributes to the development of a large number of human diseases. In particular, clinical investigations in the field of oncology have demonstrated a correlation between upregulated 14-3-3 levels and poor survival of cancer patients. These studies highlight the rapid emergence of 14-3-3 proteins as a novel class of molecular target for potential therapeutic intervention. The current status of 14-3-3 modulator discovery is discussed.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | | | | | | | | |
Collapse
|
37
|
Zhou WH, Tang F, Xu J, Wu X, Feng ZY, Li HG, Lin DJ, Shao CK, Liu Q. Aberrant upregulation of 14-3-3ơ expression serves as an inferior prognostic biomarker for gastric cancer. BMC Cancer 2011; 11:397. [PMID: 21933426 PMCID: PMC3184120 DOI: 10.1186/1471-2407-11-397] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2011] [Accepted: 09/20/2011] [Indexed: 12/18/2022] Open
Abstract
Background 14-3-3ơ is an intracellular, phosphoserine binding protein and proposed to be involved in tumorigenesis. However, the expression dynamics of 14-3-3ơ and its clinicopathological/prognostic significance in human tumors are still controversial. Methods The method of immunohistochemistry (IHC) and Western blot were utilized to examine the protein expression of 14-3-3ơ in gastric cancer and paired normal adjacent gastric mucosal tissues. Receive operating characteristic (ROC) curve analysis was employed to determine a cutoff score for 14-3-3ơ expression in a training set (n = 66). For validation, the ROC-derived cutoff score was subjected to analysis of the association of 14-3-3ơ expression with patient outcome and clinical characteristics in a testing set (n = 86) and overall patients (n = 152). Results The expression frequency and expression levels of 14-3-3ơ were significantly higher in gastric cancer than in normal gastric mucosal tissues. Correlation analysis demonstrated that high expression of 14-3-3ơ in gastric cancer was significantly correlated with clinical stage and tumor invasion. Furthermore, in the testing set and overall patients, Kaplan-Meier analysis showed that elevated 14-3-3ơ expression predicted poorer overall survival (OS) and progression-free survival (PFS). Importantly, high 14-3-3ơ expression was also associated with shortened survival time in stage III and stage IV gastric cancer patients. Multivariate analyses revealed that 14-3-3ơ expression was an independent prognostic parameter in gastric cancer. Conclusions These findings provide evidence that high expression of 14-3-3ơ may be important in the tumor progression and servers as an independent molecular marker for poor prognosis of gastric cancer. Thus, overexpression of 14-3-3ơ identifies patients at high risk and is a novel therapeutic molecular target for this tumor.
Collapse
Affiliation(s)
- Wei-hua Zhou
- Department of Hematology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhang C, Elkahloun AG, Robertson M, Gills JJ, Tsurutani J, Shih JH, Fukuoka J, Hollander MC, Harris CC, Travis WD, Jen J, Dennis PA. Loss of cytoplasmic CDK1 predicts poor survival in human lung cancer and confers chemotherapeutic resistance. PLoS One 2011; 6:e23849. [PMID: 21887332 PMCID: PMC3161069 DOI: 10.1371/journal.pone.0023849] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 07/26/2011] [Indexed: 01/15/2023] Open
Abstract
The dismal lethality of lung cancer is due to late stage at diagnosis and inherent therapeutic resistance. The incorporation of targeted therapies has modestly improved clinical outcomes, but the identification of new targets could further improve clinical outcomes by guiding stratification of poor-risk early stage patients and individualizing therapeutic choices. We hypothesized that a sequential, combined microarray approach would be valuable to identify and validate new targets in lung cancer. We profiled gene expression signatures during lung epithelial cell immortalization and transformation, and showed that genes involved in mitosis were progressively enhanced in carcinogenesis. 28 genes were validated by immunoblotting and 4 genes were further evaluated in non-small cell lung cancer tissue microarrays. Although CDK1 was highly expressed in tumor tissues, its loss from the cytoplasm unexpectedly predicted poor survival and conferred resistance to chemotherapy in multiple cell lines, especially microtubule-directed agents. An analysis of expression of CDK1 and CDK1-associated genes in the NCI60 cell line database confirmed the broad association of these genes with chemotherapeutic responsiveness. These results have implications for personalizing lung cancer therapy and highlight the potential of combined approaches for biomarker discovery.
Collapse
Affiliation(s)
- Chunyu Zhang
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Abdel G. Elkahloun
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Matthew Robertson
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joell J. Gills
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Junji Tsurutani
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Medical Oncology Department, Kinki University School of Medicine, Osaka-Sayama, Osaka, Japan
| | - Joanna H. Shih
- Biometric Research Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Junya Fukuoka
- Laboratory of Population Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Surgical Pathology, Toyama University Hospital, Toyama, Japan
| | - M. Christine Hollander
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Curtis C. Harris
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - William D. Travis
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Jin Jen
- Laboratory of Population Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Department of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Phillip A. Dennis
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
39
|
Boyarskikh UA, Kondrakhin YV, Yevshin IS, Sharipov RN, Komelkov AV, Musatkina EA, Tchevkina EM, Sukoyan MA, Kolpakov FA, Kashkin KN, Filipenko ML. Prediction of nonsmall cell lung cancer sensitivity to cisplastin and paclitaxel based on marker gene expression. Mol Biol 2011. [DOI: 10.1134/s0026893311030034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
Esposito L, Conti D, Ailavajhala R, Khalil N, Giordano A. Lung Cancer: Are we up to the Challenge? Curr Genomics 2011; 11:513-8. [PMID: 21532835 PMCID: PMC3048313 DOI: 10.2174/138920210793175903] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2010] [Revised: 06/08/2010] [Accepted: 07/26/2010] [Indexed: 12/31/2022] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide among both men and women, with more than 1 million deaths annually. Non–small cell lung cancer (NSCLC) accounts for about 80% of all lung cancers. Although recent advances have been made in diagnosis and treatment strategies, the prognosis of NSCLC patients is poor and it is basically due to a lack of early diagnostic tools. However, in the last years genetic and biochemical studies have provided more information about the protein and gene’s mutations involved in lung tumors. Additionally, recent proteomic and microRNA’s approaches have been introduced to help biomarker discovery. Here we would like to discuss the most recent discoveries in lung cancer pathways, focusing on the genetic and epigenetic factors that play a crucial role in malignant cell proliferation, and how they could be helpful in diagnosis and targeted therapy.
Collapse
Affiliation(s)
- Luca Esposito
- Oncology Research Centre of Mercogliano, Avellino, Italy
| | | | | | | | | |
Collapse
|
41
|
Shiba-Ishii A, Kano J, Morishita Y, Sato Y, Minami Y, Noguchi M. High expression of stratifin is a universal abnormality during the course of malignant progression of early-stage lung adenocarcinoma. Int J Cancer 2011; 129:2445-53. [PMID: 21207417 DOI: 10.1002/ijc.25907] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Accepted: 12/10/2010] [Indexed: 11/08/2022]
Abstract
Adenocarcinoma in situ (AIS) of the lung has an extremely favorable prognosis, with a 5-year survival rate of 100%. However, early invasive adenocarcinoma (EIA) often has a fatal outcome. In this study, we compared the expression profiles of AIS with those of EIA showing lymph node metastasis or a fatal outcome, and screened the differentially expressed genes by cDNA microarray. From the genes selected, we focused on Stratifin (SFN, 14-3-3 σ), which showed significantly higher expression in EIA than in AIS. Immunohistochemistry for SFN revealed that more than 95% of EIAs were immunopositive for SFN, in comparison to only 13% of AISs (p <0.05). Interestingly, positivity was detected not only in the invasive region but also in the in situ spreading component of EIA. Functionally, SFN facilitates the cell proliferation capacity of lung adenocarcinoma. These results indicate that SFN overexpression is a universal abnormality during the stepwise progression from in situ to invasive adenocarcinoma of the lung.
Collapse
Affiliation(s)
- Aya Shiba-Ishii
- Department of Pathology, Institute of Basic Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba-shi, Ibaraki 305-8575, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Neal CL, Yu D. 14-3-3ζ as a prognostic marker and therapeutic target for cancer. Expert Opin Ther Targets 2011; 14:1343-54. [PMID: 21058923 DOI: 10.1517/14728222.2010.531011] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
IMPORTANCE OF THE FIELD The ubiquitously expressed 14-3-3ζ protein is involved in numerous important cellular pathways involved in cancer. Recent research suggests 14-3-3ζ may play a central role regulating multiple pathways responsible for cancer initiation and progression. This review will provide an overview of 14-3-3 proteins and address the role of 14-3-3ζ overexpression in cancer. AREAS COVERED IN THIS REVIEW The review covers the basic role of 14-3-3 in regulation of multiple pathways with a focus on 14-3-3ζ as a clinically relevant biomarker for cancer recurrence. WHAT THE READER WILL GAIN 14-3-3ζ overexpression has been found in multiple cancers; however, the clinical implications were unclear. Recently, 14-3-3ζ has been identified as a biomarker for poor prognosis and chemoresistance in multiple tumor types, indicating a potential clinical application for using 14-3-3ζ in selecting treatment options and predicting cancer patients' outcome. TAKE HOME MESSAGE 14-3-3ζ is a potential prognostic marker of cancer recurrence and predictive marker for therapeutic resistance. The overexpression of 14-3-3ζ in multiple cancers suggests that it may be a common target to intervene tumor progression; therefore, more efforts are needed for the development of 14-3-3 inhibitors.
Collapse
Affiliation(s)
- Christopher L Neal
- The University of Texas M. D. Anderson Cancer Center, Department of Molecular and Cellular Oncology, Houston, TX 77030, USA
| | | |
Collapse
|
43
|
Li Z, Dong Z, Myer D, Yip-Schneider M, Liu J, Cui P, Schmidt CM, Zhang JT. Role of 14-3-3σ in poor prognosis and in radiation and drug resistance of human pancreatic cancers. BMC Cancer 2010; 10:598. [PMID: 21040574 PMCID: PMC2991307 DOI: 10.1186/1471-2407-10-598] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Accepted: 11/01/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Pancreatic cancer is the fourth leading cause of death in the US. Unlike other solid tumors such as testicular cancer which are now curable, more than 90% of pancreatic cancer patients die due to lack of response to therapy. Recently, the level of 14-3-3σ mRNA was found to be increased in pancreatic cancers and this increased expression may contribute to the failure in treatment of pancreatic cancers. In the present study, we tested this hypothesis. METHODS Western blot analysis was used to determine 14-3-3σ protein level in fresh frozen tissues and was correlated to clinical outcome. A stable cell line expressing 14-3-3σ was established and the effect of 14-3-3σ over-expression on cellular response to radiation and anticancer drugs were tested using SRB assay and clonogenic assays. Cell cycle distribution and apoptosis analyses were performed using propidium iodide staining and PARP cleavage assays. RESULTS We found that 14-3-3σ protein level was increased significantly in about 71% (17 of 24) of human pancreatic cancer tissues and that the 14-3-3σ protein level in cancers correlated with lymph node metastasis and poor prognosis. Furthermore, we demonstrated that over-expression of 14-3-3σ in a pancreatic cancer cell line caused resistance to γ-irradiation as well as anticancer drugs by causing resistance to treatment-induced apoptosis and G2/M arrest. CONCLUSION The increased level of 14-3-3σ protein likely contributes to the poor clinical outcome of human pancreatic cancers by causing resistance to radiation and anticancer drugs. Thus, 14-3-3σ may serve as a prognosis marker predicting survival of pancreatic cancer patients and guide the clinical treatment of these patients.
Collapse
Affiliation(s)
- Zhaomin Li
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Yeh CN, Pang ST, Jung SM, Chen TW, Jan YY, Chen MF. Cytoplasmic overexpression with membrane accentuation of stratifin in intrahepatic mass-forming cholangiocarcinoma after hepatectomy: Correlation with clinicopathologic features and patient survival. J Surg Oncol 2010; 102:608-14. [DOI: 10.1002/jso.21604] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
45
|
Ren HZ, Pan GQ, Wang JS, Wen JF, Wang KS, Luo GQ, Shan XZ. Reduced stratifin expression can serve as an independent prognostic factor for poor survival in patients with esophageal squamous cell carcinoma. Dig Dis Sci 2010; 55:2552-60. [PMID: 20108042 DOI: 10.1007/s10620-009-1065-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 11/20/2009] [Indexed: 12/20/2022]
Abstract
UNLABELLED Stratifin plays an important role in cancer biology by interfering with intracellular signalling pathways and cell-cycle checkpoints. Decreased expression of stratifin gene has been reported to be a poor prognostic indicator in a variety of human malignant tumors. AIM To clarify the role and prognostic significance of stratifin in esophageal squamous cell carcinoma (ESCC). METHODS The alteration of stratifin messenger RNA (mRNA) and protein was analyzed by reverse-transcription and quantitative real-time polymerase chain reaction (QRT-PCR) and Western blotting in 20 paired ESCC and nonneoplastic esophageal mucosa tissues, respectively. Then, immunohistochemistry (IHC) was used to evaluate expression of stratifin in tissues of 148 ESCC patients (including the former 20 pairs of tissues) and correlate it with clinicopathological parameters and prognosis of ESCC patients. RESULTS The stratifin level of mRNA and protein was markedly downregulated in ESCC tissue compared with in corresponding nonneoplastic esophageal epithelium (P<0.05). Similarly, the positive rate of stratifin protein expression was lower in the esophageal cancer than in paired nonneoplastic esophageal epithelium as detected by IHC (P=0.007). Statistically, the downregulation of stratifin expression was correlated with tumor infiltration depth (P=0.003), lymph node metastasis (P=0.008), distant metastasis (P=0.013), and lymphovascular invasion (P=0.007) of ESCC. Furthermore, the reduced stratifin expression was associated with shorter 5-year survival rate of ESCC patients after curative surgery (P<0.0001). On the basis of univariate and multivariate Cox regression analysis, we found that reduced stratifin expression, T4 stage, lymph node metastasis, and distant metastasis were independent risk factors for worse prognosis in ESCC patients. CONCLUSION The present report indicates that stratifin could be a useful indicator for prognosis of this disease, as well as a potential target for more effective therapy.
Collapse
MESH Headings
- 14-3-3 Proteins/analysis
- 14-3-3 Proteins/genetics
- Adult
- Aged
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Blotting, Western
- Carcinoma, Squamous Cell/chemistry
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/secondary
- Carcinoma, Squamous Cell/surgery
- Chi-Square Distribution
- Down-Regulation
- Esophageal Neoplasms/chemistry
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/mortality
- Esophageal Neoplasms/pathology
- Esophageal Neoplasms/surgery
- Esophagectomy
- Exonucleases/analysis
- Exonucleases/genetics
- Exoribonucleases
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Kaplan-Meier Estimate
- Lymphatic Metastasis
- Male
- Middle Aged
- Neoplasm Invasiveness
- Neoplasm Staging
- Predictive Value of Tests
- Proportional Hazards Models
- RNA, Messenger/analysis
- Reverse Transcriptase Polymerase Chain Reaction
- Risk Assessment
- Risk Factors
- Time Factors
- Treatment Outcome
Collapse
Affiliation(s)
- Hong-Zheng Ren
- Department of Pathology, Xiangya Basic Medical College, Central South University, Changsha, China
| | | | | | | | | | | | | |
Collapse
|
46
|
Suzuki M, Yoshino I. Aberrant methylation in non-small cell lung cancer. Surg Today 2010; 40:602-7. [DOI: 10.1007/s00595-009-4094-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Accepted: 02/16/2009] [Indexed: 01/15/2023]
|
47
|
Kang S, Dong SM, Park NH. Frequent promoter hypermethylation of TGFBI in epithelial ovarian cancer. Gynecol Oncol 2010; 118:58-63. [PMID: 20421128 DOI: 10.1016/j.ygyno.2010.03.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Revised: 03/30/2010] [Accepted: 03/31/2010] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Using pharmacologic unmasking and genome-wide differential methylation analysis, we identified a novel methylated gene in ovarian cancers. METHODS Two ovarian cancer cells (OVCAR-3, ES-2) that showed synergistic growth inhibition by 5-aza-dC and cisplatin were selected. After treatment with 5-aza-dC, differential expression profiles were compared using microarray that contained 38,500 genes. Reactivation of candidate genes and their promoter methylation were validated by real-time RT-PCR, MS-PCR and bisulfite sequencing. Methylation status was tested by MS-PCR in 56 patients with epithelial ovarian cancer and compared to the 38 normal ovarian tissues. RESULTS We identified 103 candidate genes that were reactivated by 5-aza-dC treatment. Among those, SFN and TGFBI were commonly reactivated in both cells. Since SFN is a well known methylated marker, we selected TGFBI for further validation. Bisulfite sequencing revealed complete promoter methylation in ES-2 and partial methylation in OVCAR-3. In addition, silencing of TGFBI at the transcription level was reversed by 5-aza-dC treatment. TGFBI methylation was observed in 23 out of 38 (60.5%) cases of ovarian cancer, in no normal ovarian tissues (0 of 38, P=0.001), and in 5 out of 18 (27.8%) borderline tumors (P=0.044). In our cohort, we did not observe any association between methylation of TGFBI and clinicopathologic variables or clinical outcomes. CONCLUSION Our results confirm that TGFBI is frequently methylated in ovarian cancer. Its methylation can be used as a novel epigenetic biomarker in discriminating ovarian cancer from non-cancer or borderline tumors.
Collapse
Affiliation(s)
- Sokbom Kang
- Research Institute and Hospital, National Cancer Center, Goyang, Republic of Korea
| | | | | |
Collapse
|
48
|
Mirza S, Sharma G, Parshad R, Srivastava A, Gupta SD, Ralhan R. Clinical significance of Stratifin, ERα and PR promoter methylation in tumor and serum DNA in Indian breast cancer patients. Clin Biochem 2010; 43:380-6. [DOI: 10.1016/j.clinbiochem.2009.11.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 11/18/2009] [Accepted: 11/19/2009] [Indexed: 10/20/2022]
|
49
|
Fu X, Li J, Li K, Tian X, Zhang Y. Hypermethylation of APC promoter 1A is associated with moderate activation of Wnt signalling pathway in a subset of colorectal serrated adenomas. Histopathology 2010; 55:554-63. [PMID: 19912361 DOI: 10.1111/j.1365-2559.2009.03411.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AIMS The role of Wnt signalling pathway in serrated adenomas (SAs) remains to be identified. The aim of this study was to determine whether Wnt signalling plays a role in the pathogenesis of SAs, and to clarify the mechanism of Wnt signalling activation in SAs. METHODS AND RESULTS This study investigated immunoreactivities of adenomatous polyposis coli (APC) and beta-catenin, mutations of APC and beta-catenin genes, methylation status of APC promoter 1A in 12 SAs, and compared the findings with normal colorectal mucosa, hyperplastic polyps, traditional adenomas (TAs) and colorectal cancers (CRCs). APC expression was moderately decreased in SAs. Cytoplasmic accumulation of beta-catenin was demonstrated in 41.7% (5/12) of SAs, but membranous immunoreactivity of beta-catenin was lost in only 8.3% (1/12) of SAs. No beta-catenin mutation was detected in any of 12 SAs, and only one SA was found to be positive for APC gene mutation. Complete methylation of APC promoter 1A was found in 41.7% (5/12) of SAs, but in no TAs or CRCs. CONCLUSIONS Hypermethylation of APC promoter 1A, instead of mutations involving APC and beta-catenin, contributes to moderate activation of Wnt signalling in a subset of SAs.
Collapse
Affiliation(s)
- Xiangsheng Fu
- Institute for Digestive Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | | | | | | | | |
Collapse
|
50
|
Van Den Broeck A, Ozenne P, Eymin B, Gazzeri S. Lung cancer: a modified epigenome. Cell Adh Migr 2010; 4:107-13. [PMID: 20139698 DOI: 10.4161/cam.4.1.10885] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Epigenetic is the study of heritable changes in gene expression that occur without changes in DNA sequence. This process is important for gene expression and genome stability and its disruption is now thought to play a key role in the onset and progression of numerous tumor types. The most studied epigenetic phenomena includes post-translational modifications in DNA and histone proteins as well as microRN As expression. As epigenetic aberrations are potentially reversible, their correction has emerged as a potential strategy for the treatment of cancer. This review highlights the roles of chromatin epigenetic modifications and of microRN As expression in lung tumorigenesis and discusses the emerging epigenetic therapies which are being developed for the treatment of lung cancer.
Collapse
Affiliation(s)
- Arnaud Van Den Broeck
- Equipe Bases Moléculaires de la Progression des Cancers du Poumon, Centre de Recherche INSERM U823, Institut Albert Bonniot, Grenoble, France
| | | | | | | |
Collapse
|