1
|
Saadh MJ, Allela OQB, Kareem RA, Baldaniya L, Ballal S, Vashishth R, Parmar M, Sameer HN, Hamad AK, Athab ZH, Adil M. Prognostic gene expression profile of colorectal cancer. Gene 2025; 955:149433. [PMID: 40122415 DOI: 10.1016/j.gene.2025.149433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer is a major global health burden, with significant heterogeneity in clinical outcomes among patients. Identifying robust prognostic gene expression signatures can help stratify patients, guide treatment decisions, and improve clinical management. This review provides an overview of current prognostic gene expression profiles in colorectal cancer research. We have synthesized evidence from numerous published studies investigating the association between tumor gene expression patterns and patient survival outcomes. The reviewed literature reveals several promising gene signatures that have demonstrated the ability to predict disease-free survival and overall survival in CRC patients, independent of standard clinicopathological risk factors. These genes are crucial in fundamental biological processes, including cell cycle control, epithelial-mesenchymal transition, and immune regulation. The implementation of prognostic gene expression tests in clinical practice holds great potential for enabling more personalized management strategies for colorectal cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Manisha Parmar
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India.
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq.
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq.
| | | |
Collapse
|
2
|
Shur S, Sommer AK, Latchford A, Spier I, Katz LH. A review of APC somatic mosaicism and specific APC variants - I1307K and promotor variants. Fam Cancer 2025; 24:39. [PMID: 40237877 PMCID: PMC12003607 DOI: 10.1007/s10689-025-00464-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 03/30/2025] [Indexed: 04/18/2025]
Abstract
In the majority of patients with a classical Familial Adenomatous Polyposis (FAP) a pathogenic APC germline variant is identified; usually these are truncating variants in the coding region of APC. However, there are some special circumstances in which FAP is not the result of a pathogenic heterozygous germline variant in APC (mosaicism) and tspecific APC variants which do not cause FAP (I1307K and promotor variants). This paper will discuss these three conditions. APC somatic (postzygotic) mosaicism can be identified in up to 50% of unexplained adenomatous polyposis cases. The ability to identify APC postzygotic mosaicism depends on the the detection method (today usually next-generation sequencing) and also the tissue being analysed (investigation of multiple colorectal adenomas is more sensitive than leukocyte DNA). Identifying mosaicism has important implications in terms of an individual's management and managing risk in family members. The I1307K variant in APC is prevalent among Ashkenazi Jews (AJ) but can also be found in Sephardi Jews and individuals of non-Jewish descent. While this variant does not cause polyposis, it increases the risk of colorectal cancer (CRC) by 1.68-fold in AJ individuals. However, the link between the I1307K variant and CRC risk in non-AJ populations, is less well-established. Furthermore, its potential impact on other types of cancer remains unclear. Consequently, the classification of this variant, along with appropriate screening and surveillance recommendations, remains a subject of ongoing debate among leading medical and genetic organizations. Variants in the APC promotor 1B region cause the relatively newly described condition of gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS). It is said to have an isolated gastric phenotype, with neither duodenal, large bowel nor extra-intestinal manifestations. There are many uncertainties regarding this condition, it's penetrance and management. Lack of clinical data and poor understanding of the natural history of the condition remain significant barriers to developing guidelines to manage this condition.
Collapse
Affiliation(s)
- Shira Shur
- The Gonczarowski Family Institute of Gastroenterology and Liver Diseases, Shamir Medical Center, Zerifin, Israel
| | - Anna K Sommer
- Institute of Human Genetics, Medical Faculty University of Bonn, Bonn, Germany
| | - Andrew Latchford
- Polyposis Registry, Centre for Familial Intestinal Cancer, St Mark's Hospital, London, UK
- Department of surgery and cancer, Imperial College London, London, UK
| | - Isabel Spier
- Institute of Human Genetics, Medical Faculty University of Bonn, Bonn, Germany
- National Center for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Germany
| | - Lior H Katz
- Department of Gastroenterology, Hadassah Medical Center, Hadassah Ein-kerem, Jerusalem, Israel.
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
3
|
Ivey A, Pratt H, Boone BA. Molecular pathogenesis and emerging targets of gastric adenocarcinoma. J Surg Oncol 2022; 125:1079-1095. [PMID: 35481910 PMCID: PMC9069999 DOI: 10.1002/jso.26874] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/15/2022] [Accepted: 03/19/2022] [Indexed: 12/24/2022]
Abstract
Gastric adenocarcinoma (GC) is a devastating disease and is the third leading cause of cancer deaths worldwide. This heterogeneous disease has several different classification systems that consider histological appearance and genomic alterations. Understanding the etiology of GC, including infection, hereditary conditions, and environmental factors, is of particular importance and is discussed in this review. To improve survival in GC, we also must improve our therapeutic strategies. Here, we discuss new targets that warrant further exploration.
Collapse
Affiliation(s)
- Abby Ivey
- Department of Cancer Cell Biology, West Virginia University Cancer Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Hillary Pratt
- Department of Cancer Cell Biology, West Virginia University Cancer Institute, West Virginia University, Morgantown, West Virginia, USA
| | - Brian A Boone
- Department of Cancer Cell Biology, West Virginia University Cancer Institute, West Virginia University, Morgantown, West Virginia, USA
- Department of Surgery, Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, West Virginia, USA
| |
Collapse
|
4
|
Gastric Xanthelasma, Microsatellite Instability and Methylation of Tumor Suppressor Genes in the Gastric Mucosa: Correlation and Comparison as a Predictive Marker for the Development of Synchronous/Metachronous Gastric Cancer. J Clin Med 2021; 11:jcm11010009. [PMID: 35011751 PMCID: PMC8745081 DOI: 10.3390/jcm11010009] [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: 11/24/2021] [Revised: 12/13/2021] [Accepted: 12/17/2021] [Indexed: 11/23/2022] Open
Abstract
A predictive marker for the development of synchronous/metachronous gastric cancer (GC) would be highly desirable in order to establish an effective strategy for endoscopic surveillance. Herein, we examine the significance of gastric xanthelasma (GX) and molecular abnormalities for the prediction of synchronous/metachronous GC. Patients (n = 115) were followed up (range, 12–122; median, 55 months) in whom the presence of GX and molecular alterations, including microsatellite instability (MSI) and methylation of human mutL homolog 1 (hMLH1), cyclin-dependent kinase inhibitor 2A (CDKN2A) and adenomatous polyposis coli (APC) genes, had been confirmed in non-neoplastic gastric mucosa when undergoing endoscopic submucosal dissection (ESD) for early GC. At the start of surveillance, the numbers of positive subjects were as follows: GX, 59 (51.3%); MSI, 48 (41.7%); hMLH1, 37 (32.2%); CDKN2A, 7 (6.1%); APC, 18 (15.7%). After ESD treatment, synchronous/metachronous GCs occurred in patients with the following positive factors: GX, 16 (27.1%); MSI, 7 (14.6%); hMLH1, 6 (16.2%); CDKN2A, 3 (42.9%); APC, 3 (16.7%). The presence of GX had no significant relationship to positivity for MSI or methylation of hMLH1, CDKN2A or APC. GX was significantly (p = 0.0059) and independently (hazard ratio, 3.275; 95% confidence interval, 1.134–9.346) predictive for the development of synchronous/metachronous GC, whereas those genetic alterations were not predictive. GX is a simple and powerful marker for predicting the development of synchronous or metachronous GC.
Collapse
|
5
|
Zhu L, Li X, Yuan Y, Dong C, Yang M. APC Promoter Methylation in Gastrointestinal Cancer. Front Oncol 2021; 11:653222. [PMID: 33968756 PMCID: PMC8103321 DOI: 10.3389/fonc.2021.653222] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/30/2021] [Indexed: 12/30/2022] Open
Abstract
The adenomatous polyposis coli (APC) gene, known as tumor suppressor gene, has the two promoters 1A and 1B. Researches on APC have usually focused on its loss-of-function variants causing familial adenomatous polyposis. Hypermethylation, however, which is one of the key epigenetic alterations of the APC CpG sequence, is also associated with carcinogenesis in various cancers. Accumulating studies have successively explored the role of APC hypermethylation in gastrointestinal (GI) tumors, such as in esophageal, colorectal, gastric, pancreatic, and hepatic cancer. In sporadic colorectal cancer, the hypermethylation of CpG island in APC is even considered as one of the primary causative factors. In this review, we systematically summarized the distribution of APC gene methylation in various GI tumors, and attempted to provide an improved general understanding of DNA methylation in GI tumors. In addition, we included a robust overview of demethylating agents available for both basic and clinical researches. Finally, we elaborated our findings and perspectives on the overall situation of APC gene methylation in GI tumors, aiming to explore the potential research directions and clinical values.
Collapse
Affiliation(s)
- Lila Zhu
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyu Li
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Yuan
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Caixia Dong
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mengyuan Yang
- Department of Medical Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
6
|
Abstract
Purpose of review Gastric cancer is a leading cause of cancer death in the world. Between 1% and 3% of cases are associated with specific genetic cancer risk syndromes. The purpose of this article is to review the latest insights, as well as gaps in knowledge, regarding some of the most common hereditary gastric cancer syndromes: hereditary diffuse gastric cancer (HDGC), gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS), Lynch syndrome, the adenomatous polyposis syndromes, and the hamartomatous polyposis syndromes. Recent findings Patients carrying pathogenic variants in CDH1, but not meeting clinical criteria for HDGC, are increasingly being identified thanks to multigene panel testing; their absence from previous analyses overestimated gastric cancer penetrance. GAPPS is a recently described hereditary gastric cancer syndrome associated with specific point mutations in the promoter 1B region of the APC gene. Summary Risk of gastric cancer is highest among carriers of pathogenic variants in CDH1, with cumulative incidences approximately 40% and 30% for men and women, respectively. Mutations associated with Lynch syndrome and adenomatous polyposis syndromes confer greatest risk for gastric cancer in East Asian populations. Risk of gastric cancer in GAPPS and hamartomatous polyposis syndromes is difficult to estimate due to their rarity, but mutation status likely determines risk. Future research is needed to more precisely define risk of gastric cancer in these syndromes, so strategies for screening and prophylactic gastrectomy can be optimized.
Collapse
|
7
|
Gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS) - A Helicobacter-opposite point. Best Pract Res Clin Gastroenterol 2021; 50-51:101728. [PMID: 33975682 DOI: 10.1016/j.bpg.2021.101728] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/08/2021] [Indexed: 01/31/2023]
Abstract
Gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS) is a rare familial gastric cancer syndrome with an autosomal dominant pattern of inheritance. It is characterised by fundic gland polyposis of the gastric body and is associated with a significant risk of gastric adenocarcinoma. Unlike sporadic gastric cancer, Helicobacter pylori is usually absent in patients with GAPPS. This opposite-point finding has so far not been fully clarified. Prophylactic total gastrectomy is indicated in all cases of GAPPS with fundic gland polyposis and the presence of any dysplasia. If no dysplasia is found at histology, prophylactic gastrectomy is suggested at between 30 and 35 years of age, or at five years earlier than the age at which the youngest family member developed gastric cancer. Different phenotypes of GAPPS demand an individual approach to particular family members.
Collapse
|
8
|
Saelee P, Pongtheerat T. APC Promoter Hypermethylation as a Prognostic Marker in Breast Cancer Patients. Asian Pac J Cancer Prev 2020; 21:3627-3632. [PMID: 33369461 PMCID: PMC8046330 DOI: 10.31557/apjcp.2020.21.12.3627] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Adenomatous polyposis coli (APC) promoter hypermethylation implicated in breast cancer development through Wnt signaling pathway, hypermethylation may result in inactivation of APC expression. This study aimed to investigated whether hypermethylation of APC promoter, the aggressive behavior of breast cancer cells, and correlated them with clinicopathological parameters and survival. Methods: Sixty-one fresh tissues of breast tumor were evaluated for APC promoter hypermethylation with methylation-specific PCR techniques (MS-PCR) and APC mRNA expression level analysis by quantitative real-time reverse transcription-PCR. Results: Our results show aberrant APC hypermethylation status was founded in 27 of 61 cases (44%), and significantly associated with chemotherapy treatment (OR= 6.9, 95%CI=1.5-31.01, P = 0.01), distant metastasis (OR = 5.52, 95%CI = 1.27-24.08, P = 0.04) as well as APC methylated status also associated with shorter overall survival than those without (8.4 and 11.0 years respectively, P = 0.02). Conclusion: The findings indicated hypermethylation of APC promoter may be used as a useful prognostic biomarker in breast cancer patients.
Collapse
Affiliation(s)
- Pensri Saelee
- Research Division, National Cancer Institute, Bangkok 10400, Thailand
| | - Tanett Pongtheerat
- Unit of Biochemistry, Department of Medical Sciences, Faculty of Science, Rangsit University, Patumthani, Thailand
| |
Collapse
|
9
|
Mahdiannasser M, Haghpanah V, Damavandi E, Kabuli M, Tavangar SM, Larijani B, Ghadami M. Investigation of promoter methylation of FSCN1 gene and FSCN1 protein expression in differentiated thyroid carcinomas. Mol Biol Rep 2020; 47:2161-2169. [PMID: 32072403 DOI: 10.1007/s11033-020-05315-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 02/07/2020] [Indexed: 01/18/2023]
Abstract
FSCN1 gene encodes an actin-bundling protein, FSCN1, which is involved in formation of actin-based structures that contribute to cell migration. High levels of FSCN1 expression is observed in cells with extended membranes and protrusions. Moreover, up-regulation of FSCN1 has been reported in several epithelial carcinomas. Therefore, FSCN1 is thought to play a role in cell movement and invasion. However, the mechanism behind FSCN1 up-regulation is not known. We investigated the expression of FSCN1 using immunohistochemistry. Methylation-specific PCR was adopted to analyze the methylation status of FSCN1 promoter as a potential regulatory mechanism in FSCN1 expression. The samples included papillary thyroid carcinoma, follicular thyroid carcinoma and goiter samples (controls). Methylation of FSCN1 promoter was observed in 50% of follicular, 48.6% of papillary and 60% of controls. The promoter was unmethylated in 16.7% of follicular samples, 5.7% of papillary samples and 26.7% of controls. In the remaining 33.3% of follicular and 45.7% of papillary samples as well as 13.3% of controls, both methylated and unmethylated alleles were amplified, a condition referred to as semi-methylation. The results showed that FSCN1 promoter was significantly hypomethylated in papillary cases while the methylation status was not significantly altered in follicular cases. On the other hand, FSCN1 was expressed in only nine papillary samples. Regarding protein expression and methylation status, we suggest that hypomethylation of FSCN1 promoter in papillary thyroid carcinoma does not lead to overexpression of FSCN1 and that there might be other regulatory mechanisms involved in FSCN1 up-regulation.
Collapse
Affiliation(s)
- Mojdeh Mahdiannasser
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Poursina St, Tehran, Iran
| | - Vahid Haghpanah
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Poursina St, District 6, Tehran, Tehran Province, Iran.,Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Elia Damavandi
- Specialized Medical Genetic Center (SMGC) of ACECR, 4th floor, No 65, Aboureihan St, Enghelab Ave., Tehran, Iran.,Department of Photo Healing and Regeneration, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Majid Kabuli
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Poursina St, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Jalal Al Ahmad Junction, Karegar Shomali St, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Poursina St, District 6, Tehran, Tehran Province, Iran
| | - Mohsen Ghadami
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Poursina St, Tehran, Iran. .,Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Poursina St, District 6, Tehran, Tehran Province, Iran. .,Cardiac Primary Research Center, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Hognon C, Besancenot V, Gruez A, Grandemange S, Monari A. Cooperative Effects of Cytosine Methylation on DNA Structure and Dynamics. J Phys Chem B 2019; 123:7365-7371. [PMID: 31365827 DOI: 10.1021/acs.jpcb.9b05835] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The behavior of the structural parameters of DNA considering different levels of methylation in CpG islands is studied by means of full-atom molecular dynamics simulations and electronic circular dichroism, both in an artificial model system and in a gene promoter sequence. It is demonstrated that methylation although intrinsically brings quite local perturbations may, if its level is high enough, induce cooperative effects that strongly modify the DNA backbone torsional parameters altering the helicity as compared to the nonmethylated case. Because methylation of the CpG island is correlated with the regulation of gene expression, understanding the structural modifications induced in DNA is crucial to characterize all the fine equilibria into play in epigenetics phenomena.
Collapse
Affiliation(s)
- Cécilia Hognon
- Université de Lorraine and CNRS, UMR 7019 LPCT , F-54000 Nancy , France
| | | | - Arnaud Gruez
- Université de Lorraine and CNRS, UMR 7356 IMOPA , F-54000 Nancy , France
| | | | - Antonio Monari
- Université de Lorraine and CNRS, UMR 7019 LPCT , F-54000 Nancy , France
| |
Collapse
|
11
|
Wang J, Wen T, Li Z, Che X, Gong L, Yang X, Zhang J, Tang H, He L, Qu X, Liu Y. MicroRNA-1224 Inhibits Tumor Metastasis in Intestinal-Type Gastric Cancer by Directly Targeting FAK. Front Oncol 2019; 9:222. [PMID: 31019895 PMCID: PMC6458237 DOI: 10.3389/fonc.2019.00222] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 03/13/2019] [Indexed: 12/20/2022] Open
Abstract
Intestinal-type gastric cancer (GC) of the Lauren classification system has specific epidemiological characteristics and carcinogenesis patterns. MicroRNAs (miRNAs) have prognostic significance, and some can be used as prognostic biomarkers in GC. In this study, we identified miR-1224 as a potential survival-related miRNA in intestinal-type GC patients by The Cancer Genome Atlas (TCGA) analysis. Using quantitative real-time PCR (qRT-PCR), we showed that the relative expression of miR-1224 was significantly decreased in intestinal-type GC tissues compared to matched adjacent normal mucosa tissues (p < 0.01). We found that high miR-1224 expression was associated with no lymph-node metastasis (p < 0.05) and good prognosis (p = 0.028) in 90 intestinal-type GC tissues. Transfection of intestinal-type GC cells with miR-1224 mimics showed that miR-1224 suppressed cell migration in vitro (wound healing assay and Transwell migration assay), whereas the transfection of cells with miR-1224 inhibitor promoted cell migration in vitro. miR-1224 also suppressed intestinal-type GC cell metastasis in a xenograft mouse model. Furthermore, bioinformatics, luciferase reporter, Western blotting, and immunohistochemistry (IHC) studies demonstrated that miR-1224 directly bound to the focal adhesion kinase (FAK) gene, and downregulated its expression, which decreased STAT3 and NF-κB signaling and subsequent the epithelial-to-mesenchymal transition (EMT). Repression of FAK is required for the miR-1224-mediated inhibition of cell migration in intestinal-type GC. The present study demonstrated that miR-1224 is downregulated in intestinal-type GC. miR-1224 inhibits the metastasis of intestinal-type GC by suppressing FAK-mediated activation of the STAT3 and NF-κB pathways, and subsequent EMT. miR-1224 could represent an important prognostic factor in intestinal-type GC.
Collapse
Affiliation(s)
- Jin Wang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Ti Wen
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Zhi Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Libao Gong
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Xianghong Yang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jingdong Zhang
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Huali Tang
- Department of Medical Oncology, The Central Hospital of Zhuanghe, Zhuanghe, China
| | - Lingzi He
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Xiujuan Qu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| | - Yunpeng Liu
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.,Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Rudloff U. Gastric adenocarcinoma and proximal polyposis of the stomach: diagnosis and clinical perspectives. Clin Exp Gastroenterol 2018; 11:447-459. [PMID: 30584346 PMCID: PMC6284852 DOI: 10.2147/ceg.s163227] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS) is a recently described, rare gastric polyposis syndrome. It is characterized by extensive involvement of the fundus and body of the stomach with fundic gland polyps sparing the antrum and lesser curvature, an autosomal dominant inheritance pattern with incomplete penetrance, and a significant predisposition for the development of gastric adenocarcinoma. Due to the recent discovery of APC promotor IB mutations (c.-191T>C, c.-192A>G, and c.-195A>C), which reduce binding of the transcription factor Yin Yang 1 (YY1) and transcriptional activity of the promotor, as its underlying genetic perturbation, GAPPS has been added to the growing molecular class of APC-associated disorders. Recent reports on family members afflicted by gastric polyposis due to GAPPS have described the development of metastatic cancer or the presence of invasive gastric adenocarcinoma in total gastrectomy specimens after variable periods of endoscopic surveillance emphasizing the need for an improved understanding of the to-date poorly characterized natural history of the syndrome. There are, however, currently no guidelines on screening, timing of prophylactic gastrectomy, or endoscopic surveillance for GAPPS available. In this review, we summarize the clinical, pathological, and genetic aspects of GAPPS as well as management approaches to this rare cancer predisposition syndrome, highlighting the need for early recognition, a multidisciplinary approach, and the creation of prospective family registries and consensus guidelines in the near future.
Collapse
Affiliation(s)
- Udo Rudloff
- Rare Tumor Initiative, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA,
| |
Collapse
|
13
|
Zhang J, Shen Z, Liu H, Liu S, Shu W. Diagnostic potential of methylated DAPK in brushing samples of nasopharyngeal carcinoma. Cancer Manag Res 2018; 10:2953-2964. [PMID: 30214290 PMCID: PMC6118259 DOI: 10.2147/cmar.s171796] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background The death-associated protein kinase (DAPK) gene is an important member of the apoptotic pathway and is inactivated by abnormal methylation in numerous cancers, including nasopharyngeal carcinoma (NPC). However, the diagnostic value of DAPK methylation in brushing samples and tissue samples of NPC remains unclear. Methods We conducted a systematic meta-analysis based on 17 studies (including 386 tissue cases, 233 brushing cases, and 296 blood cases). Results Our results revealed an association between methylated DAPK and increased risk of NPC in blood, brushing, and tissue samples. In addition, the comparison of the pooled sensitivity, specificity, and area under the curve of methylated DAPK in brushing and tissue samples demonstrated the non-inferior effectiveness of methylated DAPK in brushing samples to monitor the development of NPC.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, People's Republic of China
| | - Zhisen Shen
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, People's Republic of China
| | - Huigao Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Zhenhai Longsai Hospital, Ningbo, People's Republic of China
| | - Shuai Liu
- Department of Radiology, Ningbo Medical Center Lihuili Eastern Hospital, Ningbo, People's Republic of China
| | - Wenxiu Shu
- Department of Oncology and Hematology, Ningbo Medical Center Lihuili Eastern Hospital, Ningbo, People's Republic of China,
| |
Collapse
|
14
|
Yamagishi M, Fujikawa D, Watanabe T, Uchimaru K. HTLV-1-Mediated Epigenetic Pathway to Adult T-Cell Leukemia-Lymphoma. Front Microbiol 2018; 9:1686. [PMID: 30087673 PMCID: PMC6066519 DOI: 10.3389/fmicb.2018.01686] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 07/06/2018] [Indexed: 11/13/2022] Open
Abstract
Human T-cell leukemia virus type 1 (HTLV-1), the first reported human oncogenic retrovirus, is the etiologic agent of highly aggressive, currently incurable diseases such as adult T-cell leukemia-lymphoma (ATL) and HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP). HTLV-1 proteins, including Tax and HBZ, have been shown to have critical roles in HTLV-1 pathogenicity, yet the underlying mechanisms of HTLV-1-driven leukemogenesis are unclear. The frequent disruption of genetic and epigenetic gene regulation in various types of malignancy, including ATL, is evident. In this review, we illustrate a focused range of topics about the establishment of HTLV-1 memory: (1) genetic lesion in the Tax interactome pathway, (2) gene regulatory loop/switch, (3) disordered chromatin regulation, (4) epigenetic lock by the modulation of epigenetic factors, (5) the loss of gene fine-tuner microRNA, and (6) the alteration of chromatin regulation by HTLV-1 integration. We discuss the persistent influence of Tax-dependent epigenetic changes even after the disappearance of HTLV-1 gene expression due to the viral escape from the immune system, which is a remaining challenge in HTLV-1 research. The summarized evidence and conceptualized description may provide a better understanding of HTLV-1-mediated cellular transformation and the potential therapeutic strategies to combat HTLV-1-associated diseases.
Collapse
Affiliation(s)
- Makoto Yamagishi
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Dai Fujikawa
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshiki Watanabe
- The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kaoru Uchimaru
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
15
|
Cao Y, Song J, Chen J, Xiao J, Ni J, Wu C. Overexpression of NEK3 is associated with poor prognosis in patients with gastric cancer. Medicine (Baltimore) 2018; 97:e9630. [PMID: 29504992 PMCID: PMC5779761 DOI: 10.1097/md.0000000000009630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The NIMA-related kinase 3 (NEK3) plays an important role in cell migration, cell proliferation, and cell viability. Recently, NEK3 was reported to enhance the malignancy of breast cancer. However, its role in gastric cancer has not been completely characterized. In this study, we explored the prognostic significance of NEK3 in human gastric cancer. Reverse transcription-polymerase chain reaction and western blot were performed to detect the NEK3 mRNA and protein expression in 6 paired fresh human gastric cancer tissues and surrounding normal tissues. NEK3 levels in gastric cancer and its adjacent normal samples of 168 cases were detected by immunohistochemistry, and the relationships between the NEK3 level and various clinicopathological features were analyzed. NEK3 mRNA and protein were significantly overexpressed in gastric cancer tissues, compared with adjacent normal tissues. Immunohistochemistry staining assay showed the percentage of high NEK3 expression in gastric cancer samples was higher than that in adjacent normal samples. NEK3 overexpression was significantly correlated with pT stage, pathologic TNM stage, lymph node metastasis, and poor prognosis of gastric cancer. Cox multivariate regression analyses suggested that NEK3 was an independent prognostic factor for survival of patients with gastric cancer. The data demonstrate that NEK3 is overexpressed in gastric cancer, which promotes the malignancy of gastric cancer. NEK3 may be as a prognostic biomarker and a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Yongfeng Cao
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou
- Department of Oncology, The Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jiaye Song
- Department of Oncology, The Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jia Chen
- Department of Oncology, The Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jinzhang Xiao
- Department of Oncology, The Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jingyi Ni
- Department of Oncology, The Affiliated Tumor Hospital of Nantong University, Nantong, Jiangsu, China
| | - Changping Wu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou
| |
Collapse
|
16
|
Ayala-Calvillo E, Mojica-Vázquez LH, García-Carrancá A, González-Maya L. Wnt/β‑catenin pathway activation and silencing of the APC gene in HPV‑positive human cervical cancer‑derived cells. Mol Med Rep 2017; 17:200-208. [PMID: 29115417 PMCID: PMC5780127 DOI: 10.3892/mmr.2017.7853] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/06/2017] [Indexed: 01/21/2023] Open
Abstract
Although persistent infections with high-risk human papilloma virus (HPV) constitute the most significant cofactor for the development of cervical cancer, they are insufficient on their own. Mutations or epigenetic inactivation of the tumor suppressor adenomatous polyposis coli (APC), the two acting as prominent oncogenic mechanisms in a number of types of cancer, are frequently associated with aberrant activation of the Wnt/β-catenin pathway. According to these observations, it was hypothesized that APC alteration may lead to β-catenin deregulation and the abnormal expression of direct targets of the Wnt pathway in HPV-infected cervical cancer cells. The present study confirmed that the stabilization of β-catenin correlates with enhanced transcriptional activity of the β-catenin/T-cell factor complex in cervical cancer cell lines. Sequence analysis of the ‘hot-spot’ in the mutation cluster region did not exhibit genetic alterations that may be associated with APC gene inactivation. In addition, it was identified that there was a good correlation with the hypermethylation status of the APC promoter 1A and the abnormal accumulation of endogenous β-catenin in cell lines and biopsies infected with HPV16, although not HPV18. Removal of the epigenetic markers led to an increase in APC levels and a reduction of β-catenin expression in two transcriptional targets of the Wnt pathway: Matrix metalloproteinase-7 and vascular endothelial growth factor. The present study suggested that the increase in Wnt activity in certain cervical cancer-derived cells may be associated with an alteration in the methylation status of the APC gene promoter 1A.
Collapse
Affiliation(s)
- Erick Ayala-Calvillo
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, CP 62209, México
| | - Luis Humberto Mojica-Vázquez
- Centro de Ciencias de la Atmósfera, Genotoxicología Ambiental‑Universidad Nacional Autónoma de México, CDMX, CP 14080, México
| | - Alejandro García-Carrancá
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas‑Universidad Nacional Autónoma de México, CDMX, CP 14080, México
| | - Leticia González-Maya
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, CP 62209, México
| |
Collapse
|
17
|
Wang T, Liu Y, Zhao M. Mutational analysis of driver genes with tumor suppressive and oncogenic roles in gastric cancer. PeerJ 2017; 5:e3585. [PMID: 28729958 PMCID: PMC5516769 DOI: 10.7717/peerj.3585] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/26/2017] [Indexed: 12/18/2022] Open
Abstract
Gastric cancer (GC) is a complex disease with heterogeneous genetic mechanisms. Genomic mutational profiling of gastric cancer not only expands our knowledge about cancer progression at a fundamental genetic level, but also could provide guidance on new treatment decisions, currently based on tumor histology. The fact that precise medicine-based treatment is successful in a subset of tumors indicates the need for better identification of clinically related molecular tumor phenotypes, especially with regard to those driver mutations on tumor suppressor genes (TSGs) and oncogenes (ONGs). We surveyed 313 TSGs and 160 ONGs associated with 48 protein coding and 19 miRNA genes with both TSG and ONG roles. Using public cancer mutational profiles, we confirmed the dual roles of CDKN1A and CDKN1B. In addition to the widely recognized alterations, we identified another 82 frequently mutated genes in public gastric cancer cohort. In summary, these driver mutation profiles of individual GC will form the basis of personalized treatment of gastric cancer, leading to substantial therapeutic improvements.
Collapse
Affiliation(s)
- Tianfang Wang
- School of Science and Engineering, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydore DC, Australia
| | - Yining Liu
- The School of Public Health, Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou, China
| | - Min Zhao
- School of Science and Engineering, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, Maroochydore DC, Australia
| |
Collapse
|
18
|
Li BQ, Liu PP, Zhang CH. Correlation between the methylation of APC gene promoter and colon cancer. Oncol Lett 2017; 14:2315-2319. [PMID: 28781669 PMCID: PMC5530209 DOI: 10.3892/ol.2017.6455] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/08/2017] [Indexed: 11/21/2022] Open
Abstract
The present study was planned to explore the correlation between the methylation of APC (adenomatous polyposis coli) and colon carcinogenesis. Colon cancer tissues and tumor-adjacent normal tissues of 60 colon cancer patients (who received surgical operation in our hospital from January 2012 to December 2014) were collected. SW1116 cells in human colon cancer tissues were selected for culturing. 5-aza-2c-deoxycytidine (5-aza-dC) was utilized as an inhibitor of the methylation for APC gene. Methylation specific PCR (MSP) was utilized for detection of APC methylation in SW1116 cells. The MTT and Transwell assays were performed to detect the effect of the methylation of APC gene on the proliferation and invasive abilities of SW1116 cells. The correlation between the methylation of APC gene and pathological parameters of colon cancer patients was analyzed. MSP results revealed that 41 cases (68.33%) showed methylation of APC gene in colon cancer tissues. No methylation of APC gene was found in tumor-adjacent normal tissues. 5-aza-dC was able to inhibit the methylation of APC gene in SW1116 cells. APC gene methylation was correlated with tumor size, differentiation degree, lymph node metastasis and Dukes staging. In conclusion, the levels of the methylation of APC in colon cancer tissues and SW1116 cells are relatively high. The methylation of APC promoted the proliferation and invasion abilities of SW1116 cells. Furthermore, methylation is correlated with a variety of clinicopathological features of colon cancer patients.
Collapse
Affiliation(s)
- Bing-Qiang Li
- Department of Gastrointestinal Surgery, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| | - Peng-Peng Liu
- Department of Hepatobiliary Surgery, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| | - Cai-Hua Zhang
- Department of Gastrointestinal Surgery, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
19
|
Tekcham DS, Poojary SS, Bhunia S, Barbhuiya MA, Gupta S, Shrivastav BR, Tiwari PK. Epigenetic regulation of APC in the molecular pathogenesis of gallbladder cancer. Indian J Med Res 2017; 143:S82-S90. [PMID: 27748282 PMCID: PMC5080933 DOI: 10.4103/0971-5916.191792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background & objectives: Loss of function of adenomatous polyposis coli (APC) has been reported in cancer. The two promoters of APC, 1A and 1B also have roles in cancer. But, the epigenetic role of APC promoters is not yet clear in gallbladder cancer (GBC) and gallstone diseases (GSD). We undertook this study to determine the epigenetic role of APC in GBC and GSD. Methods: Methylation-specific (MS)-PCR was used to analyze the methylation of APC gene. The expression of APC gene was studied by semi-quantitative PCR, real-time PCR and immunohistochemistry (IHC) in GBC, GSD and adjacent normal tissues. Results: Of the two promoters, APC 1A promoter was found methylated in 96 per cent GBC (P=0.0155) and 80 per cent GSD (P=0.015). Exon 1 was downregulated in grade II (P=0.002) and grade III (P=0.0001) of GBC, while exon 2 was normally expressed. Scoring analysis of IHC revealed 0 or negativity in 34.48 per cent (P=0.057) and 1+ in 24.14 per cent (P=0.005) GBC cases suggesting loss of APC expression. Interpretation & conclusions: The present findings indicate epigenetic silencing of APC in advanced GBC. The methylation pattern, followed by expression analysis of APC may be suggested for diagnostic, prognostic and therapeutic purposes in GBC in future.
Collapse
Affiliation(s)
- Dinesh Singh Tekcham
- Centre for Genomics; School of Studies in Zoology, Jiwaji University, Gwalior, Madhya Pradesh, India
| | - Satish S Poojary
- Centre for Genomics, Jiwaji University, Gwalior, Madhya Pradesh, India
| | - Shushruta Bhunia
- Centre for Genomics, Jiwaji University, Gwalior, Madhya Pradesh, India
| | | | - Sanjeev Gupta
- Department of Pathology, Cancer Hospital and Research Institute, Gwalior, Madhya Pradesh, India
| | - Braj Raj Shrivastav
- Department of Surgical Oncology, Cancer Hospital and Research Institute; Department of Surgery, Gajra Raja Medical College, Gwalior, Madhya Pradesh, India
| | - Pramod Kumar Tiwari
- Centre for Genomics; School of Studies in Zoology, Jiwaji University, Gwalior, Madhya Pradesh, India
| |
Collapse
|
20
|
Xing X, Cai W, Ma S, Wang Y, Shi H, Li M, Jiao J, Yang Y, Liu L, Zhang X, Chen M. Down-regulated expression of OPCML predicts an unfavorable prognosis and promotes disease progression in human gastric cancer. BMC Cancer 2017; 17:268. [PMID: 28407749 PMCID: PMC5391589 DOI: 10.1186/s12885-017-3203-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 03/17/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND OPCML belongs to the IgLON family of Ig domain-containing GPI-anchored cell adhesion molecules and was recently found to be involved in carcinogenesis, while its role in gastric cancer remains unclear. METHODS We assessed expression and biological behavior of OPCML in gastric cancer. RESULTS OPCML expression was markedly reduced in tumor tissues and cancer cell lines. Decreased OPCML expression had a significant association with unfavorable tumor stage (p = 0.007) and grading (p < 0.001). Furthermore, the results revealed that OPCML was an independent prognostic factor for overall survival in gastric cancer (p = 0.002). In addition, ectopic expression of OPCML in cancer cells significantly inhibited cell viability (p < 0.01) and colony formation (p < 0.001), arrest cell cycle in G0/G1 phase and induced apoptosis, and suppressed tumor formation in nude mice. The alterations of phosphorylation status of AKT and its substrate GSK3β, up-regulation of pro-apoptotic regulators including caspase-3, caspase-9 and PARP, and up-regulation of cell cycle regulator p27, were implicated in the biological activity of OPCML in cancer cells. CONCLUSION Down-regulated OPCML expression might serve as an independent predictor for unfavorable prognosis of patients, and the biological behavior supports its role as a tumor suppressor in gastric cancer.
Collapse
Affiliation(s)
- Xiangbin Xing
- Department of Gastroenterology, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan II Road, Guangzhou, 510080 China
| | - Weibin Cai
- Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, 510089 China
| | - Sanmei Ma
- Department of Biotechnology, Jinan University, Guangzhou, 510632 China
| | - Yongfei Wang
- Department of Biotechnology, Jinan University, Guangzhou, 510632 China
| | - Huijuan Shi
- Department of Pathology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Ming Li
- Department of Gastroenterology, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan II Road, Guangzhou, 510080 China
- Department of Biotechnology, Jinan University, Guangzhou, 510632 China
| | - Jinxia Jiao
- Department of Gastroenterology, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan II Road, Guangzhou, 510080 China
- Department of Biotechnology, Jinan University, Guangzhou, 510632 China
| | - Yang Yang
- Department of Gastroenterology, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan II Road, Guangzhou, 510080 China
- Department of Biotechnology, Jinan University, Guangzhou, 510632 China
| | - Longshan Liu
- Department of Laboratory of General Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080 China
| | - Xiangliang Zhang
- Department of Abdominal Surgery (Section 2), Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, 510095 China
| | - Minhu Chen
- Department of Gastroenterology, the First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan II Road, Guangzhou, 510080 China
| |
Collapse
|
21
|
Debouki-Joudi S, Trifa F, Khabir A, Sellami-Boudawara T, Frikha M, Daoud J, Mokdad-Gargouri R. CpG methylation of APC promoter 1A in sporadic and familial breast cancer patients. Cancer Biomark 2017; 18:133-141. [DOI: 10.3233/cbm-160005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Saoussen Debouki-Joudi
- Laboratory of Molecular Biotechnology of Eukaryotes, Department of Cancer Genetics, Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Fatma Trifa
- Laboratory of Molecular Biotechnology of Eukaryotes, Department of Cancer Genetics, Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | | | | | - Mounir Frikha
- Centre Hospitalo-Universitaire Habib Bourguiba, Sfax 3000, Tunisia
| | - Jamel Daoud
- Centre Hospitalo-Universitaire Habib Bourguiba, Sfax 3000, Tunisia
| | - Raja Mokdad-Gargouri
- Laboratory of Molecular Biotechnology of Eukaryotes, Department of Cancer Genetics, Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
22
|
Wang H, Duan XL, Qi XL, Meng L, Xu YS, Wu T, Dai PG. Concurrent Hypermethylation of SFRP2 and DKK2 Activates the Wnt/β-Catenin Pathway and Is Associated with Poor Prognosis in Patients with Gastric Cancer. Mol Cells 2017; 40:45-53. [PMID: 28152305 PMCID: PMC5303888 DOI: 10.14348/molcells.2017.2245] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/15/2016] [Accepted: 12/20/2016] [Indexed: 11/27/2022] Open
Abstract
Aberrant hypermethylation of Wnt antagonists has been observed in gastric cancer. A number of studies have focused on the hypermethylation of a single Wnt antagonist and its role in regulating the activation of signaling. However, how the Wnt antagonists interacted to regulate the signaling pathway has not been reported. In the present study, we systematically investigated the methylation of some Wnt antagonist genes (SFRP2, SFRP4, SFRP5, DKK1, DKK2, and APC) and their regulatory role in carcinogenesis. We found that aberrant promoter methylation of SFRP2, SFRP4, DKK1, and DKK2 was significantly increased in gastric cancer. Moreover, concurrent hypermethylation of SFRP2 and DKK2 was observed in gastric cancer and this was significantly associated with increased expression of β-catenin, indicating that the joint inactivation of these two genes promoted the activation of the Wnt signaling pathway. Further analysis using a multivariate Cox proportional hazards model showed that DKK2 methylation was an independent prognostic factor for poor overall survival, and the predictive value was markedly enhanced when the combined methylation status of SFRP2 and DKK2 was considered. In addition, the methylation level of SFRP4 and DKK2 was correlated with the patient's age and tumor differentiation, respectively. In conclusion, epigenetic silencing of Wnt antagonists was associated with gastric carcinogenesis, and concurrent hypermethylation of SFRP2 and DKK2 could be a potential marker for a prognosis of poor overall survival.
Collapse
Affiliation(s)
- Hao Wang
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi’an, Shaanxi,
China
| | - Xiang-Long Duan
- Second Department of General Surgery, Shaanxi Provincial People’s Hospital, Xi’an, Shaanxi,
China
- Department of General Surgery, The First Hospital of Yulin, Yulin, Shaanxi,
China
| | - Xiao-Li Qi
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi’an, Shaanxi,
China
| | - Lei Meng
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi’an, Shaanxi,
China
- Department of Surgical Oncology, The First Affiliated Hospital of Xi’an Jiaotong University. Xi’an, Shaanxi,
China
| | - Yi-Song Xu
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi’an, Shaanxi,
China
| | - Tong Wu
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi’an, Shaanxi,
China
| | - Peng-Gao Dai
- National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi’an, Shaanxi,
China
| |
Collapse
|
23
|
Song X, Xin N, Wang W, Zhao C. Wnt/β-catenin, an oncogenic pathway targeted by H. pylori in gastric carcinogenesis. Oncotarget 2016; 6:35579-88. [PMID: 26417932 PMCID: PMC4742126 DOI: 10.18632/oncotarget.5758] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 08/26/2015] [Indexed: 12/14/2022] Open
Abstract
A section of gastric cancers presents nuclear β-catenin accumulation correlated with H. pylori infection. H. pylori stimulate Wnt/β-catenin pathway by activating oncogenic c-Met and epidermal growth factor receptor (EGFR), or by inhibiting tumor suppressor Runx3 and Trefoil factor 1 (TFF1). H. pylori also trigger Wnt/β-catenin pathway by recruiting macrophages. Moreover, Wnt/β-catenin pathway is found involved in H. pylori-induced gastric cancer stem cell generation. Recently, by using gastroids, researchers have further revealed that H. pylori induce gastric epithelial cell proliferation through β-catenin. These findings indicate that Wnt/β-catenin is an oncogenic pathway activated by H. pylori. Therefore, this pathway is a potential therapy target for H. pylori-related gastric cancer.
Collapse
Affiliation(s)
- Xiaowen Song
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Na Xin
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Wei Wang
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Chenghai Zhao
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China
| |
Collapse
|
24
|
Tekcham DS, Tiwari PK. Epigenetic regulation in gallbladder cancer: Promoter methylation profiling as emergent novel biomarkers. Asia Pac J Clin Oncol 2016; 12:332-348. [PMID: 27385126 DOI: 10.1111/ajco.12507] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 03/27/2016] [Accepted: 04/05/2016] [Indexed: 12/12/2022]
Abstract
DNA methylation, once considered to rule the sex determination in Mary Lyon's hypothesis, has now reached the epicenter of human diseases, from monogenic (e.g. Prader Willi syndrome, Angelman syndromes and Beckwith-Wiedemann syndrome) to polygenic diseases, like cancer. Technological developments from gold standard to high throughput technologies have made tremendous advancement to define the epigenetic mechanism of cancer. Gallbladder cancer (GBC) is a fatal health issue affecting mostly the middle-aged women, whose survival rate is very low due to late symptomatic diagnosis. DNA methylation has become one of the key molecular mechanisms in the tumorigenesis of gallbladder. Various molecules have been reported to be epigenetically altered in GBC. In this review, we have discussed the classes of epigenetics, an overview of DNA methylation, technological approaches for its study, profile of methylated genes, their likely roles in GBC, future prospects of biomarker development and other discovery approaches, including therapeutics.
Collapse
Affiliation(s)
- Dinesh Singh Tekcham
- Centre for Genomics, Molecular and Human Genetics, Jiwaji University, Gwalior, Madhya Pradesh, India
| | - Pramod Kumar Tiwari
- Centre for Genomics, Molecular and Human Genetics, Jiwaji University, Gwalior, Madhya Pradesh, India
| |
Collapse
|
25
|
Liu L, Cao L, Gong B, Yu J. Novel biomarkers for the identification and targeted therapy of gastric cancer. Expert Rev Gastroenterol Hepatol 2016. [PMID: 26220043 DOI: 10.1586/17474124.2015.1072466] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gastric cancer development follows the pathologic pattern such that chronic inflammation in the gastric mucosa progressively transforms normal mucosa into atrophy, intestinal metaplasia, adenoma/dysplasia and eventually invasive and metastatic tumors. The accumulation of multiple genetic and epigenetic alterations leads to the dysregulation of oncogenes and tumor suppressors, which was considered as the driver behind events during the tumorigenesis. Almost all gastric cancers are adenocarcinomas, which share considerable heterogeneity with distinct morphology, pathogenesis and clinical behavior. Therefore, identifying subtypes of gastric cancers with molecular and genetic features will be beneficial for the early identification and selection of new effective agents for targeted treatment. High-throughput sequencing techniques such as whole genomic, epigenome and transcriptome sequencing and proteomics platforms have identified major genomic characteristics that exhibit identification and prognostic impacts and distinct response patterns. In this article, the authors aim to summarize the information regarding the most promising molecules that may have clinical application as non-invasive biomarkers and therapy targets.
Collapse
Affiliation(s)
- Lei Liu
- a 1 Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | |
Collapse
|
26
|
Li J, Woods SL, Healey S, Beesley J, Chen X, Lee JS, Sivakumaran H, Wayte N, Nones K, Waterfall JJ, Pearson J, Patch AM, Senz J, Ferreira MA, Kaurah P, Mackenzie R, Heravi-Moussavi A, Hansford S, Lannagan TRM, Spurdle AB, Simpson PT, da Silva L, Lakhani SR, Clouston AD, Bettington M, Grimpen F, Busuttil RA, Di Costanzo N, Boussioutas A, Jeanjean M, Chong G, Fabre A, Olschwang S, Faulkner GJ, Bellos E, Coin L, Rioux K, Bathe OF, Wen X, Martin HC, Neklason DW, Davis SR, Walker RL, Calzone KA, Avital I, Heller T, Koh C, Pineda M, Rudloff U, Quezado M, Pichurin PN, Hulick PJ, Weissman SM, Newlin A, Rubinstein WS, Sampson JE, Hamman K, Goldgar D, Poplawski N, Phillips K, Schofield L, Armstrong J, Kiraly-Borri C, Suthers GK, Huntsman DG, Foulkes WD, Carneiro F, Lindor NM, Edwards SL, French JD, Waddell N, Meltzer PS, Worthley DL, Schrader KA, Chenevix-Trench G. Point Mutations in Exon 1B of APC Reveal Gastric Adenocarcinoma and Proximal Polyposis of the Stomach as a Familial Adenomatous Polyposis Variant. Am J Hum Genet 2016; 98:830-842. [PMID: 27087319 DOI: 10.1016/j.ajhg.2016.03.001] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 03/02/2016] [Indexed: 12/15/2022] Open
Abstract
Gastric adenocarcinoma and proximal polyposis of the stomach (GAPPS) is an autosomal-dominant cancer-predisposition syndrome with a significant risk of gastric, but not colorectal, adenocarcinoma. We mapped the gene to 5q22 and found loss of the wild-type allele on 5q in fundic gland polyps from affected individuals. Whole-exome and -genome sequencing failed to find causal mutations but, through Sanger sequencing, we identified point mutations in APC promoter 1B that co-segregated with disease in all six families. The mutations reduced binding of the YY1 transcription factor and impaired activity of the APC promoter 1B in luciferase assays. Analysis of blood and saliva from carriers showed allelic imbalance of APC, suggesting that these mutations lead to decreased allele-specific expression in vivo. Similar mutations in APC promoter 1B occur in rare families with familial adenomatous polyposis (FAP). Promoter 1A is methylated in GAPPS and sporadic FGPs and in normal stomach, which suggests that 1B transcripts are more important than 1A in gastric mucosa. This might explain why all known GAPPS-affected families carry promoter 1B point mutations but only rare FAP-affected families carry similar mutations, the colonic cells usually being protected by the expression of the 1A isoform. Gastric polyposis and cancer have been previously described in some FAP-affected individuals with large deletions around promoter 1B. Our finding that GAPPS is caused by point mutations in the same promoter suggests that families with mutations affecting the promoter 1B are at risk of gastric adenocarcinoma, regardless of whether or not colorectal polyps are present.
Collapse
Affiliation(s)
- Jun Li
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Susan L Woods
- School of Medicine, University of Adelaide and Cancer Theme, SAHMRI, Adelaide, SA 5000, Australia
| | - Sue Healey
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Jonathan Beesley
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Xiaoqing Chen
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Jason S Lee
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Haran Sivakumaran
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Nicci Wayte
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Katia Nones
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Joshua J Waterfall
- Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - John Pearson
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Anne-Marie Patch
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Janine Senz
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Manuel A Ferreira
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Pardeep Kaurah
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Robertson Mackenzie
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | | | - Samantha Hansford
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 2B5, Canada
| | - Tamsin R M Lannagan
- School of Medicine, University of Adelaide and Cancer Theme, SAHMRI, Adelaide, SA 5000, Australia
| | - Amanda B Spurdle
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Peter T Simpson
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; School of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Leonard da Silva
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; School of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia
| | - Sunil R Lakhani
- UQ Centre for Clinical Research, The University of Queensland, Brisbane, QLD 4029, Australia; School of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia; Anatomical Pathology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, QLD 4029, Australia
| | - Andrew D Clouston
- Centre for Liver Disease Research, TRI Building, University of Queensland, Woolloongabba, QLD 4102, Australia; Envoi Specialist Pathologists, Bishop Street, Kelvin Grove, QLD 4059, Australia
| | - Mark Bettington
- School of Medicine, The University of Queensland, Brisbane, QLD 4006, Australia; Envoi Specialist Pathologists, Bishop Street, Kelvin Grove, QLD 4059, Australia; The Conjoint Gastroenterology Laboratory, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Florian Grimpen
- Departments of Gastroenterology and Hepatology, Royal Brisbane and Women's Hospital, Brisbane, QLD 4006, Australia
| | - Rita A Busuttil
- Cancer Genetics and Genomics Laboratory, Peter MacCallum Cancer Centre, Locked Bag 1, Melbourne, VIC 8006, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Natasha Di Costanzo
- Cancer Genetics and Genomics Laboratory, Peter MacCallum Cancer Centre, Locked Bag 1, Melbourne, VIC 8006, Australia
| | - Alex Boussioutas
- Cancer Genetics and Genomics Laboratory, Peter MacCallum Cancer Centre, Locked Bag 1, Melbourne, VIC 8006, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Gastroenterology, Royal Melbourne Hospital, Parkville, VIC 3010, Australia
| | - Marie Jeanjean
- Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - George Chong
- Molecular Pathology Centre, Department of Pathology, Jewish General Hospital - McGill University, Montreal, QC H3T 1E2, Canada
| | - Aurélie Fabre
- AP-HM Timone, Medical Genetics Department, 13385 Marseille, France; Aix Marseille Université, INSERM, GMGF UMR_S 910, 13385 Marseille, France; Oncology Unit, Generale de Sante, Clairval Hospital, 13009 Marseille, France
| | - Sylviane Olschwang
- AP-HM Timone, Medical Genetics Department, 13385 Marseille, France; Aix Marseille Université, INSERM, GMGF UMR_S 910, 13385 Marseille, France; Oncology Unit, Generale de Sante, Clairval Hospital, 13009 Marseille, France
| | - Geoffrey J Faulkner
- Mater Research Institute, University of Queensland, TRI Building, Woolloongabba, QLD 4102, Australia
| | - Evangelos Bellos
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia; Department of Genomics of Common Disease, Imperial College London, London W12 0NN, UK
| | - Lachlan Coin
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Kevin Rioux
- Department of Medicine, Division of Gastroenterology, Department of Microbiology and Infectious Diseases, Gastrointestinal Research Group, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Oliver F Bathe
- Departments of Surgery and Oncology, University of Calgary, Calgary, AB T2N 4N1, Canada; Division of Surgical Oncology, Tom Baker Cancer Centre, 1331 29(th) St NW, Calgary, AB T2N 4N1, Canada
| | - Xiaogang Wen
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP)/Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto 4200-135, Portugal; Centro Hospitalar Vila Nova de Gaia/Espinho, Porto 4430-027, Portugal
| | - Hilary C Martin
- Wellcome Trust Centre for Human Genetics, Oxford OX3 7BN, UK
| | - Deborah W Neklason
- Department of Internal Medicine, Huntsman Cancer Institute at University of Utah, Salt Lake City, UT 84112, USA
| | - Sean R Davis
- Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Robert L Walker
- Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Kathleen A Calzone
- Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Itzhak Avital
- Department of Surgery, Saint Peter's University Hospital, Rutgers University, New Brunswick, NJ 08901, USA
| | - Theo Heller
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Disease (NIDDK), NIH, Bethesda, MD 20892, USA
| | - Christopher Koh
- Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Disease (NIDDK), NIH, Bethesda, MD 20892, USA
| | - Marbin Pineda
- Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Udo Rudloff
- Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Martha Quezado
- Laboratory of Pathology, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Pavel N Pichurin
- Department of Medical Genetics, Mayo Clinic, Rochester, MN 55905, USA
| | - Peter J Hulick
- Center for Medical Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | | | - Anna Newlin
- Center for Medical Genetics, NorthShore University HealthSystem, Evanston, IL 60201, USA
| | - Wendy S Rubinstein
- National Center for Biotechnology Information (NCBI), National Library of Medicine (NLM), NIH, Bethesda, MD 20892, USA
| | - Jone E Sampson
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Kelly Hamman
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - David Goldgar
- Department of Dermatology and Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Nicola Poplawski
- Adult Genetics Unit, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA 5006, Australia; University Department of Paediatrics, University of Adelaide, Adelaide, SA 5005, Australia
| | - Kerry Phillips
- Adult Genetics Unit, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA 5006, Australia; University Department of Paediatrics, University of Adelaide, Adelaide, SA 5005, Australia
| | - Lyn Schofield
- Genetic Services of Western Australia, King Edward Memorial Hospital, Subiaco, WA 6008, Australia
| | - Jacqueline Armstrong
- Adult Genetics Unit, SA Pathology at the Women's and Children's Hospital, North Adelaide, SA 5006, Australia
| | - Cathy Kiraly-Borri
- Genetic Services of Western Australia, King Edward Memorial Hospital, Subiaco, WA 6008, Australia
| | - Graeme K Suthers
- University Department of Paediatrics, University of Adelaide, Adelaide, SA 5005, Australia
| | - David G Huntsman
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada; Department of Pathology and Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC V6Z 2K5, Canada
| | - William D Foulkes
- Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, Montreal, QC H3T 1E2, Canada; Program in Cancer Genetics, Departments of Oncology and Human Genetics, McGill University, Montreal, QC H3A 1B1, Canada
| | - Fatima Carneiro
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP)/Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto 4200-135, Portugal; Medical Faculty of the University of Porto/Centro Hospitalar São João, Porto 4200-319, Portugal
| | - Noralane M Lindor
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Stacey L Edwards
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Juliet D French
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia
| | - Nicola Waddell
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia; Institute for Molecular Bioscience, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Paul S Meltzer
- Genetics Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), NIH, Bethesda, MD 20892, USA
| | - Daniel L Worthley
- School of Medicine, University of Adelaide and Cancer Theme, SAHMRI, Adelaide, SA 5000, Australia
| | - Kasmintan A Schrader
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada; Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC V5Z 1L3, Canada
| | - Georgia Chenevix-Trench
- Department of Genetics and Computational Biology, QIMR Berghofer, Herston, QLD 4029, Australia.
| |
Collapse
|
27
|
Hoyos-Giraldo LS, Escobar-Hoyos LF, Saavedra-Trujillo D, Reyes-Carvajal I, Muñoz A, Londoño-Velasco E, Tello A, Cajas-Salazar N, Ruíz M, Carvajal S, Santella RM. Gene-specific promoter methylation is associated with micronuclei frequency in urothelial cells from individuals exposed to organic solvents and paints. JOURNAL OF EXPOSURE SCIENCE & ENVIRONMENTAL EPIDEMIOLOGY 2016; 26:257-262. [PMID: 25993025 DOI: 10.1038/jes.2015.28] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/25/2015] [Accepted: 02/28/2015] [Indexed: 06/04/2023]
Abstract
Sufficient epidemiologic evidence has established an etiologic link between bladder cancer risk and occupational exposure as a painter to organic solvents. Currently, it remains to be established whether gene-specific promoter methylation contributes to bladder cancer development, including by enhancing chromosome breakage or loss. We investigated the effect of chronic exposure to organic solvents and paints on DNA methylation profiles in the promoter regions of four genes (GSTP1, p16(INK4a), APC and CDH1) and micronucleus (MN) frequency in exfoliated urothelial cells from voided urine from Colombian male non-smoking car painters and age-matched unexposed individuals. The exposed group had a higher percentage of individuals with >2 MNs/2000 cells compared with the unexposed group (P=0.04). Gene-specific analysis showed a significantly higher percentage of individuals with methylated GSTP1, p16(INK4a) and APC in the exposed group. Poisson regression analysis indicated that exposed individuals with methylated GSTP1 and p16(INK4a) promoters were more than twofold more likely to have an increase in MN frequency as compared with the reference. Finally, among exposed individuals with GSTP1 and p16(INK4a) methylated promoters, those with a greater age had a higher RR of increased MN frequency compared with younger exposed individuals with methylated promoters. These results support the conclusion that gene-specific promoter methylation may increase MN frequency in a dependent or independent interaction with occupational exposure to organic solvents.
Collapse
Affiliation(s)
- L S Hoyos-Giraldo
- Department of Biology, Research Group Genetic Toxicology and Cytogenetics, Faculty of Natural Sciences and Education, Universidad del Cauca, Popayán, Cauca, Colombia
| | - L F Escobar-Hoyos
- Department of Biology, Research Group Genetic Toxicology and Cytogenetics, Faculty of Natural Sciences and Education, Universidad del Cauca, Popayán, Cauca, Colombia
- Departments of Pharmacological Sciences and Pathology, Stony Brook University, Stony Brook, New York, USA
| | - D Saavedra-Trujillo
- Department of Biology, Research Group Genetic Toxicology and Cytogenetics, Faculty of Natural Sciences and Education, Universidad del Cauca, Popayán, Cauca, Colombia
| | - I Reyes-Carvajal
- Department of Biology, Research Group Genetic Toxicology and Cytogenetics, Faculty of Natural Sciences and Education, Universidad del Cauca, Popayán, Cauca, Colombia
| | - A Muñoz
- Department of Biology, Research Group Genetic Toxicology and Cytogenetics, Faculty of Natural Sciences and Education, Universidad del Cauca, Popayán, Cauca, Colombia
| | - E Londoño-Velasco
- Department of Biology, Research Group Genetic Toxicology and Cytogenetics, Faculty of Natural Sciences and Education, Universidad del Cauca, Popayán, Cauca, Colombia
| | - A Tello
- Department of Biology, Research Group Genetic Toxicology and Cytogenetics, Faculty of Natural Sciences and Education, Universidad del Cauca, Popayán, Cauca, Colombia
| | - N Cajas-Salazar
- Department of Biology, Research Group Genetic Toxicology and Cytogenetics, Faculty of Natural Sciences and Education, Universidad del Cauca, Popayán, Cauca, Colombia
| | - M Ruíz
- Department of Morphology, Research Group Health and Work, Faculty of Health Sciences, Universidad del Cauca, Popayán, Cauca, Colombia
| | - S Carvajal
- Department of Biology, Research Group Genetic Toxicology and Cytogenetics, Faculty of Natural Sciences and Education, Universidad del Cauca, Popayán, Cauca, Colombia
| | - R M Santella
- Department of Environmental Health Sciences, Mailman School of Public Health of Columbia University, New York, New York, USA
| |
Collapse
|
28
|
Vidal AF, Cruz AMP, Magalhães L, Pereira AL, Anaissi AKM, Alves NCF, Albuquerque PJBS, Burbano RMR, Demachki S, Ribeiro-dos-Santos Â. hsa-miR-29c and hsa-miR-135b differential expression as potential biomarker of gastric carcinogenesis. World J Gastroenterol 2016; 22:2060-70. [PMID: 26877610 PMCID: PMC4726678 DOI: 10.3748/wjg.v22.i6.2060] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/10/2015] [Accepted: 11/13/2015] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the expression profiles of hsa-miR-29c and hsa-miR-135b in gastric mucosal samples and their values as gastric carcinogenesis biomarkers. METHODS The expression levels of hsa-miR-29c and hsa-miR-135b in normal gastric mucosa, non-atrophic chronic gastritis, intestinal metaplasia and intestinal-type gastric adenocarcinoma were analysed using quantitative real-time PCR. The difference between hsa-miR-29c and hsa-miR-135b expression profiles in the grouped samples was evaluated by ANOVA and Student's t-test tests. The results were adjusted for multiple testing by using Bonferroni's correction. P values ≤ 0.05 were considered statistically significant. To evaluate hsa-miR-29c and hsa-miR-135b expressions as potential biomarkers of gastric carcinogenesis, we performed a receiver operating characteristic curve analysis and the derived area under the curve, and a Categorical Principal Components Analysis. In silico identification of the genetic targets of hsa-miR-29c and hsa-miR-135b was performed using different prediction tools, in order to identify possible genes involved in gastric carcinogenesis. RESULTS The expression levels of hsa-miR-29c were higher in normal gastric mucosal samples, and decreased progressively in non-atrophic chronic gastritis samples, intestinal metaplasia samples and intestinal-type gastric adenocarcinoma samples. The expression of hsa-miR-29c in the gastric lesions showed that non-atrophic gastritis have an intermediate profile to gastric normal mucosa and intestinal-type gastric adenocarcinoma, and that intestinal metaplasia samples presented an expression pattern similar to that in intestinal-type gastric adenocarcinoma. This microRNA (miRNA) has a good discriminatory accuracy between normal gastric samples and (1) intestinal-type gastric adenocarcinoma; and (2) intestinal metaplasia, and regulates the DMNT3A oncogene. hsa-miR-135b is up-regulated in non-atrophic chronic gastritis and intestinal metaplasia samples and down-regulated in normal gastric mucosa and intestinal-type gastric adenocarcinoma samples. Non-atrophic chronic gastritis and intestinal metaplasia are significantly different from normal gastric mucosa samples. hsa-miR-135b expression presented a greater discriminatory accuracy between normal samples and gastric lesions. This miRNA was associated with Helicobacter pylori presence in non-atrophic chronic gastritis samples and regulates the APC and KLF4 tumour suppressor genes. CONCLUSION Our results provide evidence of epigenetic alterations in non-atrophic chronic gastritis and intestinal metaplasia and suggest that hsa-miR-29c and hsa-miR-135b are promising biomarkers of gastric carcinogenesis.
Collapse
|
29
|
Copy number variants associated with 18p11.32, DCC and the promoter 1B region of APC in colorectal polyposis patients. Meta Gene 2015; 7:95-104. [PMID: 26909336 PMCID: PMC4733217 DOI: 10.1016/j.mgene.2015.12.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 12/16/2015] [Accepted: 12/21/2015] [Indexed: 01/05/2023] Open
Abstract
Familial Adenomatous Polyposis (FAP) is the second most common inherited predisposition to colorectal cancer (CRC) associated with the development of hundreds to thousands of adenomas in the colon and rectum. Mutations in APC are found in ~ 80% polyposis patients with FAP. In the remaining 20% no genetic diagnosis can be provided suggesting other genes or mechanisms that render APC inactive may be responsible. Copy number variants (CNVs) remain to be investigated in FAP and may account for disease in a proportion of polyposis patients. A cohort of 56 polyposis patients and 40 controls were screened for CNVs using the 2.7M microarray (Affymetrix) with data analysed using ChAS (Affymetrix). A total of 142 CNVs were identified unique to the polyposis cohort suggesting their involvement in CRC risk. We specifically identified CNVs in four unrelated polyposis patients among CRC susceptibility genes APC, DCC, MLH1 and CTNNB1 which are likely to have contributed to disease development in these patients. A recurrent deletion was observed at position 18p11.32 in 9% of the patients screened that was of particular interest. Further investigation is necessary to fully understand the role of these variants in CRC risk given the high prevalence among the patients screened.
Collapse
Key Words
- ALL, acute lymphoblastic leukaemia
- BH, Bengamini and Hochberg
- CHAS, Chromosome Analysis Suite
- CN, copy number
- CNV
- CNV, copy number variation
- COSMIC, Catalogue of Somatic Mutations in Cancer
- CRC, colorectal cancer
- Cancer
- DGV, Database of genomic variants
- DNA, deoxyribose nucleic acid
- FAP, familial adenomatous polyposis
- HMDD, human microRNA disease database
- KEGG, Kyoto Encyclopaedia of Genes and Genomes
- Kb, kilobase
- LOH, loss of heterozygosity
- MLPA, multiplex ligation-dependant probe amplification
- MMR, mismatch repair
- NTC, no template control
- QC, quality control
- RNA, ribose nucleic acid
- SNP, single nucleotide polymorphism
- TAM, Tool for the annotation of microRNAs
- TCGA, The Cancer Genome Atlas
- UCSC, University of California, Santa Cruz
- diagnostic testing
- lncRNA, link RNA
- long non-coding RNAs
- mapd, median absolute pairwise difference
- miR, microRNA
- ng, nanogram
- polyposis
Collapse
|
30
|
Characteristics of DNA methylation changes induced by traffic-related air pollution. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2015; 796:46-53. [PMID: 26778509 DOI: 10.1016/j.mrgentox.2015.12.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/04/2015] [Accepted: 12/14/2015] [Indexed: 12/20/2022]
Abstract
Traffic-related air pollution (TRAP) is a potential risk factor for numerous respiratory disorders, including lung cancer, while alteration of DNA methylation may be one of the underlying mechanisms. However, the effects of TRAP mixtures on DNA methylation have not been investigated. We have studied the effects of brief or prolonged TRAP exposures on DNA methylation in the rat. The exposures were performed in spring and autumn, with identical study procedures. In each season, healthy Wistar rats were exposed to TRAP at for 4 h, 7 d, 14 d, or 28 d. Global DNA methylation (LINE-1 and Alu) and specific gene methylation (p16(CDKN2A), APC, and iNOS) in the DNA from blood and lung tissues were quantified by pyrosequencing. Multiple linear regression was applied to assess the influence of air pollutants on DNA methylation levels. The levels of PM2.5, PM10, and NO2 in the high and moderate groups were significantly higher than in the control group. The DNA methylation levels were not significantly different between spring and autumn. When spring and autumn data were analyzed together, PM2.5, PM10, and NO2 exposures were associated with changes in%5mC (95% CI) in LINE-1, iNOS, p16(CDKN2A), and APC ranging from -0.088 (-0.150, -0.026) to 0.102 (0.049, 0.154) per 1 μg/m(3) increase in the pollutant concentration. Prolonged exposure to a high level of TRAP was negatively associated with LINE-1 and iNOS methylation, and positively associated with APC methylations in the DNA from lung tissues but not blood. These findings show that TRAP exposure is associated with decreased methylation of LINE-1 and iNOS, and increased methylation of p16(CDKN2A) and APC.
Collapse
|
31
|
MALDI-TOF Mass Array Analysis of Nell-1 Promoter Methylation Patterns in Human Gastric Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:136941. [PMID: 26090379 PMCID: PMC4452250 DOI: 10.1155/2015/136941] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 10/10/2014] [Accepted: 11/06/2014] [Indexed: 12/19/2022]
Abstract
Mass spectrometry (MS) enables rapid and sensitive qualitative and quantitative analyses of biomolecules (proteins, peptides, oligosaccharides, lipids, DNA, and RNA), drugs, and metabolites. MS has become an essential tool in modern biomedical research, including the analysis of DNA methylation. DNA methylation has been reported in many cancers, suggesting that it can be utilized as an early biomarker to improve the early diagnosis rate. Using matrix-assisted laser desorption/ionization time-of-flight MS and MassCLEAVE reagent, we compared Nell-1 hypermethylation levels among tumor tissues, paracarcinoma tissues, and normal tissues from gastric cancer patients. Almost 80% of the CpG sites in the amplicons produced were covered by the analysis. Our results indicate a significant difference in methylation status between gastric cancer tissue (a higher level) and normal tissue. The same trend was identified in gastric cancer tissue versus paracarcinoma tissue. We also detected lower relative expression of Nell-1 by real-time PCR. Furthermore, immunohistochemical analyses revealed that Nell-1 staining was less intense in cancer tissue relative to normal tissue and that the tumor cells had spread to the muscle layer. These findings may serve as a guide for the early diagnosis of gastric cancer.
Collapse
|
32
|
Kamat N, Khidhir MA, Hussain S, Alashari MM, Rannug U. Chemotherapy induced microsatellite instability and loss of heterozygosity in chromosomes 2, 5, 10, and 17 in solid tumor patients. Cancer Cell Int 2014; 14:118. [PMID: 25493073 PMCID: PMC4260186 DOI: 10.1186/s12935-014-0118-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Accepted: 10/27/2014] [Indexed: 01/05/2023] Open
Abstract
Background The inevitable side effects of the currently used chemotherapy are associated with serious syndromes. Genotoxic effects and consequent genetic instability may play an important role in these syndromes. The aim of the study was to evaluate chemotherapy-related microsatellite instability (MSI), loss of heterozygosity (LOH), and loss of mismatch repair (MMR) expression in solid tumor patients. Methods Samples were collected from 117 de novo patients with solid tumors of different origins. Specimens, taken pre- and post-treatment, were screened for MSI and LOH in 10 microsatellite sequences in blood, and expression of five MMR proteins were analyzed in cancer tissues using immunohistochemistry. Statistical analysis included the use of; Fisher’s exact test, Chi Square, and an inter-rater reliability test using Cohen’s kappa coefficient. Results Microsatellite analysis showed that 66.7% of the patients had MSI, including 23.1% high-positive MSI and 43.6% low-positive MSI. A large portion (41%) of the patients exhibited LOH in addition to MSI. MSI and LOH were detected in seven loci in which incidence rates ranged from 3.8% positive for Bat-26 to 34.6% positive for Tp53-Alu. Immunohistochemistry revealed that human mutL homolog 1 (hMLH1) expression was deficient in 29.1% of the patients, whereas 18.8%, 23.9%, 13.4%, and 9.7% were deficient for human mutS homolog 2 (hMSH2), P53, human mutS homolog 6 (hMSH6) and human post-meiotic segregation increased 2 (hPMS2), respectively. There was a significant correlation between MSI and LOH incidence in Tp53-Alu, Mfd41, and APC with low or deficient expression of hMLH1, hMSH2, and P53. A significant association between MSI and LOH, and incidence of secondary tumors was also evident. Conclusions The negative correlation between MMR expression, MSI, and LOH and increased resistance to anti-cancer drugs and development of secondary cancers demonstrates a useful aid in early detection of potential chemotherapy-related side-effects. The diagnostic value demonstrated in our earlier study on breast cancer patients was confirmed for other solid tumors. Electronic supplementary material The online version of this article (doi:10.1186/s12935-014-0118-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nasir Kamat
- Department of Molecular Biosciences, the Wenner-Gren Institute (MBW), Stockholm University, SE-106 91 Stockholm, Sweden
| | - Mohammed A Khidhir
- Department of Genetics Research, Management of Natural Conservations, AlAin City, UAE
| | - Sabir Hussain
- Department of Oncology and Hematology, Tawam Hospital, AlAin City, UAE
| | - Mouied M Alashari
- Department of Pathology, University of Utah, Salt Lake City, Utah 84112 USA
| | - Ulf Rannug
- Department of Molecular Biosciences, the Wenner-Gren Institute (MBW), Stockholm University, SE-106 91 Stockholm, Sweden
| |
Collapse
|
33
|
Li Z, Chang X, Dai D, Deng P, Sun Q. RASSF10 is an epigenetically silenced tumor suppressor in gastric cancer. Oncol Rep 2014; 31:1661-8. [PMID: 24573726 DOI: 10.3892/or.2014.3039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 01/14/2014] [Indexed: 11/06/2022] Open
Abstract
To better understand the role of the N-Terminal Ras association domain family (RASSF) genes in the development of gastric cancer, we examined the expression of RASSF7 and RASSF10 and RASSF10 methylation in gastric cancer. We found that RASSF10 expression was lost in six gastric cancer cell lines, and was rescued by a DNA demethylating agent and a histone deacetylase inhibitor. However, RASSF7 expression was strong in four cancer cell lines as well as in 87% of primary gastric cancer tissues. In contrast, RASSF7 expression was moderate in the GES-1 cell line and negative in 33.3% of the corresponding non-cancerous tissues. Analysis of RASSF10 methylation by methylation-specific PCR (MSP) and sequencing revealed that the methylation frequency in primary gastric carcinoma tissues was significantly higher compared to that in adjacent non-carcinoma tissues (61.6 vs. 38.4%; p<0.01). The methylation frequency in the tumor with invasion depth at T3 and T4 was significantly higher compared to that with invasion depth at T1 and T2 (67.1 vs. 37.5%; p<0.05). Hypermethylation of RASSF10 was found in the patients with lymph node metastasis, compared to those with unaffected lymph nodes (68.8 vs. 40.9%; p<0.05). Among the 4 gross types of the Borrmann classification, i.e. EGC, Borrmann Ⅰ, Borrmann Ⅱ, Borrmann Ⅲ and Borrmann Ⅳ, the last one was more frequently methylated (85.7 vs. 56.9%; p<0.05). The present study revealed that RASSF10 is an epigenetically silenced gene involved in tumor invasion and metastasis in gastric cancer, suggesting that the methylation status of RASSF10 may be a useful indicator to predict the malignant degree of gastric cancer.
Collapse
Affiliation(s)
- Zhenhua Li
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Xiaojing Chang
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Dongqiu Dai
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Peng Deng
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, P.R. China
| | - Qiang Sun
- Department of Gastrointestinal Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110032, P.R. China
| |
Collapse
|
34
|
Otani K, Li X, Arakawa T, Chan FKL, Yu J. Epigenetic-mediated tumor suppressor genes as diagnostic or prognostic biomarkers in gastric cancer. Expert Rev Mol Diagn 2013; 13:445-55. [PMID: 23782252 DOI: 10.1586/erm.13.32] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Gastric cancer is believed to result in part from the accumulation of multiple genetic and epigenetic alterations leading to oncogene overexpression and tumor suppressor loss. Tumor suppressor genes are inactivated more frequently by promoter methylation than by mutation in gastric cancer. Identification of genes inactivated by promoter methylation is a powerful approach to discover novel tumor suppressor genes. We have previously identified tumor suppressor genes in gastric cancer by genome-wide methylation screening. The biological functions of these genes are related to cell adhesion, ubiquitination, transcription, p53 regulation and diverse signaling pathways. Some of the tumor suppressor genes are of particular clinical importance as they can be used as predictive biomarkers for early diagnosis or ongoing prognosis of gastric cancer.
Collapse
Affiliation(s)
- Koji Otani
- Department of Medicine and Therapeutics, Institute of Digestive Disease, Li KaShing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | | | | | | | | |
Collapse
|
35
|
Ayed-Guerfali DB, Hassairi B, Khabir A, Sellami-Boudawara T, Gargouri A, Mokdad-Gargouri R. Expression of APC, β-catenin and E-cadherin in Tunisian patients with gastric adenocarcinoma: clinical significance. Tumour Biol 2013; 35:1775-83. [PMID: 24197976 DOI: 10.1007/s13277-013-1236-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 09/18/2013] [Indexed: 12/13/2022] Open
Abstract
Aberrant activation of the Wnt signalling pathway is a key feature of many cancers. β-Catenin, adenomatous polyposis coli (APC) and E-cadherin are major players in this pathway. The aim of this study is to examine the expression of β-catenin, APC and E-cadherin in tumour tissues of 80 Tunisian patients with gastric carcinoma and to determine the methylation status of the APC promoter in tumour tissues. Associations between protein expression and clinico-pathological parameters, including prognosis, were performed. Positive expression of β-catenin, APC and E-cadherin was observed in 77.5, 68.7 and 60% of cases, respectively. Tumours lacking membranous expression of β-catenin had greater extent of lymph node metastasis, poor differentiation and advanced T-stage. The expression of E-cadherin correlated with poor differentiation (P = 0.05) and β-catenin expression (P = 0.004). With regards to prognosis, the overall survival time was significantly prolonged for patients showing normal β-catenin expression (exclusively or predominantly membranous staining) alone or combined with positive APC expression (P log rank = 0.008 and 0.003, respectively). The methylated pattern of APC promoter 1A was detected in 43.8% of cases and correlated with T-stage (P = 0.046) and distant metastasis (P = 0.037). No correlation was found between the methylated profile of APC promoter 1A and the expression of APC protein in tumour tissues. Our findings suggest that deregulation of the Wnt pathway via abnormal expression of β-catenin and E-cadherin occurred frequently in gastric carcinoma and correlated with worse clinical behaviour.
Collapse
Affiliation(s)
- Dorra Ben Ayed-Guerfali
- Center of Biotechnology of Sfax, University of Sfax, Sidi Mansour Street Km 6, BP 1177, 3038, Sfax, Tunisia
| | | | | | | | | | | |
Collapse
|
36
|
Sameer AS. Colorectal cancer: a researcher’s perspective of the molecular angel’s gone eccentric in the Vale of Kashmir. Tumour Biol 2013; 34:1301-1315. [PMID: 23417859 DOI: 10.1007/s13277-013-0692-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 02/03/2013] [Indexed: 02/06/2023] Open
|
37
|
Sameer AS. Colorectal cancer: molecular mutations and polymorphisms. Front Oncol 2013; 3:114. [PMID: 23717813 PMCID: PMC3651991 DOI: 10.3389/fonc.2013.00114] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 04/25/2013] [Indexed: 01/10/2023] Open
Abstract
Colorectal cancer (CRC) is one of the major causes of mortality and morbidity, and is the third most common cancer in men and the second most common cancer in women worldwide. The incidence of CRC shows considerable variation among racially or ethnically defined populations in multiracial/ethnic countries. The tumorigenesis of CRC is either because of the chromosomal instability (CIN) or microsatellite instability (MIN) or involving various proto-oncogenes, tumor-suppressor genes, and also epigenetic changes in the DNA. In this review I have focused on the mutations and polymorphisms of various important genes of the CIN and MIN pathways which have been implicated in the development of CRC.
Collapse
Affiliation(s)
- Aga Syed Sameer
- Department of Biochemistry, Sher-I-Kashmir Institute of Medical Sciences Associated Medical College, Bemina, SrinagarKashmir, India
| |
Collapse
|
38
|
Yamagishi M, Watanabe T. Molecular hallmarks of adult T cell leukemia. Front Microbiol 2012; 3:334. [PMID: 23060864 PMCID: PMC3444139 DOI: 10.3389/fmicb.2012.00334] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 08/29/2012] [Indexed: 12/20/2022] Open
Abstract
The molecular hallmarks of adult T cell leukemia (ATL) comprise outstanding deregulations of signaling pathways that control the cell cycle, resistance to apoptosis, and proliferation of leukemic cells, all of which have been identified by early excellent studies. Nevertheless, we are now confronted the therapeutic difficulties of ATL that is a most aggressive T cell leukemia/lymphoma. Using next-generation strategies, emerging molecular characteristics such as specific surface markers and an additional catalog of signals affecting the fate of leukemic cells have been added to the molecular hallmarks that constitute an organizing principle for rationalizing the complexities of ATL. Although human T cell leukemia virus type 1 is undoubtedly involved in ATL leukemogenesis, most leukemic cells do not express the viral protein Tax. Instead, cellular gene expression changes dominate homeostasis disorders of infected cells and characteristics of ATL. In this review, we summarize the state of the art of ATL molecular pathology, which supports the biological properties of leukemic cells. In addition, we discuss the recent discovery of two molecular hallmarks of potential generality; an abnormal microRNA pattern and epigenetic reprogramming, which strongly involve the imbalance of the molecular network of lymphocytes. Global analyses of ATL have revealed the functional impact of crosstalk between multifunctional pathways. Clinical and biological studies on signaling inhibitory agents have also revealed novel oncogenic drivers that can be targeted in future. ATL cells, by deregulation of such pathways and their interconnections, may become masters of their own destinies. Recognizing and understanding of the widespread molecular applicability of these concepts will increasingly affect the development of novel strategies for treating ATL.
Collapse
Affiliation(s)
- Makoto Yamagishi
- Laboratory of Tumor Cell Biology, Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo Minato-ku, Tokyo, Japan
| | | |
Collapse
|
39
|
Bruno A, Dovizio M, Tacconelli S, Patrignani P. Mechanisms of the antitumoural effects of aspirin in the gastrointestinal tract. Best Pract Res Clin Gastroenterol 2012. [PMID: 23199511 DOI: 10.1016/j.bpg.2012.10.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
A recent clinical study showed that after five years of taking aspirin, at doses of at least 75 mg once daily, death rates were 54% less for gastrointestinal (GI) cancers. The finding of aspirin benefit at low-doses used for cardioprevention, locates the antiplatelet effect of aspirin at the centre of its antitumour efficacy. At low-doses, aspirin acts mainly by an irreversible inactivation of platelet cyclooxygenase (COX)-1 activity. We propose that platelet activation is involved in the early stages of colorectal carcinogenesis in man through the induction of a COX-2-mediated paracrine signalling between stromal cells and epithelial cells within adenomas. In this scenario, aspirin causes a chemopreventive effect by countering platelet activation which seems to play a role in early event in GI tumourigenesis.
Collapse
Affiliation(s)
- Annalisa Bruno
- Department of Medicine and Aging, G. d'Annunzio University, School of Medicine, Italy
| | | | | | | |
Collapse
|
40
|
Yamanaka S, Olaru AV, An F, Luvsanjav D, Jin Z, Agarwal R, Tomuleasa C, Popescu I, Alexandrescu S, Dima S, Chivu M, Montgomery EA, Torbenson M, Meltzer SJ, Selaru FM. MicroRNA-21 inhibits Serpini1, a gene with novel tumour suppressive effects in gastric cancer. Dig Liver Dis 2012; 44:589-96. [PMID: 22464652 PMCID: PMC3360813 DOI: 10.1016/j.dld.2012.02.016] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Revised: 02/16/2012] [Accepted: 02/24/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND A thorough understanding of gastric cancer at the molecular level is urgently needed. One prominent oncogenic microRNA, miR-21, was previously reported to be upregulated in gastric cancer. METHODS We performed an unbiased search for downstream messenger RNA targets of miR-21, based on miR-21 dysregulation, by using human tissue specimens and the MKN28 human gastric carcinoma cell line. Molecular techniques include microRNA microarrays, cDNA microarrays, qRT-PCR for miR and mRNA expression, transfection of MKN28 with miR-21 inhibitor or Serpini1 followed by Western blotting, cell cycle analysis by flow cytometry and luciferase reporter assay. RESULTS This search identified Serpini1 as a putative miR-21 target. Luciferase assays demonstrated direct interaction between miR-21 and Serpini1 3'UTR. miR-21 and Serpini1 expression levels were inversely correlated in a subgroup of gastric cancers, suggesting a regulatory mechanism that included both of these molecules. Furthermore, Serpini1 induced growth retardation of MKN28 and induced vigorous G1/S arrest suggesting its potential tumour-suppressive function in the stomach. CONCLUSION Taken together, these data suggest that in a subgroup of gastric cancers, miR-21 is upregulated, inducing downregulation of Serpini1, which in turn releases the G1-S transition checkpoint, with the end result being increased tumour growth.
Collapse
Affiliation(s)
- Sumitaka Yamanaka
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexandru V. Olaru
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Fangmei An
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Delgermaa Luvsanjav
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zhe Jin
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rachana Agarwal
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ciprian Tomuleasa
- Department of Cancer Immunology, Prof. Dr. Ion Chiricuta Comprehensive Cancer Center and Iuliu Hatieganu University of Medicine and Pharmacy, Romania
| | - Irinel Popescu
- Clinic of General Surgery and Liver Transplantation "Dan Setlacec", Fundeni Clinical Institute of Digestive Diseases and Liver Transplantation, Bucharest, Romania
| | - Sorin Alexandrescu
- Clinic of General Surgery and Liver Transplantation "Dan Setlacec", Fundeni Clinical Institute of Digestive Diseases and Liver Transplantation, Bucharest, Romania
| | - Simona Dima
- Clinic of General Surgery and Liver Transplantation "Dan Setlacec", Fundeni Clinical Institute of Digestive Diseases and Liver Transplantation, Bucharest, Romania
| | - Mihaela Chivu
- Clinic of General Surgery and Liver Transplantation "Dan Setlacec", Fundeni Clinical Institute of Digestive Diseases and Liver Transplantation, Bucharest, Romania
| | | | - Michael Torbenson
- Department of Pathology, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Stephen J. Meltzer
- Department of Pathology, Johns Hopkins University Hospital, Baltimore, MD, USA
| | - Florin M. Selaru
- Division of Gastroenterology and Hepatology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
41
|
Yue MX, Fu XW, Zhou GB, Hou YP, DU M, Wang L, Zhu SE. Abnormal DNA methylation in oocytes could be associated with a decrease in reproductive potential in old mice. J Assist Reprod Genet 2012; 29:643-50. [PMID: 22618193 DOI: 10.1007/s10815-012-9780-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Accepted: 04/24/2012] [Indexed: 12/28/2022] Open
Abstract
PURPOSE This study was designed to evaluate DNA methylation and the expression of DNA methyltransferases (Dnmt1, Dnmt3a, Dnmt3b and Dnmt3L) in metaphaseII (MII) oocytes and the DNA methylation of pre-implantation embryos during mouse aging to address whether such aging-related changes are associated with decreased reproductive potential in aged mice. METHODS Oocytes (MII) from 6 to 8 weeks old female mice are referred to as the 'young group'; oocytes from the same group that were maintained until 35-40 weeks old are referred to as the 'old group.' The oocytes were fertilized both in vitro and in vivo to obtain embryos. The DNA methylation levels in the oocytes (MII) and pre-implantation embryos were assessed using fluorescence staining. The expression levels of the Dnmt genes in the oocytes (MII) were assessed using Western blotting. RESULTS The DNA methylation levels in the oocytes and pre-implantation embryos (in vivo and in vitro) decreased significantly during the aging of the mice. The expression levels of all of the examined Dnmt proteins in the old group were lower than young group. Both the cleavage and blastocyst rate were significantly lower in the oocytes of the older mice (69.9 % vs. 80.9 %, P < 0.05; 33.9 % vs. 56.4 %, P < 0.05). The pregnancy rate of the old mice was lower than that of the young mice (46.7 % vs. 100 %, P < 0.05). The stillbirth and fetal malformation rate was significantly higher in the old group than in the young group (17.2 % vs. 2.9 %, P < 0.05). CONCLUSIONS The decreased expression of Dnmt1, Dnmt3a, Dnmt3b and Dnmt3L in oocytes (MII) and the change of genome-wide DNA methylation in oocytes and pre-implantation embryos due to aging may be related to lower reproductive potential in old female mice.
Collapse
Affiliation(s)
- Ming-xing Yue
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
42
|
Jain S, Chang TT, Hamilton JP, Lin SY, Lin YJ, Evans AA, Selaru FM, Lin PW, Chen SH, Block TM, Hu CT, Song W, Meltzer SJ, Su YH. Methylation of the CpG sites only on the sense strand of the APC gene is specific for hepatocellular carcinoma. PLoS One 2011; 6:e26799. [PMID: 22073196 PMCID: PMC3206845 DOI: 10.1371/journal.pone.0026799] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Accepted: 10/03/2011] [Indexed: 12/12/2022] Open
Abstract
Hypermethylation of the promoter of the tumor suppressor gene, adenomatous polyposis coli (APC), occurs in various malignancies, including hepatocellular carcinoma (HCC). However, reports on the specificity of the methylation of the APC gene for HCC have varied. To gain insight into how these variations occur, bisulfite PCR sequencing was performed to analyze the methylation status of both sense and antisense strands of the APC gene in samples of HCC tissue, matched adjacent non-HCC liver tissue, hepatitis, cirrhosis, and normal liver tissues. DNA derived from fetal liver and 12 nonhepatic normal tissue was also examined. These experiments revealed liver-specific, antisense strand-biased CpG methylation of the APC gene and suggested that, although methylation of the antisense strand of the APC gene exists in normal liver and other non-HCC disease liver tissue, methylation of the sense strand of the APC gene occurs predominantly in HCC. To determine the effect of the DNA strand on the specificity of the methylated APC gene as a biomarker for HCC detection, quantitative methylation-specific PCR assays for sense and antisense strand DNA were developed and performed on DNA isolated from HCC (n = 58), matched adjacent non-HCC (n = 58), cirrhosis (n = 41), and hepatitis (n = 39). Receiver operating characteristic curves were constructed. With the cutoff value set at the limit of detection, the specificity of sense and antisense strand methylation was 84% and 43%, respectively, and sensitivity was 67.2% and 72.4%, respectively. This result demonstrated that the identity of the methylated DNA strand impacted the specificity of APC for HCC detection. Interestingly, methylation of the sense strand of APC occurred in 40% of HCCs from patients with serum AFP levels less than 20 ng/mL, suggesting a potential role for APC as a biomarker to complement AFP in HCC screening.
Collapse
Affiliation(s)
- Surbhi Jain
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Ting-Tsung Chang
- Department of Medicine, Infectious Diseases and Signaling Research Center, College of Medicine, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - James P. Hamilton
- Division of Gastroenterology and Hepatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Selena Y. Lin
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Yih-Jyh Lin
- Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan, Republic of China
| | - Alison A. Evans
- School of Public Health, Drexel University, Philadelphia, Pennsylvania, United States of America
| | - Florin M. Selaru
- Division of Gastroenterology and Hepatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Pin- Wen Lin
- Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan, Republic of China
| | - Shun-Hua Chen
- Department of Microbiology, Medical College, National Cheng Kung University, Tainan, Taiwan, Republic of China
| | - Timothy M. Block
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Chi-Tan Hu
- Department of General Medicine, Buddhist Tzu Chi General Hospital, Hualien, Taiwan, Republic of China
| | - Wei Song
- JBS Science Inc., Philadelphia, Pennsylvania, United States of America
| | - Stephen J. Meltzer
- Division of Gastroenterology and Hepatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, United States of America
| | - Ying-Hsiu Su
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
43
|
Jennes I, Zuntini M, Mees K, Palagani A, Pedrini E, De Cock G, Fransen E, Vanden Berghe W, Sangiorgi L, Wuyts W. Identification and functional characterization of the human EXT1 promoter region. Gene 2011; 492:148-59. [PMID: 22037484 DOI: 10.1016/j.gene.2011.10.034] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2011] [Revised: 10/02/2011] [Accepted: 10/11/2011] [Indexed: 01/02/2023]
Abstract
BACKGROUND Mutations in Exostosin-1 (EXT1) or Exostosin-2 (EXT2) cause the autosomal dominant disorder multiple osteochondromas (MO). This disease is mainly characterized by the appearance of multiple cartilage-capped protuberances arising from children's metaphyses and is known to display clinical inter- and intrafamilial variations. EXT1 and EXT2 are both tumor suppressor genes encoding proteins that function as glycosyltransferases, catalyzing the biosynthesis of heparan sulfate. At present, however, very little is known about the regulation of these genes. Two of the most intriguing questions concerning the pathogenesis of MO are how disruption of a ubiquitously expressed gene causes this cartilage-specific disease and how the clinical intrafamilial variation can be explained. Since mutations in the EXT1 gene are responsible for ~65% of the MO families with known causal mutation, our aim was to isolate and characterize the EXT1 promoter region to elucidate the transcriptional regulation of this tumor suppressor gene. METHODS In the present study, luciferase reporter gene assays were used to experimentally confirm the in silico predicted EXT1 core promoter region. Subsequently, we evaluated the effect of single nucleotide polymorphisms (SNP's) on EXT1 promoter activity and transcription factor binding using luciferase assays, electrophoretic mobility shift assays (EMSA), and enzyme-linked immunosorbent assays (ELISA). Finally, a genotype-phenotype study was performed with the aim to identify one or more genetic modifiers influencing the clinical expression of MO. RESULTS Transient transfection of HEK293 cells with a series of luciferase reporter constructs mapped the EXT1 core promoter at approximately -917 bp upstream of the EXT1 start codon, within a 123 bp region. This region is conserved in mammals and located within a CpG-island containing a CAAT- and a GT-box. A polymorphic G/C-SNP at -1158 bp (rs34016643) was demonstrated to be located in a USF1 transcription factor binding site, which is lost with the presence of the C-allele resulting in a ~56% increase in EXT1 promoter activity. A genotype-phenotype study was suggestive for association of the C-allele with shorter stature, but also with a smaller number of osteochondromas. CONCLUSIONS We provide for the first time insight into the molecular regulation of EXT1. Although a larger patient population will be necessary for statistical significance, our data suggest the polymorphism rs34016643, in close proximity of the EXT1 promoter, to be a potential regulatory SNP, which could be a primary modifier that might explain part of the clinical variation observed in MO patients.
Collapse
Affiliation(s)
- Ivy Jennes
- Department of Medical Genetics, University of Antwerp, and Antwerp University Hospital, Edegem, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Hou L, Zhang X, Tarantini L, Nordio F, Bonzini M, Angelici L, Marinelli B, Rizzo G, Cantone L, Apostoli P, Bertazzi PA, Baccarelli A. Ambient PM exposure and DNA methylation in tumor suppressor genes: a cross-sectional study. Part Fibre Toxicol 2011; 8:25. [PMID: 21878113 PMCID: PMC3180673 DOI: 10.1186/1743-8977-8-25] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Accepted: 08/30/2011] [Indexed: 12/20/2022] Open
Abstract
Exposure to ambient air particles matter (PM) has been associated with increased risk of lung cancer. Aberrant tumor suppressor gene promoter methylation has emerged as a promising biomarker for cancers, including lung cancer. Whether exposure to PM is associated with peripheral blood leukocyte (PBL) DNA methylation in tumor suppressor genes has not been evaluated. In 63 male healthy steel workers with well-characterized exposure to metal-rich particles nearby Brescia, Italy, we evaluated whether exposure to PM and metal components was associated with PBL DNA methylation in 4 tumor suppressor genes (i.e., APC, p16, p53 and RASSF1A). Blood samples were obtained on the 1st (baseline) and 4th day (post-exposure) of the same work week and DNA methylation was measured using pyrosequencing. A linear mixed model was used to examine the correlations of the exposure with promoter methylation levels. Mean promoter DNA methylation levels of APC or p16 were significantly higher in post-exposure samples compared to that in baseline samples (p-values = 0.005 for APC, and p-value = 0.006 for p16). By contrast, the mean levels of p53 or RASSF1A promoter methylation was decreased in post-exposure samples compared to that in baseline samples (p-value = 0.015 for p53; and p-value < 0.001 for RASSF1A). In post-exposure samples, APC methylation was positively associated with PM10 (β = 0.27, 95% CI: 0.13-0.40), and PM1 (β = 0.23, 95% CI: 0.09-0.38). In summary, ambient PM exposure was associated with PBL DNA methylation levels of tumor suppressor genes of APC, p16, p53 and RASSF1A, suggesting that such methylation alterations may reflect processes related to PM-induced lung carcinogenesis.
Collapse
Affiliation(s)
- Lifang Hou
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, 680 N, Lakeshore Drive, Chicago, 60611, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Sameer AS, Shah ZA, Abdullah S, Chowdri NA, Siddiqi MA. Analysis of molecular aberrations of Wnt pathway gladiators in colorectal cancer in the Kashmiri population. Hum Genomics 2011; 5:441-452. [PMID: 21807601 PMCID: PMC3525962 DOI: 10.1186/1479-7364-5-5-441] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 03/17/2011] [Indexed: 12/31/2022] Open
Abstract
The development and progression of colorectal cancer (CRC) is a multi-step process, and the Wnt pathways with its two molecular gladiators adenomatous polyposis coli (APC) and β-catenin plays an important role in transforming a normal tissue into a malignant one. In this study, we aimed to investigate the role of aberrations in the APC and β-catenin genes in the pathogenesis of CRC in the Kashmir valley, and to correlate it with various clinicopathological variables. We examined the paired tumour and normal-tissue specimens of 86 CRC patients for the occurrence of aberrations in the mutation cluster region (MCR) of the APC gene and exon 3 of the β-catenin gene by polymerase chain reaction-single-strand conformation polymorphism (PCR-SSCP) and/or PCR-direct sequencing. Analysis of promoter hypermethylation of the APC gene was also carried out using methylation-specific PCR (MS-PCR). The overall mutation rate of the MCR of the APC gene among 86 CRC cases was 12.8 per cent (11 of 86). Promoter hypermethylation of APC was observed in 54.65 per cent (47 of 86) of cases. Furthermore, we found a significant association between tumour location, tumour grade and node status and the methylation status of the APC gene (p ≤ 0.05). Although the number of mutations in the APC and β-catenin genes in our CRC cases was very low, the study confirms the role of epigenetic gene silencing of the pivotal molecular gladiator, APC, of the Wnt pathway in the development of CRC in the Kashmiri population.
Collapse
Affiliation(s)
- A Syed Sameer
- Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of
Medical Sciences, Soura, Srinagar, Kashmir, 190011, India
- Department of Clinical Biochemistry, Sher-I-Kashmir Institute of Medical Sciences,
Soura, Srinagar, Kashmir, 190011, India
| | - Zaffar A Shah
- Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of
Medical Sciences, Soura, Srinagar, Kashmir, 190011, India
| | - Safiya Abdullah
- Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of
Medical Sciences, Soura, Srinagar, Kashmir, 190011, India
| | - Nissar A Chowdri
- Department of General Surgery, Sher-I-Kashmir Institute of Medical Sciences,
Soura, Srinagar, Kashmir, 190011, India
| | - Mushtaq A Siddiqi
- Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of
Medical Sciences, Soura, Srinagar, Kashmir, 190011, India
| |
Collapse
|
46
|
Inactivation of promoter 1B of APC causes partial gene silencing: evidence for a significant role of the promoter in regulation and causative of familial adenomatous polyposis. Oncogene 2011; 30:4977-89. [PMID: 21643010 PMCID: PMC3240859 DOI: 10.1038/onc.2011.201] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Familial adenomatous polyposis (FAP) is caused by germline mutations in the adenomatous polyposis coli (APC) gene. Two promoters, 1A and 1B, have been recognized in APC, and 1B is thought to have a minor role in the regulation of the gene. We have identified a novel deletion encompassing half of this promoter in the largest family (Family 1) of the Swedish Polyposis Registry. The mutation leads to an imbalance in allele-specific expression of APC, and transcription from promoter 1B was highly impaired in both normal colorectal mucosa and blood from mutation carriers. To establish the significance of promoter 1B in normal colorectal mucosa (from controls), expression levels of specific transcripts from each of the promoters, 1A and 1B, were examined, and the expression from 1B was significantly higher compared with 1A. Significant amounts of transcripts generated from promoter 1B were also determined in a panel of 20 various normal tissues examined. In FAP-related tumors, the APC germline mutation is proposed to dictate the second hit. Mutations leaving two or three out of seven 20-amino-acid repeats in the central domain of APC intact seem to be required for tumorigenesis. We examined adenomas from mutation carriers in Family 1 for second hits in the entire gene without any findings, however, loss of the residual expression of the deleterious allele was observed. Three major conclusions of significant importance in relation to the function of APC can be drawn from this study; (i) germline inactivation of promoter 1B is disease causing in FAP; (ii) expression of transcripts from promoter 1B is generated at considerable higher levels compared with 1A, demonstrating a hitherto unknown importance of 1B; (iii) adenoma formation in FAP, caused by impaired function of promoter 1B, does not require homozygous inactivation of APC allowing for alternative genetic models as basis for adenoma formation.
Collapse
|
47
|
David S, Meltzer SJ. Epigenetic Alterations as Contributors to the Pathogenesis, Detection, Prognosis and Treatment of Human Pre-invasive Neoplasia. PRE-INVASIVE DISEASE: PATHOGENESIS AND CLINICAL MANAGEMENT 2011:41-63. [DOI: 10.1007/978-1-4419-6694-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
48
|
Genomic and epigenetic profiles of gastric cancer: Potential diagnostic and therapeutic applications. Surg Today 2010; 41:24-38. [DOI: 10.1007/s00595-010-4370-5] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2010] [Accepted: 04/22/2010] [Indexed: 02/07/2023]
|
49
|
Schneider BG, Peng DF, Camargo MC, Piazuelo MB, Sicinschi LA, Mera R, Romero-Gallo J, Delgado AG, Bravo LE, Wilson KT, Peek RM, Correa P, El-Rifai W. Promoter DNA hypermethylation in gastric biopsies from subjects at high and low risk for gastric cancer. Int J Cancer 2010; 127:2588-97. [PMID: 20178103 PMCID: PMC2916942 DOI: 10.1002/ijc.25274] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gene promoter CpG island hypermethylation is associated with Helicobacter pylori (H. pylori) infection and may be an important initiator of gastric carcinogenesis. To examine factors influencing methylation, we utilized bisulfite Pyrosequencing® technology for quantitative analysis of promoter DNA methylation in RPRM, APC, MGMT and TWIST1 genes using DNA from 86 gastric biopsies from Colombian residents of areas with high and low incidence of gastric cancer. H. pylori colonies were cultured from the same subjects, and gastric pathology was evaluated. Virulence factors cagA (including segments of the 3' end, encoding EPIYA polymorphisms) and vacA s and m regions were characterized in the H. pylori strains. Using univariate analysis, we found significantly elevated levels of RPRM and TWIST1 promoter DNA methylation in biopsies from residents of the high-risk region compared to those from residents of the low-risk region. The presence of cagA and vacA s1m1 alleles were independently associated with elevated levels of promoter DNA methylation of RPRM and MGMT. Using multivariate analysis, DNA methylation of RPRM was associated with location of residence, cagA and vacA s1m1 status and methylation of TWIST1. We conclude that cagA and vacA virulence determinants are significantly associated with quantitative differences in promoter DNA methylation in these populations, but that other as yet undefined factors that differ between the populations may also contribute to variation in methylation status.
Collapse
Affiliation(s)
- Barbara G Schneider
- Division of Gastroenterology, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232-0252, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ali AHK, Kondo K, Namura T, Senba Y, Takizawa H, Nakagawa Y, Toba H, Kenzaki K, Sakiyama S, Tangoku A. Aberrant DNA methylation of some tumor suppressor genes in lung cancers from workers with chromate exposure. Mol Carcinog 2010; 50:89-99. [PMID: 21229606 DOI: 10.1002/mc.20697] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 08/25/2010] [Accepted: 09/28/2010] [Indexed: 12/16/2022]
Abstract
Our previous studies revealed a variety of genetic changes in lung cancers from chromate-exposed workers (chromate lung cancer). In the present study, we examined epigenetic changes in chromate lung cancers. Nested-methylation-specific PCR was employed in studying the methylation of CpG islands in the APC, MGMT, hMLH1 genes in 36 chromate lung cancers and 25 nonchromate lung cancers. Methylation in chromate lung cancers was detected at 86% for APC, 20% for MGMT, and 28% for hMLH1. Whereas, it occurred at lower frequencies in nonchromate lung cancers, particularly in APC (44%) and hMLH1 (0%) genes. Our previous study showed that methylation of p16 gene in chromate lung cancer and nonchromate lung cancer was 33% and 26%, respectively. The mean methylation index (MI), a reflection of the overall methylation status, was significantly higher in chromate lung cancers than nonchromate lung cancers (0.41 vs. 0.21, P=0.001). Methylation of multiple genes (particularly hMLH1, p16, and APC genes) had experienced more than 15 yr of chromate exposure in chromate lung cancer (MI: <15 yr; 0.19, ≥ 15 yr, 0.42). There is a significant correlation of p16 and hMLH1 methylation with the expressional decrease or loss of the corresponding gene products (P=0.037 and 0.024) respectively, and an inverse correlation between APC and MGMT methylation (P = 0.014). This study provides a novel evidence for the chromium carcinogenesis that chromate lung cancer is linked to the progressive methylation of some tumor suppressor genes, which may be related to genomic instability.
Collapse
Affiliation(s)
- Abdellah H K Ali
- Department of Respiratory Medicine, Sohag Faculty of Medicine, Sohag University, Sohag, Egypt
| | | | | | | | | | | | | | | | | | | |
Collapse
|