1
|
Wojtukiewicz MZ, Mysliwiec M, Tokajuk A, Kruszewska J, Politynska B, Jamroze A, Wojtukiewicz AM, Tang DG, Honn KV. Tissue factor pathway inhibitor-2 (TFPI-2)-an underappreciated partaker in cancer and metastasis. Cancer Metastasis Rev 2024; 43:1185-1204. [PMID: 39153052 PMCID: PMC11554837 DOI: 10.1007/s10555-024-10205-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024]
Abstract
The coagulation system is known to play an important role in cancer development and metastasis, but the precise mechanisms by which it does so remain incompletely understood. With this in mind, we provide an updated overview of the effects of TFPI-2, a protease inhibitor, on cancer development and metastasis. TFPI-2 interacts with the thrombin cascade and also employs other mechanisms to suppress cancer growth and dissemination, which include extracellular matrix stabilization, promotion of caspase-mediated cell apoptosis, inhibition of angiogenesis and transduction of intracellular signals. Down-regulation of TFPI-2 expression is well documented in numerous types of neoplasms, mainly via promoter methylation. However, the exact role of TFPI-2 in cancer progression and possible approaches to up-regulate TFPI-2 expression warrant further studies. Strategies to reactivate TFPI-2 may represent a promising direction for future anticancer studies and therapy development.
Collapse
Affiliation(s)
- Marek Z Wojtukiewicz
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland.
- Department of Clinical Oncology, Comprehensive Cancer Center of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland.
| | - Marta Mysliwiec
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland
| | - Anna Tokajuk
- Department of Clinical Oncology, Comprehensive Cancer Center of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland
| | - Joanna Kruszewska
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa, 15-027, Bialystok, Poland
| | - Barbara Politynska
- Department of Psychology and Philosophy, Medical University of Bialystok, 37 Szpitalna, 15-295, Bialystok, Poland
- Robinson College, University of Cambridge, Grange Road, Cambridge, CB3 9AN, UK
| | - Anmbreen Jamroze
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Anna M Wojtukiewicz
- Department of Psychology and Philosophy, Medical University of Bialystok, 37 Szpitalna, 15-295, Bialystok, Poland
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Kenneth V Honn
- Department of Pathology-School of Medicine, Bioactive Lipids Research Program, Wayne State University, 540 East Canfield Avenue, Detroit, MI, 48201, USA
- Karmanos Cancer Institute, 4100 John R St, Detroit, MI, 48201, USA
- Department of Chemistry, Wayne State University, 5101 Cass Ave, Detroit, MI, 48202, USA
| |
Collapse
|
2
|
Gu T, Vasilatos SN, Yin J, Qin Y, Zhang L, Davidson NE, Huang Y. Restoration of TFPI2 by LSD1 inhibition suppresses tumor progression and potentiates antitumor immunity in breast cancer. Cancer Lett 2024; 600:217182. [PMID: 39154703 PMCID: PMC11384719 DOI: 10.1016/j.canlet.2024.217182] [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: 05/06/2024] [Revised: 07/16/2024] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
Histone lysine-specific demethylase 1 (LSD1) is frequently overexpressed in triple negative breast cancer (TNBC), which is associated with worse clinical outcome in TNBC patients. However, the underlying mechanisms by which LSD1 promotes TNBC progression remain to be identified. We recently established a genetically engineered murine model by crossing mammary gland conditional LSD1 knockout mice with Brca1-deficient mice to explore the role of LSD1 in TNBC pathogenesis. Cre-mediated Brca1 loss led to higher incidence of tumor formation in mouse mammary glands, which was hindered by concurrent depletion of LSD1, indicating a critical role of LSD1 in promoting Brca1-deficient tumors. We also demonstrated that the silencing of a tumor suppressor gene, Tissue Factor Pathway Inhibitor 2 (TFPI2), is functionally associated with LSD1-mediated TNBC progression. Mouse Brca1-deficient tumors exhibited elevated LSD1 expression and decreased TFPI2 level compared to normal mammary tissues. Analysis of TCGA database revealed that TFPI2 expression is significantly lower in aggressive ER-negative or basal-like BC. Restoration of TFPI2 through LSD1 inhibition increased H3K4me2 enrichment at the TFPI2 promoter, suppressed tumor progression, and enhanced antitumor efficacy of chemotherapeutic agent. Induction of TFPI2 by LSD1 ablation downregulates activity of matrix metalloproteinases (MMPs) that in turn increases the level of cytotoxic T lymphocyte attracting chemokines in tumor environment, leading to enhanced tumor infiltration of CD8+ T cells. Moreover, induction of TFPI2 potentiates antitumor effect of LSD1 inhibitor and immune checkpoint blockade in poorly immunogenic TNBC. Together, our study identifies previously unrecognized roles of TFPI2 in LSD1-mediated TNBC progression, therapeutic response, and immunogenic effects.
Collapse
Affiliation(s)
- Tiezheng Gu
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shauna N Vasilatos
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jun Yin
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ye Qin
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lin Zhang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA, USA
| | - Nancy E Davidson
- Fred Hutchinson Cancer Center and University of Washington, Seattle, WA, USA
| | - Yi Huang
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA; Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Internal Medicine, Division of Hematology, Oncology, and Blood & Marrow Transplantation, Carver College of Medicine, University of Iowa, Iowa City, IA, USA; Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
3
|
Hassan N, Efing J, Kiesel L, Bendas G, Götte M. The Tissue Factor Pathway in Cancer: Overview and Role of Heparan Sulfate Proteoglycans. Cancers (Basel) 2023; 15:1524. [PMID: 36900315 PMCID: PMC10001432 DOI: 10.3390/cancers15051524] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/24/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023] Open
Abstract
Historically, the only focus on tissue factor (TF) in clinical pathophysiology has been on its function as the initiation of the extrinsic coagulation cascade. This obsolete vessel-wall TF dogma is now being challenged by the findings that TF circulates throughout the body as a soluble form, a cell-associated protein, and a binding microparticle. Furthermore, it has been observed that TF is expressed by various cell types, including T-lymphocytes and platelets, and that certain pathological situations, such as chronic and acute inflammatory states, and cancer, may increase its expression and activity. Transmembrane G protein-coupled protease-activated receptors can be proteolytically cleaved by the TF:FVIIa complex that develops when TF binds to Factor VII (PARs). The TF:FVIIa complex can activate integrins, receptor tyrosine kinases (RTKs), and PARs in addition to PARs. Cancer cells use these signaling pathways to promote cell division, angiogenesis, metastasis, and the maintenance of cancer stem-like cells. Proteoglycans play a crucial role in the biochemical and mechanical properties of the cellular extracellular matrix, where they control cellular behavior via interacting with transmembrane receptors. For TFPI.fXa complexes, heparan sulfate proteoglycans (HSPGs) may serve as the primary receptor for uptake and degradation. The regulation of TF expression, TF signaling mechanisms, their pathogenic effects, and their therapeutic targeting in cancer are all covered in detail here.
Collapse
Affiliation(s)
- Nourhan Hassan
- Department of Gynecology and Obstetrics, Münster University Hospital, Domagkstrasse 11, 48149 Münster, Germany
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Janes Efing
- Department of Gynecology and Obstetrics, Münster University Hospital, Domagkstrasse 11, 48149 Münster, Germany
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Domagkstrasse 11, 48149 Münster, Germany
| | - Gerd Bendas
- Pharmaceutical Department, University Bonn, An der Immenburg 4, 53225 Bonn, Germany
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Domagkstrasse 11, 48149 Münster, Germany
| |
Collapse
|
4
|
Ota Y, Koizume S, Nakamura Y, Yoshihara M, Takahashi T, Sato S, Myoba S, Ohtake N, Kato H, Yokose T, Miyagi E, Miyagi Y. Tissue factor pathway inhibitor‑2 is specifically expressed in ovarian clear cell carcinoma tissues in the nucleus, cytoplasm and extracellular matrix. Oncol Rep 2021; 45:1023-1032. [PMID: 33650653 PMCID: PMC7859994 DOI: 10.3892/or.2021.7944] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/22/2020] [Indexed: 12/21/2022] Open
Abstract
Tissue factor pathway inhibitor-2 (TFPI-2) is a promising candidate as a serum biomarker of ovarian clear cell carcinoma (OCCC), a lethal histological subtype of epithelial ovarian cancer (EOC). TFPI-2 is a secreted serine protease inhibitor that suppresses cancer progression through the inhibition of matrix protease activities. Previous studies have also identified TFPI-2 in the nucleus, and a possible function of nuclear TFPI-2 as a transcriptional repressor of matrix metalloproteinase-2 (MMP-2) was recently demonstrated. We are currently establishing TFPI-2 as a serum biomarker for OCCC patients; however, TFPI-2 expression in OCCC tissues has not been previously investigated. In the present study, we examined TFPI-2 expression and its localization in 11 OCCC cell lines by western blotting and enzyme-linked immune assay. Four cell lines expressed TFPI-2 in the nucleus, cytoplasm and culture plate–attached extracellular fraction, while four other cell lines expressed TFPI-2 only in the extracellular fraction. In the remaining three cell lines, TFPI-2 was not identified in any fraction. The amount of secreted soluble TFPI-2 showed similar trends to that of the plate-attached fraction. We next investigated the expression levels and distribution of TFPI-2 in surgically resected EOC tissues by immunohistochemistry. In 52 of the 77 (67.5%) OCCC tumors, TFPI-2 expression was detected in at least one of the nuclear, cytoplasmic and extracellular matrix fractions. In contrast, we did not identify TFPI-2 in the other EOC subtypes (n=65). TFPI-2-positive expression distinguished CCC from the other EOC tissues with a sensitivity of 67.5% and specificity of 100%. Although the inherent tumor suppressor function, statistical analyses failed to demonstrate correlations between TFPI-2 expression and clinical parameters, including 5-year overall survival, except for the patient age. In conclusion, we identified TFPI-2 expression in the nucleus, cytoplasm and extracellular matrix in OCCC tissues. The high specificity of TFPI-2 may support its use for diagnosis of OCCC in combination with existing markers.
Collapse
Affiliation(s)
- Yukihide Ota
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241‑8515, Japan
| | - Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241‑8515, Japan
| | - Yoshiyasu Nakamura
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241‑8515, Japan
| | - Mitsuyo Yoshihara
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241‑8515, Japan
| | - Tomoko Takahashi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241‑8515, Japan
| | - Shinya Sato
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241‑8515, Japan
| | - Shohei Myoba
- Bioscience Division, Reagent Development Department, Tosoh Corporation, Ayase, Kanagawa 252‑1123, Japan
| | - Norihisa Ohtake
- Bioscience Division, Reagent Development Department, Tosoh Corporation, Ayase, Kanagawa 252‑1123, Japan
| | - Hisamori Kato
- Department of Gynecologic Oncology, Kanagawa Cancer Center, Yokohama 241‑8515, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center, Yokohama 241‑8515, Japan
| | - Etsuko Miyagi
- Department of Obstetrics and Gynecology, Yokohama City University Graduate School of Medicine, Yokohama 236‑0004, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241‑8515, Japan
| |
Collapse
|
5
|
Li L, Yang L, Fan Z, Xue W, Shen Z, Yuan Y, Sun X, Wang D, Lian J, Wang L, Zhao J, Zhang Y. Hypoxia-induced GBE1 expression promotes tumor progression through metabolic reprogramming in lung adenocarcinoma. Signal Transduct Target Ther 2020; 5:54. [PMID: 32439898 PMCID: PMC7242448 DOI: 10.1038/s41392-020-0152-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/24/2020] [Accepted: 03/12/2020] [Indexed: 12/24/2022] Open
Abstract
Hypoxia mediates a metabolic switch from oxidative phosphorylation to glycolysis and increases glycogen synthesis. We previously found that glycogen branching enzyme (GBE1) is downstream of the hypoxia-inducible factor-1 (HIF1) signaling pathway in lung adenocarcinoma (LUAD) cells; however, the molecular mechanism underlying HIF1 regulation of GBE1 expression remains unknown. Herein, the effect of GBE1 on tumor progression via changes in metabolic signaling under hypoxia in vitro and in vivo was evaluated, and GBE1-related genes from human specimens and data sets were analyzed. Hypoxia induced GBE1 upregulation in LUAD cells. GBE1-knockdown A549 cells showed impaired cell proliferation, clone formation, cell migration and invasion, angiogenesis, tumor growth, and metastasis. GBE1 mediated the metabolic reprogramming of LUAD cells. The expression of gluconeogenesis pathway molecules, especially fructose-1,6-bisphosphatase (FBP1), was markedly higher in shGBE1 A549 cells than it was in the control cells. FBP1 inhibited the tumor progression of LUAD. GBE1-mediated FBP1 suppression via promoter methylation enhanced HIF1α levels through NF-κB signaling. GBE1 may be a negative prognostic biomarker for LUAD patients. Altogether, hypoxia-induced HIF1α mediated GBE1 upregulation, suppressing FBP1 expression by promoter methylation via NF-κB signaling in LUAD cells. FBP1 blockade upregulated HIF1α, triggered the switch to anaerobic glycolysis, and enhanced glucose uptake. Therefore, targeting HIF1α/GBE1/NF-κB/FBP1 signaling may be a potential therapeutic strategy for LUAD.
Collapse
Affiliation(s)
- Lifeng Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Internet Medical and System Applications of National Engineering Laboratory, Zhengzhou, China
| | - Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Zhirui Fan
- Integrated Traditional and Western Medicine, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Wenhua Xue
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Zhibo Shen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yongliang Yuan
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Xiangdong Sun
- Marshall B.J. Medical Research Centre, Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Dan Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Jingyao Lian
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Liping Wang
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Jie Zhao
- Internet Medical and System Applications of National Engineering Laboratory, Zhengzhou, China.
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
6
|
Yamamoto Y, Matsusaka K, Fukuyo M, Rahmutulla B, Matsue H, Kaneda A. Higher methylation subtype of malignant melanoma and its correlation with thicker progression and worse prognosis. Cancer Med 2020; 9:7194-7204. [PMID: 32406600 PMCID: PMC7541157 DOI: 10.1002/cam4.3127] [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: 01/11/2020] [Revised: 04/08/2020] [Accepted: 04/23/2020] [Indexed: 01/12/2023] Open
Abstract
Malignant melanoma (MM) is the most life‐threatening disease among all skin malignancies, and recent genome‐wide studies reported BRAF, RAS, and NF1 as the most frequently mutated driver genes. While epigenetic aberrations are known to contribute to the oncogenic activity seen in various cancers, their role in MM has not been fully investigated. To investigate the role of epigenetic aberrations in MM, we performed genome‐wide DNA methylation analysis of 51 clinical MM samples using Infinium 450k beadarray. Hierarchical clustering analysis stratified MM into two DNA methylation epigenotypes: high‐ and low‐methylation subgroups. Tumor thickness was significantly greater in case of high‐methylation tumors than low‐methylation tumors (8.3 ± 5.3 mm vs 4.5 ± 2.9 mm, P = .003). Moreover, prognosis was significantly worse in high‐methylation cases (P = .03). Twenty‐seven genes were found to undergo significant and frequent hypermethylation in high‐methylation subgroup, where TFPI2 was identified as the most frequently hypermethylated gene. MM cases with lower expression levels of TFPI2 showed significantly worse prognosis (P = .001). Knockdown of TFPI2 in two MM cell lines, CHL‐1 and G361, resulted in significant increases of cell proliferation and invasion. These indicate that MM can be stratified into at least two different epigenetic subgroups, that the MM subgroup with higher DNA methylation shows a more progressive phenotype, and that methylation of TFPI2 may contribute to the tumor progression of MM.
Collapse
Affiliation(s)
- Yosuke Yamamoto
- Department of Dermatology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Keisuke Matsusaka
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Masaki Fukuyo
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Genome Research and Development, Kazusa DNA Research Institute, Chiba, Japan
| | - Bahityar Rahmutulla
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroyuki Matsue
- Department of Dermatology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Kaneda
- Department of Molecular Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
7
|
Haen P, Mege D, Crescence L, Dignat-George F, Dubois C, Panicot-Dubois L. Thrombosis Risk Associated with Head and Neck Cancer: A Review. Int J Mol Sci 2019; 20:E2838. [PMID: 31212608 PMCID: PMC6600456 DOI: 10.3390/ijms20112838] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/30/2019] [Accepted: 06/07/2019] [Indexed: 12/12/2022] Open
Abstract
Venous thromboembolism (VTE) is a common complication for cancer patients. VTE-associated risk varies according to the type of tumor disease. Head and neck cancer is a common cancer worldwide, and most tumors are squamous cell carcinomas due to tobacco and alcohol abuse. The risk of VTE associated with head and neck (H&N) cancer is considered empirically low, but despite the high incidence of H&N cancer, few data are available on this cancer; thus, it is difficult to state the risk of VTE. Our review aims to clarify this situation and tries to assess the real VTE risk associated with H&N cancer. We report that most clinical studies have concluded that there is a very low thrombosis risk associated with H&N cancer. Even with the biases that often exist, this clinical review seems to confirm that the risk of VTE was empirically hypothesized. Furthermore, we highlight that H&N cancer has all the biological features of a cancer associated with a high thrombosis risk, including a strong expression of procoagulant proteins, modified thrombosis/fibrinolysis mechanisms, and secretions of procoagulant microparticles and procoagulant cytokines. Thus, this is a paradoxical situation, and some undiscovered mechanisms that could explain this clinical biological ambivalence might exist.
Collapse
Affiliation(s)
- Pierre Haen
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
- Department of Maxillo-Facial Surgery, Army Training Hospital, Laveran, 13013 Marseille, France.
| | - Diane Mege
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
- Department of Digestive Surgery, Timone University Hospital, AP-HM, 13005 Marseille, France.
| | - Lydie Crescence
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
| | - Françoise Dignat-George
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
- Laboratoire d'Hématologie, Centre Hospitalo-Universitaire Conception, 385 Boulevard Baille, 13385 Marseille, France.
| | - Christophe Dubois
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
| | - Laurence Panicot-Dubois
- Aix Marseille Univ, INSERM 1263, INRA, Center for CardioVascular and Nutrition Research (C2VN), 27 Boulevard Jean Moulin, 13385 Marseille, France.
| |
Collapse
|
8
|
Guo Y, Long J, Lei S. Promoter methylation as biomarkers for diagnosis of melanoma: A systematic review and meta-analysis. J Cell Physiol 2018; 234:7356-7367. [PMID: 30370527 DOI: 10.1002/jcp.27495] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 09/06/2018] [Indexed: 01/19/2023]
Abstract
Melanoma is one of the most common skin cancer that is characterized by rapid growth, early metastasis, high malignant, and mortality. Accumulating evidence demonstrated that promoter methylation of tumor-suppressor genes is implicated in the pathogenesis of melanoma. In the current study, we performed a meta-analysis to identify promising methylation biomarkers in the diagnosis of melanoma. We carried out a systematic literature search using Pubmed, Embase, and ISI web knowledge database and found that gene promoter methylation of 50 genes was reported to be associated with the risk of melanoma. Meta-analysis revealed that hypermethylation of claudin 11 (CLDN11; odds ratio [OR], 16.82; 95% confidence interval [CI], 1.97-143.29; p = 0.010), O-6-methylguanine-DNA methyltransferase (MGMT; OR, 5.59; 95% CI, 2.51-12.47; p < 0.0001), cyclin-dependent kinase inhibitor 2A (p16; OR, 6.57; 95% CI, 2.19-19.75; p = 0.0008), retinoic acid receptor β (RAR-β2; OR, 24.31; 95% CI, 4.58-129.01; p = 0.0002), and Ras association domain family member (RASSF1A; OR, 9.35; 95% CI, 4.73-18.45; p < 0.00001) was significantly higher in melanoma patients compared with controls. CLDN11 (OR, 14.52; 95% CI, 1.84-114.55; p = 0.01), MGMT (OR, 8.08; 95% CI, 1.84-35.46; p = 0.006), p16 (OR, 9.44; 95% CI, 2.68-33.29; p = 0.0005), and RASSF1A (OR, 7.72; 95% CI, 1.05-56.50; p = 0.04) hypermethylation was significantly increased in primary melanoma compared with controls. Methylation frequency of CLDN11 (OR, 25.56; 95% CI, 2.32-281.66; p = 0.008), MGMT (OR, 4.64; 95% CI, 1.98-10.90; p = 0.0004), p16 (OR, 4.31; 95% CI, 1.33-13.96; p = 0.01), and RASSF1A (OR, 10.10; 95% CI, 2.87-35.54; p = 0.0003) was significantly higher in metastasis melanoma compared with controls. These findings indicated that CLDN11, MGMT, p16, RAR-β2, and RASSF1A hypermethylation is a risk factor and a potential biomarker for melanoma. CLDN11, MGMT, p16, and RASSF1A promoter methylation may take part in the development of melanoma and become useful biomarkers in the early diagnosis of the disease.
Collapse
Affiliation(s)
- Yu Guo
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jianhong Long
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Shaorong Lei
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Andresen MS, Ali HO, Myklebust CF, Sandset PM, Stavik B, Iversen N, Skretting G. Estrogen induced expression of tissue factor pathway inhibitor-2 in MCF7 cells involves lysine-specific demethylase 1. Mol Cell Endocrinol 2017; 443:80-88. [PMID: 28088469 DOI: 10.1016/j.mce.2017.01.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 12/20/2016] [Accepted: 01/10/2017] [Indexed: 02/07/2023]
Abstract
Hormone-sensitive cancers can be influenced by estrogens, a process usually mediated through the estrogen receptor (ER). Tissue factor pathway inhibitor type 2 (TFPI-2) is a Kunitz-type serine protease inhibitor involved in regulating the extracellular matrix. The present study demonstrates that the expression of TFPI-2 can be induced by estrogens. Breast cancer data from GOBO displayed increased levels of TFPI-2 and increased survival in patients with ERα+ tumors. Treatment of MCF7 cells (ERα+) with 17β-estradiol (E2) or 17α-ethinyl estradiol (EE2) increased TFPI-2 mRNA and protein levels. This effect was mitigated with fulvestrant and by knocking down ERα, indicating that estrogen mediated TFPI-2 induction was through ERα. Upon knock down of DNA cytosine-5 methyltransferase 1 (DNMT1) or lysine-specific demethylase 1 (LSD1) in MCF7 cells, reduced effect of E2 on TFPI-2 mRNA levels was observed. Our data thus suggest that estrogen induced TFPI-2 expression in MCF7 cells is mediated by ERα and also by the action of LSD1.
Collapse
Affiliation(s)
- Marianne S Andresen
- Department of Haematology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway
| | - Huda Omar Ali
- Department of Haematology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Christiane Filion Myklebust
- Department of Haematology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway
| | - Per Morten Sandset
- Department of Haematology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Benedicte Stavik
- Department of Haematology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway
| | - Nina Iversen
- Dept. of Medical Genetics, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Grethe Skretting
- Department of Haematology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway.
| |
Collapse
|
10
|
Ghilardi C, Silini A, Figini S, Anastasia A, Lupi M, Fruscio R, Giavazzi R, Bani MR. Trypsinogen 4 boosts tumor endothelial cells migration through proteolysis of tissue factor pathway inhibitor-2. Oncotarget 2016; 6:28389-400. [PMID: 26318044 PMCID: PMC4695067 DOI: 10.18632/oncotarget.4949] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 07/02/2015] [Indexed: 11/25/2022] Open
Abstract
Proteases contribute to cancer in many ways, including tumor vascularization and metastasis, and their pharmacological inhibition is a potential anticancer strategy. We report that human endothelial cells (EC) express the trypsinogen 4 isoform of the serine protease 3 (PRSS3), and lack both PRSS2 and PRSS1. Trypsinogen 4 expression was upregulated by the combined action of VEGF-A, FGF-2 and EGF, angiogenic factors representative of the tumor microenvironment. Suppression of trypsinogen 4 expression by siRNA inhibited the angiogenic milieu-induced migration of EC from cancer specimens (tumor-EC), but did not affect EC from normal tissues. We identified tissue factor pathway inhibitor-2 (TFPI-2), a matrix associated inhibitor of cell motility, as the functional target of trypsinogen 4, which cleaved TFPI-2 and removed it from the matrix put down by tumor-EC. Silencing tumor-EC for trypsinogen 4 accumulated TFPI2 in the matrix. Showing that angiogenic factors stimulate trypsinogen 4 expression, which hydrolyses TFPI-2 favoring a pro-migratory situation, our study suggests a new pathway linking tumor microenvironment signals to endothelial cell migration, which is essential for angiogenesis and blood vessel remodeling. Abolishing trypsinogen 4 functions might be an exploitable strategy as anticancer, particularly anti-vascular, therapy.
Collapse
Affiliation(s)
- Carmen Ghilardi
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Antonietta Silini
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Sara Figini
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Alessia Anastasia
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Monica Lupi
- Laboratory of Cancer Pharmacology, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Robert Fruscio
- Clinic of Obstetrics and Gynecology, University of Milan-Bicocca, San Gerardo Hospital, Monza, Italy
| | - Raffaella Giavazzi
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| | - Maria Rosa Bani
- Laboratory of Biology and Treatment of Metastases, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy
| |
Collapse
|
11
|
Sun FK, Sun Q, Fan YC, Gao S, Zhao J, Li F, Jia YB, Liu C, Wang LY, Li XY, Ji XF, Wang K. Methylation of tissue factor pathway inhibitor 2 as a prognostic biomarker for hepatocellular carcinoma after hepatectomy. J Gastroenterol Hepatol 2016; 31:484-492. [PMID: 26313014 DOI: 10.1111/jgh.13154] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/06/2015] [Accepted: 08/09/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM Methylation of tissue factor pathway inhibitor 2 (TFPI2) gene has been detected in hepatocellular carcinoma (HCC). However, the clinicopathologcial significance and prognostic value of TFPI2 methylation in HCC remains largely unknown. This study aimed to investigate the prognostic value of TFPI2 methylation in HCC after hepatectomy. METHODS Methylation status of TFPI2 gene was examined in 178 surgical specimens of HCC and 20 normal liver samples using methylation-specific polymerase chain reaction. RESULTS Methylation of TFPI2 gene was detected in 44.9% (80 of 178) of primary HCC samples, 10.7% (19 of 178) of the corresponding non-tumorous liver samples, and 5.0% (1/20) of the normal liver samples. The mRNA concentrations of TFPI2 in primary HCC tissues were significantly lower than those in corresponding non-tumorous liver tissues and those in normal liver tissues. TFPI2 methylation was significantly associated with higher TNM stage. Patients with TFPI2 methylation demonstrated a significantly poorer prognosis than those without TFPI2 methylation for both overall survival and disease-free survival (P < 0.001, respectively). Multivariate analyses confirmed that TFPI2 methylation was an independent prognostic factor for both overall survival (P = 0.002) and disease-free survival (P = 0.000) in HCC after hepatectomy. Moreover, TFPI2 methylation was found to be the only independent predictor for early tumor recurrence of HCC after resection based on multivariate analysis (P = 0.002). CONCLUSIONS Methylation of TFPI2 predicts high risk of advanced tumor stage, early tumor recurrence, and poor prognosis, and it could be a potential prognostic biomarker in patients with HCC after hepatectomy.
Collapse
Affiliation(s)
- Feng-Kai Sun
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Qi Sun
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Yu-Chen Fan
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| | - Shuai Gao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Zhao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Feng Li
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Yi-Bin Jia
- Department of Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Chuan Liu
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Li-Yuan Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Xin-You Li
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiang-Fen Ji
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
| | - Kai Wang
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan, China
- Institute of Hepatology, Shandong University, Jinan, China
| |
Collapse
|
12
|
Yu J, Liu RL, Luo XP, Shi HM, Ma D, Pan JJ, Ni HC. Tissue Factor Pathway Inhibitor-2 Gene Polymorphisms Associate With Coronary Atherosclerosis in Chinese Population. Medicine (Baltimore) 2015; 94:e1675. [PMID: 26496276 PMCID: PMC4620828 DOI: 10.1097/md.0000000000001675] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tissue factor pathway inhibitor-2 (TFPI-2) may play critical roles in the pathogenesis of atherosclerosis. In this study, we aimed to investigate the association between TFPI-2 gene polymorphisms and coronary atherosclerosis.Four hundred and seven patients with coronary atherosclerosis and 306 individuals with normal coronary artery were enrolled in the present study. Nine single-nucleotide polymorphisms (SNPs) (rs3763473, rs59805398, rs60215632, rs59999573, rs59740167, rs34489123, rs4517, rs4264, and rs4271) were detected with polymerase chain reaction-direct sequencing method. Severity of coronary atherosclerosis was assessed by Gensini score. After the baseline investigation, patients with coronary atherosclerosis were followed up for incidence of cardiovascular events (CVEs).Eight SNPs were in accordance with the Hardy-Weinberg equilibrium, and 8 haplotypes were constructed based on rs59999573, rs59740167, and rs34489123 after linkage disequilibrium and haplotype analysis. Two SNPs (rs59805398 and rs34489123) and 5 haplotypes correlated with coronary atherosclerosis even after adjustment by Gensini score. At follow-up (median 53 months, range 1-60 months), 85 patients experienced CVE. However, there was no strong association between the gene polymorphisms and the occurrence of CVE.Tissue factor pathway inhibitor-2 gene polymorphisms were associated with coronary atherosclerosis in the Chinese population, suggesting that the information about TFPI-2 gene polymorphisms was useful for assessing the risk of developing coronary atherosclerosis, but there was not enough evidence showing it could predict occurrence of CVE.
Collapse
Affiliation(s)
- Jia Yu
- From the Department of Cardiology, Huashan Hospital, Fudan University, Shanghai, 200040, China (JY, R-IL, X-PL, H-MS, J-JP, H-CN); and Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, 200032, China (DM)
| | | | | | | | | | | | | |
Collapse
|
13
|
Cecchi F, Lih CJ, Lee YH, Walsh W, Rabe DC, Williams PM, Bottaro DP. Expression array analysis of the hepatocyte growth factor invasive program. Clin Exp Metastasis 2015; 32:659-76. [PMID: 26231668 DOI: 10.1007/s10585-015-9735-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 07/13/2015] [Indexed: 02/17/2023]
Abstract
Signaling by human hepatocyte growth factor (hHGF) via its cell surface receptor (MET) drives mitogenesis, motogenesis and morphogenesis in a wide spectrum of target cell types and embryologic, developmental and homeostatic contexts. Oncogenic pathway activation also contributes to tumorigenesis and cancer progression, including tumor angiogenesis and metastasis, in several prevalent malignancies. The HGF gene encodes full-length hHGF and two truncated isoforms known as NK1 and NK2. NK1 induces all three HGF activities at modestly reduced potency, whereas NK2 stimulates only motogenesis and enhances HGF-driven tumor metastasis in transgenic mice. Prior studies have shown that mouse HGF (mHGF) also binds with high affinity to human MET. Here we show that, like NK2, mHGF stimulates cell motility, invasion and spontaneous metastasis of PC3M human prostate adenocarcinoma cells in mice through human MET. To identify target genes and signaling pathways associated with motogenic and metastatic HGF signaling, i.e., the HGF invasive program, gene expression profiling was performed using PC3M cells treated with hHGF, NK2 or mHGF. Results obtained using Ingenuity Pathway Analysis software showed significant overlap with networks and pathways involved in cell movement and metastasis. Interrogating The Cancer Genome Atlas project also identified a subset of 23 gene expression changes in PC3M with a strong tendency for co-occurrence in prostate cancer patients that were associated with significantly decreased disease-free survival.
Collapse
Affiliation(s)
- Fabiola Cecchi
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1501, USA
| | - Chih-Jian Lih
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research, Inc. and Frederick National Laboratory for Cancer Research, Frederick, MD, 21702-1201, USA
| | - Young H Lee
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1501, USA
| | - William Walsh
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research, Inc. and Frederick National Laboratory for Cancer Research, Frederick, MD, 21702-1201, USA
| | - Daniel C Rabe
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1501, USA
| | - Paul M Williams
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research, Inc. and Frederick National Laboratory for Cancer Research, Frederick, MD, 21702-1201, USA
| | - Donald P Bottaro
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1501, USA. .,Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bldg 10 CRC Rm 2-3952, 10 Center Drive MSC 1107, Bethesda, MD, 20892-1107, USA.
| |
Collapse
|
14
|
Lai YH, He RY, Chou JL, Chan MWY, Li YF, Tai CK. Promoter hypermethylation and silencing of tissue factor pathway inhibitor-2 in oral squamous cell carcinoma. J Transl Med 2014; 12:237. [PMID: 25179542 PMCID: PMC4160550 DOI: 10.1186/s12967-014-0237-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 08/21/2014] [Indexed: 02/02/2023] Open
Abstract
Background The treatment of oral squamous cell carcinoma (OSCC) following early detection is associated with good outcomes. Therefore, the survival and prognosis of OSCC patients could be hugely improved by identifying reliable biomarkers for the early diagnosis of the disease. Our previous methylation microarray analysis results have suggested that the gene encoding tissue factor pathway inhibitor-2 (TFPI-2) is a potential clinical predictor as well as a key regulator involved in OSCC malignancy. Methods Methylation of the TFPI-2 promoter in oral tissue specimens was evaluated by bisulfite sequencing assay, quantitative methylation-specific PCR, and pyrosequencing assay. The differences in methylation levels among the groups were compared using the Mann–Whitney U test. The area under the receiver operating characteristic curve (AUROC) was used to evaluate the discrimination ability for detecting OSCC. Cellular TFPI-2 expression was analyzed by quantitative reverse-transcription PCR before and after treatment with 5′-aza-2′-deoxycytidine and trichostatin A, to confirm whether TFPI-2 was epigenetically silenced in OSCC cells. We investigated whether TFPI-2 plays a role as a tumor suppressor by establishing TFPI-2-overexpressing OSCC cells and subjecting them to in vitro cellular proliferation, migration, and invasion assays, as well as an in vivo metastasis assay. Results TFPI-2 was hypermethylated in OSCC tissues versus normal oral tissues (P < 0.0001), with AUROC = 0.91, when using a pyrosequencing assay to quantify the methylation level. TFPI-2 silencing in OSCC was regulated by both DNA methylation and chromatin histone modification. Restoration of TFPI-2 counteracted the invasiveness of OSCC by inhibiting the enzymatic activity of matrix metalloproteinase-2, and consequently interfered with OSCC metastasis in vivo. Conclusions Our data suggest strongly that TFPI-2 is a down-regulated tumor suppressor gene in OSCC, probably involving epigenetic silencing mechanisms. The loss of TFPI-2 expression is a key event for oral tumorigenesis, especially in the process of tumor metastasis.
Collapse
Affiliation(s)
| | | | | | | | - Yu-Fen Li
- Department of Life Science and Institutes of Molecular Biology and Biomedical Science, National Chung Cheng University, Min-Hsiung, Chia-Yi, Taiwan.
| | | |
Collapse
|
15
|
Leppert U, Eisenreich A. The role of tissue factor isoforms in cancer biology. Int J Cancer 2014; 137:497-503. [PMID: 24806794 DOI: 10.1002/ijc.28959] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 05/02/2014] [Indexed: 12/17/2022]
Abstract
Tissue Factor (TF) is an evolutionary conserved glycoprotein, which is of immense importance for a variety of biologic processes. TF is expressed in two naturally occurring protein isoforms, membrane-bound "full-length" (fl)TF and soluble alternatively spliced (as)TF. The TF isoform expression is differentially modulated on post-transcriptional level via regulatory factors, such as serine/arginine-rich (SR) proteins, SR protein kinases and micro (mi)RNAs. Both isoforms mediate a variety of physiologic- and pathophysiologic-relevant functions, such as thrombogenicity, angiogenesis, cell signaling, tumor cell proliferation and metastasis. In this review, we will depict the main mechanisms regulating the TF isoform expression in cancer and under other pathophysiologic-relevant conditions. Moreover, we will summarize and discuss the latest findings regarding the role of TF and its isoforms in cancer biology.
Collapse
Affiliation(s)
- Ulrike Leppert
- Charité - Universitätsmedizin Berlin, Campus Mitte, Charite Centrum 04/13, Berlin, Germany
| | - Andreas Eisenreich
- Charité - Universitätsmedizin Berlin, Campus Mitte, Charite Centrum 04/13, Berlin, Germany
| |
Collapse
|
16
|
Yue D, Fan Q, Chen X, Li F, Wang L, Huang L, Dong W, Chen X, Zhang Z, Liu J, Wang F, Wang M, Zhang B, Zhang Y. Epigenetic inactivation of SPINT2 is associated with tumor suppressive function in esophageal squamous cell carcinoma. Exp Cell Res 2014; 322:149-158. [PMID: 24269829 DOI: 10.1016/j.yexcr.2013.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 11/11/2013] [Accepted: 11/13/2013] [Indexed: 10/26/2022]
Abstract
Hepatocyte growth factor activator inhibitor type 2 (SPINT2), a Kunitz-type serine proteinase inhibitor, has been identified as a putative tumor suppressor gene silenced by promoter methylation. We aimed to investigate whether SPINT2 might act as an esophageal squamous cell carcinoma (ESCC) tumor suppressor gene. Four ESCC cell lines, Fifty-two ESCC tissues and twenty-nine neighboring non-cancerous tissues were included in this study. The expression of SPINT2 was monitored by real time PCR. Bisulfite genomic sequencing and methylation-specific PCR were used to analyze methylation status. The effect of SPINT2 on cell proliferation and apoptosis in EC109 and EC9706 cells was observed by CCK-8 assay and flow cytometric analysis. We found that silencing of SPINT2 was associated with promoter methylation in ESCC cell lines. The densely methylated SPINT2 promoter region was confirmed by bisulfite genomic sequencing. Ectopic expression of SPINT2 inhibited cell proliferation through inducing cell apoptosis in vitro. Furthermore, methylation-specific PCR analysis revealed that SPINT2 promoter methylation was prominent in carcinoma tissues (52.08%) compared with neighboring non-cancerous tissues (22.58%). Kaplan-Meier analysis showed that patients with SPINT2 hypermethylation had shorter survival time. The tumor suppressor gene of SPINT2 is commonly silenced by promoter hypermethylation in human ESCC and SPINT2 hypermethylation is correlated with poor overall survival, implicating SPINT2 is an underlying prognostic marker for human ESCC.
Collapse
Affiliation(s)
- Dongli Yue
- The Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; The Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Qingxia Fan
- The Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xinfeng Chen
- The Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Feng Li
- The Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Liping Wang
- The Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Lan Huang
- The Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Wenjie Dong
- The Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Xiaoqi Chen
- The Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Zhen Zhang
- The Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Jinyan Liu
- The Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; The School of Life Sciences, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Fei Wang
- The Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; The School of Life Sciences, Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Meng Wang
- The Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; The Department of Gastroenterology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Bin Zhang
- The Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; The Department of Hematology/Oncology, School of Medicine, Northwestern University, Chicago 60611, USA
| | - Yi Zhang
- The Biotherapy Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; The Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; The School of Life Sciences, Zhengzhou University, Zhengzhou 450052, Henan, China; The institute of Clinical-Medicine, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China.
| |
Collapse
|
17
|
Wang GJ, Wang YB, Li DIN, Li C, Deng BB. Expression of tissue factor pathway inhibitor-2 in gastric stromal tumor and its clinical significance. Exp Ther Med 2013; 7:513-517. [PMID: 24396436 PMCID: PMC3881048 DOI: 10.3892/etm.2013.1448] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 12/02/2013] [Indexed: 01/30/2023] Open
Abstract
The aim of this study was to explore the expression of tissue factor pathway inhibitor-2 (TFPI-2) in gastric stromal tissue and its clinical significance. TFPI-2 expression was detected by immunohistochemical analysis, RT-PCR and western blotting in tumor, peritumoral and gastric normal tissues from 72 patients with gastric stromal tumors. The level of TFPI-2 expression was observed to be significantly higher in gastric normal tissue than in peritumoral tissue, and was significantly higher in peritumoral tissue than in tumor tissue (P<0.01). As the NIH grade increased, the level of TFPI-2 expression decreased (P<0.01). A low expression level of TFPI-2 was closely associated with invasion and metastasis of gastric stromal tumors. In conclusion, the level of TFPI-2 expression was higher in gastric normal tissue than in gastric stromal tumors. Low expression levels of TFPI-2 may be associated with invasion and metastasis of gastric stromal tumors.
Collapse
Affiliation(s)
- Gui-Jun Wang
- Department of General Surgery, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Yu-Bin Wang
- Department of General Surgery, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - DI-Nuo Li
- Department of General Surgery, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Chen Li
- Liaoning Province Key Laboratory of Medical Tissue Engineering, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Bei-Bei Deng
- Department of Gynecology, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| |
Collapse
|
18
|
Lavergne M, Jourdan ML, Blechet C, Guyetant S, Pape AL, Heuze-Vourc'h N, Courty Y, Lerondel S, Sobilo J, Iochmann S, Reverdiau P. Beneficial role of overexpression of TFPI-2 on tumour progression in human small cell lung cancer. FEBS Open Bio 2013; 3:291-301. [PMID: 23905012 PMCID: PMC3722576 DOI: 10.1016/j.fob.2013.06.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/05/2013] [Accepted: 06/23/2013] [Indexed: 12/18/2022] Open
Abstract
Tissue factor pathway inhibitor-2 (TFPI-2) is a potent inhibitor of plasmin, a protease which is involved in tumour progression by activating (MMPs). This therefore makes TFPI-2 a potential inhibitor of invasiveness and the development of metastases. In this study, low levels of TFPI-2 expression were found in 65% of patients with small cell lung cancer (SCLC), the most aggressive type of lung cancer. To study the impact of TFPI-2 in tumour progression, TFPI-2 was overexpressed in NCI-H209 SCLC cells which were orthotopically implanted in nude mice. Investigations showed that TFPI-2 inhibited lung tumour growth. Such inhibition could be explained in vitro by a decrease in tumour cell viability, blockade of G1/S phase cell cycle transition and an increase in apoptosis shown in NCI-H209 cells expressing TFPI-2. We also demonstrated that TFPI-2 upregulation in NCI-H209 cells decreased MMP expression, particularly by downregulating MMP-1 and MMP-3. Moreover, TFPI-2 inhibited phosphorylation of the MAPK signalling pathway proteins involved in the induction of MMP transcripts, among which MMP-1 was predominant in SCLC tissues and was inversely expressed with TFPI-2 in 35% of cases. These results suggest that downregulation of TFPI-2 expression could favour the development of SCLC. The Tissue Factor Pathway Inhibitor-2 inhibits small cell lung cancer growth Monitoring of small cell lung cancer growth in a mouse orthotopic model by imaging Increasing information on the role of TFPI-2 in human lung tumour cells Increasing information on TFPI-2 and protease expression in human tissue samples
Collapse
Affiliation(s)
- Marion Lavergne
- EA 6305, Université François Rabelais de Tours, Tours F-37032, France ; Centre d'Etude des Pathologies Respiratoires, UMR 1100/EA6305, Tours F-37032, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Jazirehi AR, Torres-Collado AX, Nazarian R. Research Highlights: Highlights from the latest articles in epigenomics. Epigenomics 2013. [DOI: 10.2217/epi.13.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Ali R Jazirehi
- Department of Surgery, Division of Surgical Oncology, & Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, USA.
| | - Antoni X Torres-Collado
- Department of Surgery, Division of Surgical Oncology, & Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Ramin Nazarian
- Department of Surgery, Division of Surgical Oncology, & Jonsson Comprehensive Cancer Center, David Geffen School of Medicine at University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
20
|
Nigro CL, Wang H, McHugh A, Lattanzio L, Matin R, Harwood C, Syed N, Hatzimichael E, Briasoulis E, Merlano M, Evans A, Thompson A, Leigh I, Fleming C, Inman GJ, Proby C, Crook T. Methylated Tissue Factor Pathway Inhibitor 2 (TFPI2) DNA in Serum Is a Biomarker of Metastatic Melanoma. J Invest Dermatol 2013; 133:1278-85. [DOI: 10.1038/jid.2012.493] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
21
|
Jia Y, Yang Y, Brock MV, Cao B, Zhan Q, Li Y, Yu Y, Herman JG, Guo M. Methylation of TFPI-2 is an early event of esophageal carcinogenesis. Epigenomics 2012; 4:135-46. [PMID: 22449186 DOI: 10.2217/epi.12.11] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIMS To explore the epigenetic changes and the function of TFPI-2 in esophageal cancer. MATERIALS & METHODS Nine esophageal cancer cell lines, nine normal esophageal mucosa, 60 esophageal dysplasia and 106 advanced esophageal cancer samples were included in this study. TFPI-2 methylation was examined by methylation-specific PCR. TFPI-2 expression was evaluated by immunohistochemistry in tissue samples. The effect of TFPI-2 on proliferation, apoptosis, invasion and migration was analyzed by colony formation assay, western blot assay, transwell assay and flow cytometric analysis. RESULTS TFPI-2 expression was regulated by promoter region hypermethylation in human esophageal cancer cell lines, and TFPI-2 expression is inversely correlated with methylation in primary cancer. Methylation was found in 28.2, 33.3 and 33.3% of grade 1, 2 and 3 esophageal dysplasia, and 67% of primary esophageal cancer, but no methylation was found in normal mucosa. Methylation is significantly related to tumor differentiation. Inhibition of invasion, migration, colony formation and proliferation, and induction of apoptosis occurred with the restoration of TFPI-2 expression in the KYSE70 cell line. CONCLUSION TFPI-2 is frequently methylated in esophageal cancer with a progression tendency. TFPI-2 is a potential tumor suppressor in esophageal cancer.
Collapse
Affiliation(s)
- Yan Jia
- Department of Gastroenterology & Hepatology, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing 100853, China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Gaud G, Iochmann S, Guillon-Munos A, Brillet B, Petiot S, Seigneuret F, Touzé A, Heuzé-Vourc'h N, Courty Y, Lerondel S, Gruel Y, Reverdiau P. TFPI-2 silencing increases tumour progression and promotes metalloproteinase 1 and 3 induction through tumour-stromal cell interactions. J Cell Mol Med 2012; 15:196-208. [PMID: 20015200 PMCID: PMC3822788 DOI: 10.1111/j.1582-4934.2009.00989.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Tissue factor pathway inhibitor-2 (TFPI-2) is a potent inhibitor of plasmin which activates matrix metalloproteinases (MMPs) involved in degradation of the extracellular matrix. Its secretion in the tumour microenvironment makes TFPI-2 a potential inhibitor of tumour invasion and metastasis. As demonstrated in aggressive cancers, TFPI-2 is frequently down-regulated in cancer cells, but the mechanisms involved in the inhibition of tumour progression remained unclear. We showed in this study that stable TFPI-2 down-regulation in the National Cancer Institute (NCI)-H460 non-small cell lung cancer cell line using specific micro interfering micro-interfering RNA promoted tumour progression in a nude mice orthotopic model that resulted in an increase in cell invasion. Moreover, TFPI-2 down-regulation enhanced cell adhesion to collagen IV and laminin via an increase in α1 integrin on cell surface, and increased MMP expression (mainly MMP-1 and -3) contributing to cancer cell invasion through basement membrane components. This study also reveals for the first time that pulmonary fibroblasts incubated with conditioned media from TFPI-2 silencing cancer cells exhibited increased expression of MMPs, particularly MMP-1, -3 and -7, that are likely involved in lung cancer cell invasion through the surrounding stromal tissue, thus enhancing formation of metastases.
Collapse
Affiliation(s)
- Guillaume Gaud
- Inserm, U618, Université François Rabelais, Tours, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Genetics and epigenetics of cutaneous malignant melanoma: a concert out of tune. Biochim Biophys Acta Rev Cancer 2012; 1826:89-102. [PMID: 22503822 DOI: 10.1016/j.bbcan.2012.03.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 03/09/2012] [Accepted: 03/10/2012] [Indexed: 01/05/2023]
Abstract
Cutaneous malignant melanoma (CMM) is the most life-threatening neoplasm of the skin and is considered a major health problem as both incidence and mortality rates continue to rise. Once CMM has metastasized it becomes therapy-resistant and is an inevitably deadly disease. Understanding the molecular mechanisms that are involved in the initiation and progression of CMM is crucial for overcoming the commonly observed drug resistance as well as developing novel targeted treatment strategies. This molecular knowledge may further lead to the identification of clinically relevant biomarkers for early CMM detection, risk stratification, or prediction of response to therapy, altogether improving the clinical management of this disease. In this review we summarize the currently identified genetic and epigenetic alterations in CMM development. Although the genetic components underlying CMM are clearly emerging, a complete picture of the epigenetic alterations on DNA (DNA methylation), RNA (non-coding RNAs), and protein level (histone modifications, Polycomb group proteins, and chromatin remodeling) and the combinatorial interactions between these events is lacking. More detailed knowledge, however, is accumulating for genetic and epigenetic interactions in the aberrant regulation of the INK4b-ARF-INK4a and microphthalmia-associated transcription factor (MITF) loci. Importantly, we point out that it is this interplay of genetics and epigenetics that effectively leads to distorted gene expression patterns in CMM.
Collapse
|
24
|
Abstract
Genetics has fascinated societies since ancient times, and references to traits or behaviors that appear to be shared or different among related individuals have permeated legends, literature, and popular culture. Biomedical advances from the past century, and particularly the discovery of the DNA double helix, the increasing numbers of links that were established between mutations and medical conditions or phenotypes, and technological advances that facilitated the sequencing of the human genome, catalyzed the development of genetic testing. Genetic tests were initially performed in health care facilities, interpreted by health care providers, and included the availability of counseling. Recent years have seen an increased availability of genetic tests that are offered by companies directly to consumers, a phenomenon that became known as direct-to-consumer genetic testing. Tests offered in this setting range from the ones that are also provided in health care establishments to tests known as ‘recreational genomics,’ and consumers directly receive the test results. In addition, testing in this context often does not involve the availability of counseling and, when this is provided, it frequently occurs on-line or over the phone. As a field situated at the interface between biotechnology, biomedical research, and social sciences, direct-to-consumer genetic testing opens multiple challenges that can be appropriately addressed only by developing a complex, inter-disciplinary framework.
Collapse
|
25
|
|
26
|
TFPI-2 methylation predicts poor prognosis in non-small cell lung cancer. Lung Cancer 2011; 76:106-11. [PMID: 21983100 DOI: 10.1016/j.lungcan.2011.09.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2011] [Revised: 08/03/2011] [Accepted: 09/06/2011] [Indexed: 12/25/2022]
Abstract
BACKGROUND Methylation of human tissue factor pathway inhibitor-2 (TFPI-2) gene has been detected in several types of cancer, including non-small cell lung cancer (NSCLC). However, an association between the methylation status of TFPI-2 gene and prognosis has not yet been investigated. METHODS Methylation of TFPI-2 gene was examined in a consecutive series of 133 non-metastatic NSCLC patients using methylation-specific PCR (MSP). Univariate and multivariate analyses were conducted to investigate the association between clinical variables and overall survival time. RESULTS Methylation of TFPI-2 gene was detected in 36 of 133 patients (27.1%). Of these 36 patients, seventeen individuals (47.2%) carried stage III tumors. The 5-year disease free survival rate among patients carrying methylated TFPI-2 tumors was significantly lower as compared to those with unmethylated TFPI-2 tumors (35.5% versus 6.1%, P<0.0001). Moreover, methylation of TFPI-2 gene was found to be an independent prognostic factor for poor overall survival based on multivariate analysis models (P=0.013), as was age >62 years old (P<0.0001) and TNM stage of disease (P<0.0001). CONCLUSIONS The results of the present study suggest that methylation of TFPI-2 gene is an independent factor for an unfavorable prognosis in patients with NSCLC.
Collapse
|
27
|
Xu Y, Qin X, Zhou J, Tu Z, Bi X, Li W, Fan X, Zhang Y. Tissue factor pathway inhibitor-2 inhibits the growth and invasion of hepatocellular carcinoma cells and is inactivated in human hepatocellular carcinoma. Oncol Lett 2011; 2:779-783. [PMID: 22866126 DOI: 10.3892/ol.2011.340] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 05/11/2011] [Indexed: 11/06/2022] Open
Abstract
Human tissue factor pathway inhibitor-2 (TFPI-2) is an extracellular matrix-associated Kunitz-type serine proteinase inhibitor that inhibits the plasmin- and trypsin-mediated activation of matrix metalloproteinases and inhibits tumor progression, invasion and metastasis. Previous studies have shown that TFPI-2 is downregulated in the progression of various tumors. The purpose of this study was to investigate the expression and function of TFPI-2 in hepatocellular carcinoma (HCC). In situ hybridization was used to detect human TFPI-2 mRNA and immunohistochemistry was performed to examine the role of TFPI-2 expression in hepatocarcinoma tissues. Cell proliferation was assessed using MTT assay. In situ hybridization and immunohistochemical analyses revealed that the expression of TFPI-2 in hepatocarcinoma tissues was markedly lower than that in tumor-adjacent normal hepatic tissues. Restored expression of TFPI-2 in HepG(2) cells inhibits cell proliferation and invasion. Taken together, the results suggest that TFPI-2 has a tumor-suppression action and its inactivation may contribute to HCC.
Collapse
Affiliation(s)
- Yong Xu
- Pingshan People's Hospital, Pingshan, Shenzhen, Guangdong 518118, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Hattori N, Okochi-Takada E, Kikuyama M, Wakabayashi M, Yamashita S, Ushijima T. Methylation silencing of angiopoietin-like 4 in rat and human mammary carcinomas. Cancer Sci 2011; 102:1337-43. [PMID: 21489049 DOI: 10.1111/j.1349-7006.2011.01955.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aberrant DNA methylation is deeply involved in the development and progression of human breast cancers, but its inducers and molecular mechanisms are still unclear. To reveal such inducers and clarify the molecular mechanisms, animal models are indispensable. Here, to identify genes silenced by promoter DNA methylation in rat mammary carcinomas, we took a combined approach of methylated DNA immunoprecipitation (MeDIP)-CpG island (CGI) microarray analysis and expression microarray analysis after treatment with epigenetic drugs. MeDIP-CGI microarray revealed that among 5031 genes with promoter CGI, 465 were methylated in a carcinoma cell line induced by 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), but not in normal mammary epithelial cells. By treatment of the cell line with 5-aza-2'-deoxycytidine and trichostatin A, 29 of the 465 genes were shown to be re-expressed. In primary mammary carcinomas, five (Angptl4, Coro1a, RGD1304982, Tmem37 and Ndn) of the 29 genes were methylated in one or more of 25 samples. Quantitative expression analysis revealed that Angptl4 had high expression in normal mammary glands, but low expression in primary carcinomas. Also in humans, ANGPTL4 was unmethylated and expressed in normal mammary epithelial cells, but was methylated in 11 of 91 (12%) primary breast cancers. This is the first study to identify genes aberrantly methylated in rat mammary carcinomas, and Angptl4 is a novel methylation-silenced gene both in rat and human mammary carcinomas. The combination of the MeDIP-CGI microarray analysis and expression microarray analysis after treatment with epigenetic drugs was effective in reducing the number of methylated genes that are not methylation silenced.
Collapse
Affiliation(s)
- Naoko Hattori
- Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Ma S, Chan YP, Kwan PS, Lee TK, Yan M, Tang KH, Ling MT, Vielkind JR, Guan XY, Chan KW. MicroRNA-616 Induces Androgen-Independent Growth of Prostate Cancer Cells by Suppressing Expression of Tissue Factor Pathway Inhibitor TFPI-2. Cancer Res 2011; 71:583-92. [DOI: 10.1158/0008-5472.can-10-2587] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
30
|
Wang S, Xiao X, Zhou X, Huang T, Du C, Yu N, Mo Y, Lin L, Zhang J, Ma N, Murata M, Huang G, Zhang Z. TFPI-2 is a putative tumor suppressor gene frequently inactivated by promoter hypermethylation in nasopharyngeal carcinoma. BMC Cancer 2010; 10:617. [PMID: 21062455 PMCID: PMC2992524 DOI: 10.1186/1471-2407-10-617] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 11/09/2010] [Indexed: 12/13/2022] Open
Abstract
Background Epigenetic silencing of tumor suppressor genes play important roles in NPC tumorgenesis. Tissue factor pathway inhibitor-2 (TFPI-2), is a protease inhibitor. Recently, TFPI-2 was suggested to be a tumor suppressor gene involved in tumorigenesis and metastasis in some cancers. In this study, we investigated whether TFPI-2 was inactivated epigenetically in nasopharyngeal carcinoma (NPC). Methods Transcriptional expression levels of TFPI-2 was evaluated by RT-PCR. Methylation status were investigated by methylation specific PCR and bisulfate genomic sequencing. The role of TFPI-2 as a tumor suppressor gene in NPC was addressed by re-introducing TFPI-2 expression into the NPC cell line CNE2. Results TFPI-2 mRNA transcription was inactivated in NPC cell lines. TFPI-2 was aberrantly methylated in 66.7% (4/6) NPC cell lines and 88.6% (62/70) of NPC primary tumors, but not in normal nasopharyngeal epithelia. TFPI-2 expression could be restored in NPC cells after demethylation treatment. Ectopic expression of TFPI-2 in NPC cells induced apoptosis and inhibited cell proliferation, colony formation and cell migration. Conclusions Epigenetic inactivation of TFPI-2 by promoter hypermethylation is a frequent and tumor specific event in NPC. TFPI-2 might be considering as a putative tumor suppressor gene in NPC.
Collapse
Affiliation(s)
- Shumin Wang
- Department of Otolaryngology Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Malignant melanoma remains one of the most deadly human cancers with no effective cures for metastatic disease. The poor efficacy of current therapy in advanced melanoma highlights the need for better understanding of molecular mechanisms contributing to the disease. Recent work has shown that epigenetic changes, including aberrant DNA methylation, lead to alterations in gene expression and are as important in the development of malignant melanoma as the specific and well-characterized genetic events. Reversion of these methylation patterns could thus lead to a more targeted therapy and are currently under clinical investigation. The purpose of this review is to compile recent information on aberrant DNA methylation of melanoma, to highlight key genes and molecular pathways in melanoma development, which have been found to be epigenetically altered and to provide insight as to how DNA methylation might serve as targeted treatment option as well as a molecular and prognostic marker in malignant melanoma.
Collapse
|
32
|
Metallothionein 1E is methylated in malignant melanoma and increases sensitivity to cisplatin-induced apoptosis. Melanoma Res 2010. [DOI: 10.1097/cmr.0b013e32833d32a6] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Sigalotti L, Covre A, Fratta E, Parisi G, Colizzi F, Rizzo A, Danielli R, Nicolay HJM, Coral S, Maio M. Epigenetics of human cutaneous melanoma: setting the stage for new therapeutic strategies. J Transl Med 2010; 8:56. [PMID: 20540720 PMCID: PMC2901206 DOI: 10.1186/1479-5876-8-56] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 06/11/2010] [Indexed: 12/15/2022] Open
Abstract
Cutaneous melanoma is a very aggressive neoplasia of melanocytic origin with constantly growing incidence and mortality rates world-wide. Epigenetic modifications (i.e., alterations of genomic DNA methylation patterns, of post-translational modifications of histones, and of microRNA profiles) have been recently identified as playing an important role in melanoma development and progression by affecting key cellular pathways such as cell cycle regulation, cell signalling, differentiation, DNA repair, apoptosis, invasion and immune recognition. In this scenario, pharmacologic inhibition of DNA methyltransferases and/or of histone deacetylases were demonstrated to efficiently restore the expression of aberrantly-silenced genes, thus re-establishing pathway functions. In light of the pleiotropic activities of epigenetic drugs, their use alone or in combination therapies is being strongly suggested, and a particular clinical benefit might be expected from their synergistic activities with chemo-, radio-, and immuno-therapeutic approaches in melanoma patients. On this path, an important improvement would possibly derive from the development of new generation epigenetic drugs characterized by much reduced systemic toxicities, higher bioavailability, and more specific epigenetic effects.
Collapse
Affiliation(s)
- Luca Sigalotti
- Cancer Bioimmunotherapy Unit, Centro di Riferimento Oncologico, Istituto di Ricovero e Cura a Carattere Scientifico, Via F, Gallini 2, Aviano, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kabbarah O, Nogueira C, Feng B, Nazarian RM, Bosenberg M, Wu M, Scott KL, Kwong LN, Xiao Y, Cordon-Cardo C, Granter SR, Ramaswamy S, Golub T, Duncan LM, Wagner SN, Brennan C, Chin L. Integrative genome comparison of primary and metastatic melanomas. PLoS One 2010; 5:e10770. [PMID: 20520718 PMCID: PMC2875381 DOI: 10.1371/journal.pone.0010770] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 04/30/2010] [Indexed: 12/19/2022] Open
Abstract
A cardinal feature of malignant melanoma is its metastatic propensity. An incomplete view of the genetic events driving metastatic progression has been a major barrier to rational development of effective therapeutics and prognostic diagnostics for melanoma patients. In this study, we conducted global genomic characterization of primary and metastatic melanomas to examine the genomic landscape associated with metastatic progression. In addition to uncovering three genomic subclasses of metastastic melanomas, we delineated 39 focal and recurrent regions of amplification and deletions, many of which encompassed resident genes that have not been implicated in cancer or metastasis. To identify progression-associated metastasis gene candidates, we applied a statistical approach, Integrative Genome Comparison (IGC), to define 32 genomic regions of interest that were significantly altered in metastatic relative to primary melanomas, encompassing 30 resident genes with statistically significant expression deregulation. Functional assays on a subset of these candidates, including MET, ASPM, AKAP9, IMP3, PRKCA, RPA3, and SCAP2, validated their pro-invasion activities in human melanoma cells. Validity of the IGC approach was further reinforced by tissue microarray analysis of Survivin showing significant increased protein expression in thick versus thin primary cutaneous melanomas, and a progression correlation with lymph node metastases. Together, these functional validation results and correlative analysis of human tissues support the thesis that integrated genomic and pathological analyses of staged melanomas provide a productive entry point for discovery of melanoma metastases genes.
Collapse
Affiliation(s)
- Omar Kabbarah
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Cristina Nogueira
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), University of Porto, Porto, Portugal
| | - Bin Feng
- Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Rosalynn M. Nazarian
- Dermatopathology Unit, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Marcus Bosenberg
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Min Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kenneth L. Scott
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Lawrence N. Kwong
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yonghong Xiao
- Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Carlos Cordon-Cardo
- Department of Pathology, Columbia University, New York, New York, United States of America
| | - Scott R. Granter
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Sridhar Ramaswamy
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, United States of America
| | - Todd Golub
- The Broad Institute of MIT and Harvard and Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Lyn M. Duncan
- Dermatopathology Unit, Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Stephan N. Wagner
- DIAID, Department of Dermatology, Medical University of Vienna and Center of Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Cameron Brennan
- HOPP, Department of Neurosurgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- * E-mail: (CB); (LC)
| | - Lynda Chin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, United States of America
- Belfer Institute for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Dermatology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (CB); (LC)
| |
Collapse
|
35
|
Takada H, Wakabayashi N, Dohi O, Yasui K, Sakakura C, Mitsufuji S, Taniwaki M, Yoshikawa T. Tissue factor pathway inhibitor 2 (TFPI2) is frequently silenced by aberrant promoter hypermethylation in gastric cancer. ACTA ACUST UNITED AC 2010; 197:16-24. [PMID: 20113832 DOI: 10.1016/j.cancergencyto.2009.11.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2009] [Revised: 10/04/2009] [Accepted: 11/02/2009] [Indexed: 12/16/2022]
Abstract
Aberrant methylation of promoter CpG islands is associated with transcriptional inactivation of tumor-suppressor genes in cancer. TFPI2, a Kunitz-type serine proteinase inhibitor, has been identified as a putative tumor-suppressor gene from genome-wide screening for aberrant methylation, using a microarray combined with the methyltransferase inhibitor 5-aza-2'-deoxycytidine (5-aza-dCyd) in various types of tumors. We assessed the methylation status of TFPI2 and investigated its expression pattern in human primary gastric cancer (GC) tissues and in GC cell lines. Hypermethylation of the promoter CpG island, which was observed in more or less all of GC cell lines, was prevalent in a high proportion of primary GC tissues (15/18, or 83%), compared with noncancerous (4/18, or 22%) or normal (0/3, or 0%) stomach tissues, and expression of TFPI2 mRNA was reduced in 7 of the 17 primary GC tissues (41%). Moreover, immunohistochemical analyses showed decreased levels of TFPI-2 protein, compared with adjacent noncancerous tissues in 8 of the 20 primary GC tissues examined (40%). TFPI2 mRNA expression was restored in gene-silenced GC cells after treatment with 5-aza-dCyd. Aberrant methylation of TFPI2 promoter CpG island occurred not only in GC cells but also in primary GC tissues at a high frequency, suggesting that epigenetic silencing of TFPI2 may contribute to gastric carcinogenesis.
Collapse
Affiliation(s)
- Hisashi Takada
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kawaramachi Hirokoji, Kamigyo-ku, Kyoto, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Kuderer NM, Ortel TL, Francis CW. Impact of venous thromboembolism and anticoagulation on cancer and cancer survival. J Clin Oncol 2009; 27:4902-11. [PMID: 19738120 DOI: 10.1200/jco.2009.22.4584] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Changes in the hemostatic system and chronic hemostatic activation are frequently observed in patients with cancer, even in the absence of venous thromboembolism (VTE). VTE is a leading cause of death among patients with cancer and contributes to long-term mortality in patients with early as well as advanced-stage cancer. Mounting evidence suggests that components of the clotting cascade and associated vascular factors play an integral part in tumor progression, invasion, angiogenesis, and metastasis formation. Furthermore, there are intriguing in vitro and animal findings that anticoagulants, in particular the low molecular weight heparins (LMWHs), exert an antineoplastic effect through multiple mechanisms, including interference with tumor cell adhesion, invasion, metastasis formation, angiogenesis, and the immune system. Several relatively small randomized controlled clinical trials of anticoagulation as cancer therapy in patients without a VTE diagnosis have been completed. These comprise studies with LMWH, unfractionated heparin, and vitamin K antagonists, with overall encouraging but nonconclusive results and some limitations. Meta-analyses performed for the American Society of Clinical Oncology VTE Guidelines Committee and the Cochrane Collaboration suggest overall favorable effects of anticoagulation on survival of patients with cancer, mainly with LMWH. However, definitive clinical trials have been elusive and questions remain regarding the importance of tumor type and stage on treatment efficacy, the impact of fatal thromboembolic events, optimal anticoagulation therapy, and safety with differing chemotherapy regimens. Although the LMWHs and related agents hold promise for improving outcomes in patients with cancer, additional studies of their efficacy and safety in this setting are needed.
Collapse
Affiliation(s)
- Nicole M Kuderer
- Division of Hematology, Oncology and Cellular Therapy, Duke Comprehensive Cancer Center, Duke University Medical Center, DUMC 3841, Durham, NC 27710, USA.
| | | | | |
Collapse
|
37
|
Howell PM, Liu S, Ren S, Behlen C, Fodstad O, Riker AI. Epigenetics in human melanoma. Cancer Control 2009; 16:200-18. [PMID: 19556960 DOI: 10.1177/107327480901600302] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recent technological advances have allowed us to examine the human genome in greater detail than ever before. This has opened the door to an improved understanding of the gene expression patterns involved with cancer. METHODS A review of the literature was performed to determine the role of epigenetic modifications in human melanoma. We focused the search on histone deacetylation, methylation of gene promoter regions, demethylation of CpG islands, and the role of microRNA. We examined the relationship between human melanoma epigenetics and their importance in tumorigenesis, tumor progression, and inhibition of metastasis. The development and clinical application of select pharmacologic agents are also discussed. RESULTS We identified several articles that have extensively studied the role of epigenetics in melanoma, further elucidating the complex processes involved in gene regulation and expression. Several new agents directly affect epigenetic mechanisms in melanoma, with divergent affects on the metastatic potential of melanoma. CONCLUSIONS Epigenetic mechanisms have emerged as having a central role in gene regulation of human melanoma, including the identification of several putative tumor suppressor genes and oncogenes. Further research will focus on the development of novel therapeutics that will likely target and alter such epigenetic changes.
Collapse
Affiliation(s)
- Paul M Howell
- Basic and Translational Research Department, University of South Alabama, Mitchell Cancer Institute, Mobile, Alabama, USA
| | | | | | | | | | | |
Collapse
|
38
|
An SW, Kim NK, Chung HC. Genetic and epigenetic marker-based DNA test of stool is a promising approach for colorectal cancer screening. Yonsei Med J 2009; 50:331-4. [PMID: 19568592 PMCID: PMC2703753 DOI: 10.3349/ymj.2009.50.3.331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2009] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies and leading cause of cancer-related deaths in the world.1 However, it may be treated effectively by surgical removal of the cancerous tissue if detected at early stages. Conventional tools for screening CRC are either invasive or inaccurate. Therefore, there is an urgent need to develop a reliable screening tools for CRC to significantly reduce its morbidity. In this regard, a novel DNA markers-based detection in stool is emerging as a promising approach.
Collapse
Affiliation(s)
- Sung Whan An
- Cancer Metastasis Research Center, Yonsei University College of Medicine, Seoul, Korea.
| | | | | |
Collapse
|
39
|
Glöckner SC, Dhir M, Yi JM, McGarvey KE, Van Neste L, Louwagie J, Chan TA, Kleeberger W, de Bruïne AP, Smits KM, Khalid-de Bakker CAJ, Jonkers DMAE, Stockbrügger RW, Meijer GA, Oort FA, Iacobuzio-Donahue C, Bierau K, Herman JG, Baylin SB, Van Engeland M, Schuebel KE, Ahuja N. Methylation of TFPI2 in stool DNA: a potential novel biomarker for the detection of colorectal cancer. Cancer Res 2009; 69:4691-9. [PMID: 19435926 DOI: 10.1158/0008-5472.can-08-0142] [Citation(s) in RCA: 188] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have used a gene expression array-based strategy to identify the methylation of tissue factor pathway inhibitor 2 (TFPI2), a potential tumor suppressor gene, as a frequent event in human colorectal cancers (CRC). TFPI2 belongs to the recently described group of embryonic cell Polycomb group (PcG)-marked genes that may be predisposed to aberrant DNA methylation in early stages of colorectal carcinogenesis. Aberrant methylation of TFPI2 was detected in almost all CRC adenomas (97%, n = 56) and stages I to IV CRCs (99%, n = 115). We further explored the potential of TFPI2 as a biomarker for the early detection of CRC using stool DNA-based assays in patients with nonmetastatic CRC and average-risk noncancer controls who were candidates for screening. TFPI2 methylation was detected in stool DNA from stage I to III CRC patients with a sensitivity of 76% to 89% and a specificity of 79% to 93%. Detection of TFPI2 methylation in stool DNA may act as a useful adjunct to the noninvasive strategies for screening of CRCs in the future.
Collapse
Affiliation(s)
- Sabine C Glöckner
- Departments of Surgery, Oncology, and Pathology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Transient RNA silencing of tissue factor pathway inhibitor-2 modulates lung cancer cell invasion. Clin Exp Metastasis 2009; 26:457-67. [DOI: 10.1007/s10585-009-9245-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2008] [Accepted: 02/17/2009] [Indexed: 01/01/2023]
|
41
|
Tanemura A, Terando AM, Sim MS, van Hoesel AQ, de Maat MFG, Morton DL, Hoon DSB. CpG island methylator phenotype predicts progression of malignant melanoma. Clin Cancer Res 2009; 15:1801-7. [PMID: 19223509 DOI: 10.1158/1078-0432.ccr-08-1361] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE The CpG island methylator phenotype (CIMP) may be associated with development of malignancy through coordinated inactivation of tumor suppressor and tumor-related genes (TRG) and methylation of multiple noncoding, methylated-in-tumor (MINT) loci. These epigenetic changes create a distinct CIMP pattern that has been linked to recurrence and survival in gastrointestinal cancers. Because epigenetic inactivation of TRGs also has been shown in malignant melanoma, we hypothesized the existence of a clinically significant CIMP in cutaneous melanoma progression. EXPERIMENTAL DESIGN The methylation status of the CpG island promoter region of TRGs related to melanoma pathophysiology (WIF1, TFPI2, RASSF1A, RARbeta2, SOCS1, and GATA4) and a panel of MINT loci (MINT1, MINT2, MINT3, MINT12, MINT17, MINT25, and MINT31) in primary and metastatic tumors of different clinical stages (n=122) was assessed. RESULTS Here, we show an increase in hypermethylation of the TRGs WIF1, TFPI2, RASSF1A, and SOCS1 with advancing clinical tumor stage. Furthermore, we find a significant positive association between the methylation status of MINT17, MINT31, and TRGs. The methylation status of MINT31 is associated with disease outcome in stage III melanoma. CONCLUSIONS These findings show the significance of a CIMP pattern that is associated with advancing clinical stage of malignant melanoma. Future prospective large-scale studies may determine if CIMP-positive primary melanomas are at high risk of metastasis or recurrence.
Collapse
Affiliation(s)
- Atsushi Tanemura
- Department of Molecular Oncology, John Wayne Cancer Institute, Saint John's Health Center, Santa Monica, California 90404, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Ran Y, Pan J, Hu H, Zhou Z, Sun L, Peng L, Yu L, Sun L, Liu J, Yang Z. A Novel Role for Tissue Factor Pathway Inhibitor-2 in the Therapy of Human Esophageal Carcinoma. Hum Gene Ther 2009; 20:41-9. [PMID: 20377370 DOI: 10.1089/hum.2008.129] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Yuliang Ran
- The State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Jian Pan
- The State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Hai Hu
- The State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Zhuan Zhou
- The State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Lichao Sun
- The State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Liang Peng
- The State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Long Yu
- The State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Lixin Sun
- The State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Jun Liu
- The State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, People's Republic of China
| | - Zhihua Yang
- The State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, People's Republic of China
| |
Collapse
|
43
|
Bluff JE, Brown NJ, Reed MWR, Staton CA. Tissue factor, angiogenesis and tumour progression. Breast Cancer Res 2008; 10:204. [PMID: 18373885 PMCID: PMC2397518 DOI: 10.1186/bcr1871] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Tissue factor, the primary initiator of the coagulation cascade, maintains vascular integrity in response to injury. It is now recognised that, in addition to the role as a procoagulant activator, tissue factor participates in many tumour-related processes that contribute to malignant disease progression. The present review details the recent evidence supporting a role for tissue factor in tumour haemostasis, angiogenesis, metastasis and malignant cell survival. Furthermore, future research directions are discussed that may enhance our understanding of the role and regulation of this protein, which could ultimately lead to the innovative design and development of new anticancer therapies.
Collapse
Affiliation(s)
- Joanne E Bluff
- Microcirculation Research Group, Academic Unit of Surgical Oncology, School of Medicine and Biomedical Sciences, Beech Hill Road, Sheffield S10 2RX, UK.
| | | | | | | |
Collapse
|
44
|
Guo H, Lin Y, Zhang H, Liu J, Zhang N, Li Y, Kong D, Tang Q, Ma D. Tissue factor pathway inhibitor-2 was repressed by CpG hypermethylation through inhibition of KLF6 binding in highly invasive breast cancer cells. BMC Mol Biol 2007; 8:110. [PMID: 18053161 PMCID: PMC2233638 DOI: 10.1186/1471-2199-8-110] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Accepted: 12/03/2007] [Indexed: 12/16/2022] Open
Abstract
Background Tissue factor pathway inhibitor-2 (TFPI-2) is a matrix-associated Kunitz inhibitor that inhibits plasmin and trypsin-mediated activation of zymogen matrix metalloproteinases involved in tumor progression, invasion and metastasis. Here, we have investigated the mechanism of DNA methylation on the repression of TFPI-2 in breast cancer cell lines. Results We found that both protein and mRNA of TFPI-2 could not be detected in highly invasive breast cancer cell line MDA-MB-435. To further investigate the mechanism of TFPI-2 repression in breast cancer cells, 1.5 Kb TFPI-2 promoter was cloned, and several genetic variations were detected, but the promoter luciferase activities were not affected by the point mutation in the promoter region and the phenomena was further supported by deleted mutation. Scan mutation and informatics analysis identified a potential KLF6 binding site in TFPI-2 promoter. It was revealed, by bisulfite modified sequence, that the CpG island in TFPI-2 promoter region was hypermethylated in MDA-MB-435. Finally, using EMSA and ChIP assay, we demonstrated that the CpG methylation in the binding site of KLF-6 diminished the binding of KLF6 to TFPI-2 promoter. Conclusion In this study, we found that the CpG islands in TFPI-2 promoter was hypermethylated in highly invasive breast cancer cell line, and DNA methylation in the entire promoter region caused TFPI-2 repression by inducing inactive chromatin structure and decreasing KLF6 binding to its DNA binding sequence.
Collapse
Affiliation(s)
- Hongshen Guo
- Key Laboratory of Molecular Medicine, Ministry of Education, Yixueyuan Road 138#, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Liu S, Ren S, Howell P, Fodstad O, Riker AI. Identification of novel epigenetically modified genes in human melanoma via promoter methylation gene profiling. Pigment Cell Melanoma Res 2007; 21:545-58. [PMID: 18627528 DOI: 10.1111/j.1755-148x.2008.00484.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The inactivation of tumor-related genes through the aberrant methylation of promoter CpG islands is thought to contribute to tumor initiation and progression. We therefore investigated promoter methylation events involved in cutaneous melanoma by screening 30 genes of interest for evidence of promoter hypermethylation, examining 20 melanoma cell lines and 40 freshly procured melanoma samples. Utilizing quantitative methylation-specific PCR, we identified five genes (SOCS1, SOCS2, RAR-beta 2, TNFSF10C, and TNFSF10D) with hypermethylation frequencies ranging from 50% to 80% in melanoma cell lines as well as freshly procured tissue samples. Eighteen genes (LOX, RASSF1A, WFDC1, TM, APC, TFPI2, TNFSF10A, CDKN2A, MGMT, TIMP3, ASC, TPM1, IRF8, CIITA-PIV, CDH1, SYK, HOXB13, and DAPK1) were methylated at lower frequencies (2-30%). Two genes (CDKN1B and PTEN), previously reported as methylated in melanoma, and five other genes (RECK, IRF7, PAWR, TNFSF10B, and Rb) were not methylated in the samples screened here. Daughter melanoma cell lines showed identical methylation patterns when compared with original samples from which they were derived, as did synchronous metastatic lesions from the same patient. We identified four genes (TNFSF10C, TNFSF10D, LOX, and TPM1) that have never before been identified as hypermethylated in melanoma, with an overall methylation frequency of 60, 80, 50, and 10%, respectively, hypothesizing that these genes may play an important role in melanoma progression.
Collapse
Affiliation(s)
- Suhu Liu
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, USA
| | | | | | | | | |
Collapse
|