1
|
Suhardi VJ, Oktarina A, Hammad M, Niu Y, Li Q, Thomson A, Lopez J, McCormick J, Ayturk UM, Greenblatt MB, Ivashkiv LB, Bostrom MPG, Yang X. Prevention and treatment of peri-implant fibrosis by functionally inhibiting skeletal cells expressing the leptin receptor. Nat Biomed Eng 2024; 8:1285-1307. [PMID: 39085645 DOI: 10.1038/s41551-024-01238-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024]
Abstract
The cellular and molecular mediators of peri-implant fibrosis-a most common reason for implant failure and for surgical revision after the replacement of a prosthetic joint-remain unclear. Here we show that peri-implant fibrotic tissue in mice and humans is largely composed of a specific population of skeletal cells expressing the leptin receptor (LEPR) and that these cells are necessary and sufficient to generate and maintain peri-implant fibrotic tissue. In a mouse model of tibial implantation and osseointegration that mimics partial knee arthroplasty, genetic ablation of LEPR+ cells prevented peri-implant fibrosis and the implantation of LEPR+ cells from peri-implant fibrotic tissue was sufficient to induce fibrosis in secondary hosts. Conditional deletion of the adhesion G-protein-coupled receptor F5 (ADGRF5) in LEPR+ cells attenuated peri-implant fibrosis while augmenting peri-implant bone formation, and ADGRF5 inhibition by the intra-articular or systemic administration of neutralizing anti-ADGRF5 in the mice prevented and reversed peri-implant fibrosis. Pharmaceutical agents that inhibit the ADGRF5 pathway in LEPR+ cells may be used to prevent and treat peri-implant fibrosis.
Collapse
Affiliation(s)
- Vincentius Jeremy Suhardi
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | | | - Mohammed Hammad
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Yingzhen Niu
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Joint Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, P. R. China
| | - Qingdian Li
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Orthopedics, Guangdong Provincial People's Hospital, Southern Medical University, Guangzhou, P. R. China
| | - Andrew Thomson
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Juan Lopez
- Research Institute, Hospital for Special Surgery, New York, NY, USA
| | - Jason McCormick
- Flow Cytometry Core Facility, Weill Cornell Medicine, New York, NY, USA
| | - Ugur M Ayturk
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Matthew B Greenblatt
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Mathias P G Bostrom
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, NY, USA
- Research Institute, Hospital for Special Surgery, New York, NY, USA
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Xu Yang
- Research Institute, Hospital for Special Surgery, New York, NY, USA.
- Department of Orthopedic Surgery, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
2
|
Arroyo Villora S, Castellanos Silva P, Zenz T, Kwon JS, Schlaudraff N, Nitaj D, Meckbach C, Dammann R, Richter AM. Biomarker RIPK3 Is Silenced by Hypermethylation in Melanoma and Epigenetic Editing Reestablishes Its Tumor Suppressor Function. Genes (Basel) 2024; 15:175. [PMID: 38397165 PMCID: PMC10888250 DOI: 10.3390/genes15020175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/24/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
For several decades, cancers have demonstrably been one of the most frequent causes of death worldwide. In addition to genetic causes, cancer can also be caused by epigenetic gene modifications. Frequently, tumor suppressor genes are epigenetically inactivated due to hypermethylation of their CpG islands, actively contributing to tumorigenesis. Since CpG islands are usually localized near promoters, hypermethylation of the promoter can have a major impact on gene expression. In this study, the potential tumor suppressor gene Receptor Interacting Serine/Threonine Protein Kinase 3 (RIPK3) was examined for an epigenetic regulation and its gene inactivation in melanomas. A hypermethylation of the RIPK3 CpG island was detected by bisulfite pyrosequencing and was accompanied by a correlated loss of its expression. In addition, an increasing RIPK3 methylation rate was observed with increasing tumor stage of melanomas. For further epigenetic characterization of RIPK3, epigenetic modulation was performed using a modified CRISPR/dCas9 (CRISPRa activation) system targeting its DNA hypermethylation. We observed a reduced fitness of melanoma cells by (re-)expression and demethylation of the RIPK3 gene using the epigenetic editing-based method. The tumor suppressive function of RIPK3 was evident by phenotypic determination using fluorescence microscopy, flow cytometry and wound healing assay. Our data highlight the function of RIPK3 as an epigenetically regulated tumor suppressor in melanoma, allowing it to be classified as a biomarker.
Collapse
Affiliation(s)
- Sarah Arroyo Villora
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | | | - Tamara Zenz
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | - Ji Sun Kwon
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
- Department of Mathematics, Natural Sciences and Computer Science, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany
| | - Nico Schlaudraff
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | - Dafina Nitaj
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | - Cornelia Meckbach
- Department of Mathematics, Natural Sciences and Computer Science, University of Applied Sciences Mittelhessen, 35390 Giessen, Germany
| | - Reinhard Dammann
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| | - Antje M. Richter
- Institute for Genetics, Justus-Liebig-University Giessen, 35390 Giessen, Germany
| |
Collapse
|
3
|
Shechter O, Sausen DG, Gallo ES, Dahari H, Borenstein R. Epstein-Barr Virus (EBV) Epithelial Associated Malignancies: Exploring Pathologies and Current Treatments. Int J Mol Sci 2022; 23:14389. [PMID: 36430864 PMCID: PMC9699474 DOI: 10.3390/ijms232214389] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/06/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
Epstein-Barr virus (EBV) is one of eight known herpesviruses with the potential to infect humans. Globally, it is estimated that between 90-95% of the population has been infected with EBV. EBV is an oncogenic virus that has been strongly linked to various epithelial malignancies such as nasopharyngeal and gastric cancer. Recent evidence suggests a link between EBV and breast cancer. Additionally, there are other, rarer cancers with weaker evidence linking them to EBV. In this review, we discuss the currently known epithelial malignancies associated with EBV. Additionally, we discuss and establish which treatments and therapies are most recommended for each cancer associated with EBV.
Collapse
Affiliation(s)
- Oren Shechter
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Daniel G. Sausen
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA 23501, USA
| | - Elisa S. Gallo
- Tel-Aviv Sourasky Medical Center, Division of Dermatology, Tel-Aviv 6423906, Israel
| | - Harel Dahari
- The Program for Experimental and Theoretical Modeling, Division of Hepatology, Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| | - Ronen Borenstein
- The Program for Experimental and Theoretical Modeling, Division of Hepatology, Department of Medicine, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153, USA
| |
Collapse
|
4
|
Ntzifa A, Londra D, Rampias T, Kotsakis A, Georgoulias V, Lianidou E. DNA Methylation Analysis in Plasma Cell-Free DNA and Paired CTCs of NSCLC Patients before and after Osimertinib Treatment. Cancers (Basel) 2021; 13:cancers13235974. [PMID: 34885084 PMCID: PMC8656722 DOI: 10.3390/cancers13235974] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022] Open
Abstract
Osimertinib has been an effective second-line treatment in EGFR mutant NSCLC patients; however, resistance inevitably occurs. DNA methylation has been previously implicated in NSCLC progression and often in therapy resistance, however its distinct role in osimertinib resistance is not elucidated as yet. In the present study, we directly compared DNA methylation of nine selected genes (RASSF1A, RASSF10, APC, WIF-1, BRMS1, SLFN11, RARβ, SHISA3, and FOXA1) in plasma-cfDNA and paired CTCs of NSCLC patients who were longitudinally monitored during osimertinib treatment. Peripheral blood (PB) from 42 NSCLC patients was obtained at two time points: (a) baseline: before treatment with osimertinib and (b) at progression of disease (PD). DNA methylation of the selected genes was detected in plasma-cfDNA (n = 80) and in paired CTCs (n = 74). Direct comparison of DNA methylation of six genes between plasma-cfDNA and paired CTC samples (n = 70) revealed a low concordance, indicating that CTCs and cfDNA give complementary information. DNA methylation analysis of plasma-cfDNA and CTCs indicated that when at least one of these genes was methylated there was a statistically significant increase at PD compared to baseline (p = 0.031). For the first time, DNA methylation analysis in plasma-cfDNA and paired CTCs of NSCLC patients during osimertinib therapy indicated that DNA methylation of these genes could be a possible resistance mechanism.
Collapse
Affiliation(s)
- Aliki Ntzifa
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (A.N.); (D.L.)
| | - Dora Londra
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (A.N.); (D.L.)
| | - Theodoros Rampias
- Basic Research Center, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Athanasios Kotsakis
- Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece;
| | - Vassilis Georgoulias
- Department of Medical Oncology, Hellenic Oncology Research Group (HORG), 11471 Athens, Greece;
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (A.N.); (D.L.)
- Correspondence: ; Tel.: +30-210-727-4311
| |
Collapse
|
5
|
Yuan J, Ju Q, Zhu J, Jiang Y, Yang X, Liu X, Ma J, Sun C, Shi J. RASSF9 promotes NSCLC cell proliferation by activating the MEK/ERK axis. Cell Death Discov 2021; 7:199. [PMID: 34341331 PMCID: PMC8329231 DOI: 10.1038/s41420-021-00583-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 12/24/2022] Open
Abstract
The RAS-associated domain family 9 (RASSF9), a RAS-associated domain family gene, is expressed in a variety of tissues. However, its roles in tumorigenesis, particularly in non-small cell lung cancer (NSCLC), are still not understood well. In the present study, we aimed to examine the potential roles of RASSF9 in NSCLC and the underlying mechanisms. Our data showed that RASSF9 expression was upregulated in NSCLC tissues and cell lines. Increased expression of RASSF9 promotes NSCLC cell proliferation. On the contrary, knockdown of RASSF9 represses cell proliferation. Moreover, the effects of RASSF9 on NSCLC cell proliferation were further confirmed in vivo by using a subcutaneous tumor model. Mechanistically, pharmacological intervention studies revealed that the MEK/ERK axis is targeted by RASSF9 for transducing its regulatory roles on NSCLC cell proliferation. Collectively, our data indicate that RASSF9 plays a key role in tumorigenesis of NSCLC by stimulating tumor cell proliferation, which relies on activation of the MEK/ERK axis. Thus, RASSF9 might be a druggable target for developing novel agents for treating NSCLC.
Collapse
Affiliation(s)
- Jun Yuan
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Nantong Clinical Medical Research Center of Cardiothoracic Disease, and Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China
| | - Qianqian Ju
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Nantong Clinical Medical Research Center of Cardiothoracic Disease, and Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China.,Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Nantong University, Nantong, China.,NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jun Zhu
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Nantong Clinical Medical Research Center of Cardiothoracic Disease, and Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China
| | - Yun Jiang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Nantong Clinical Medical Research Center of Cardiothoracic Disease, and Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China
| | - Xuechao Yang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Nantong Clinical Medical Research Center of Cardiothoracic Disease, and Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaoyu Liu
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Nantong University, Nantong, China.,NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jinyu Ma
- Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Nantong University, Nantong, China.,NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Cheng Sun
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Nantong Clinical Medical Research Center of Cardiothoracic Disease, and Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China. .,Key Laboratory for Neuroregeneration of Jiangsu Province and Ministry of Education, Nantong University, Nantong, China. .,NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China.
| | - Jiahai Shi
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Nantong Clinical Medical Research Center of Cardiothoracic Disease, and Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China.
| |
Collapse
|
6
|
Dong Y, Tian J, Yan B, Lv K, Li J, Fu D. Liver-Metastasis-Related Genes are Potential Biomarkers for Predicting the Clinical Outcomes of Patients with Pancreatic Adenocarcinoma. Pathol Oncol Res 2021; 27:1609822. [PMID: 34290570 PMCID: PMC8286999 DOI: 10.3389/pore.2021.1609822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/21/2021] [Indexed: 12/26/2022]
Abstract
It is widely acknowledged that metastasis determines the prognosis of pancreatic adenocarcinoma (PAAD), and the liver is the most primary distant metastatic location of PAAD. It is worth exploring the value of liver-metastasis-related genetic prognostic signature (LM-PS) in predicting the clinical outcomes of PAAD patients post R0 resection. We collected 65 tumors and 165 normal pancreatic data from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression project (GTEx), respectively. Differentially expressed genes (DEGs) between primary tumor and normal pancreatic samples were intersected with DEGs between primary tumor samples with liver metastasis and those without new tumor events. The intersected 45 genes were input into univariate Cox regression analysis to identify the prognostic genes. Thirty-three prognostic liver-metastasis-related genes were identified and included in least absolute shrinkage and selection operator (LASSO) analysis to develop a seven-gene LM-PS, which included six risk genes (ANO1, FAM83A, GPR87, ITGB6, KLK10, and SERPINE1) and one protective gene (SMIM32). The PAAD patients were grouped into low- and high-risk groups based on the median value of risk scores. The LM-PS harbored an independent predictive ability to distinguish patients with a high-risk of death and liver metastasis after R0 resection. Moreover, a robust prognostic nomogram based on LM-PS, the number of positive lymph nodes, and histologic grade were established to predict the overall survival of PAAD patients. Besides, a transcription factor‐microRNA coregulatory network was constructed for the seven LM-PS genes, and the immune infiltration and genomic alterations were systematically explored in the TGCA-PAAD cohort.
Collapse
Affiliation(s)
- Yinlei Dong
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Junjie Tian
- Department of Urology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bingqian Yan
- Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Kun Lv
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Ji Li
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| | - Deliang Fu
- Department of Pancreatic Surgery, Pancreatic Disease Institute, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Hou Y, Li S, Du W, Li H, Wen R. The Tumor Suppressor Role of the Ras Association Domain Family 10. Anticancer Agents Med Chem 2021; 20:2207-2215. [PMID: 32664845 DOI: 10.2174/1871520620666200714141906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 04/30/2020] [Accepted: 05/17/2020] [Indexed: 11/22/2022]
Abstract
The Ras association domain family 10(RASSF10), a tumor suppressor gene, is located on human chromosome 11p15.2, which is one of the members homologous to other N-terminal RASSF families obtained through structural prediction. RASSF10 plays an important role in inhibiting proliferation, invasion, and migration, inducing apoptosis, making cancer cells sensitive to docetaxel, and capturing G2/M phase. Some studies have found that RASSF10 may inhibit the occurrence and development of tumors by regulating Wnt/β-catenin, P53, and MMP2. Methylation of tumor suppressor gene promoter is a key factor in the development and progression of many tumors. Various methylation detection methods confirmed that the methylation and downregulation of RASSF10 often occur in various tumors, such as gastric cancer, lung cancer, colon cancer, breast cancer, and leukemia. The status of RASSF10 methylation is positively correlated with tumor size, tumor type, and TNM stage. RASSF10 methylation can be used as a prognostic factor for overall survival and disease-free survival, and is also a sign of tumor diagnosis and sensitivity to docetaxel chemotherapy. In this review, we mainly elucidate the acknowledged structure and progress in the verified functions of RASSF10 and the probably relevant signaling pathways.
Collapse
Affiliation(s)
- Yulong Hou
- Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Shuofeng Li
- Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Wei Du
- Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Hailong Li
- Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| | - Rumin Wen
- Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
| |
Collapse
|
8
|
Zhao Z, Li M, Tan X, Xu D, Liu R. Methylation patterns partition pancreatic cancer into distinct prognostic subtypes. Future Oncol 2021; 17:2027-2039. [PMID: 33784823 DOI: 10.2217/fon-2020-0804] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In the initiation and progression of pancreatic cancer, DNA methylation plays a critical role. The present study attempts to explore specific prognosis subtypes based on DNA methylation data and develop an epigenetic signature to predict the overall survival (OS) of patients with pancreatic cancer.147 samples were included in the training cohort, whereas the validation cohort included 226 samples. The 298 OS-related methylation sites in the training cohort were selected for consensus clustering, and the authors identified three subtypes with a significant difference in prognosis. Cluster1 was associated with poor OS, low-grade disease and high lymph node involvement. In addition, we identified 33 specific methylation sites in Cluster1. Subsequently, we developed a robust qualitative signature consisting of 14 methylation sites to individually predict OS in the training cohort, and the predictive accuracy of this model was confirmed in the validation cohort. Functional enrichment analysis showed that the selected genes in the model were mainly enriched in known cancer-related pathways. Patients were divided into high- and low-risk groups by the model, and a significant difference in OS was observed between these groups. Classification based on the modeling of a specific DNA methylation site can reveal the heterogeneity of pancreatic cancer and provide guidance for clinicians in predicting the prognosis of pancreatic cancer and providing personalized treatment.
Collapse
Affiliation(s)
- Zhiming Zhao
- Department of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Mengyang Li
- Department of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Xianglong Tan
- Department of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Dabin Xu
- Department of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| | - Rong Liu
- Department of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Haidian District, Beijing, 100853, China
| |
Collapse
|
9
|
Gao Y, Fu Y, Wang J, Zheng X, Zhou J, Ma J. EBV as a high infection risk factor promotes RASSF10 methylation and induces cell proliferation in EBV-associated gastric cancer. Biochem Biophys Res Commun 2021; 547:1-8. [PMID: 33588233 DOI: 10.1016/j.bbrc.2021.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
Epstein-Barr virus (EBV) is the first identified human tumor-related DNA virus, and has a high infection among people worldwide. Recent studies have showed that nearly 10% of gastric cancers have shown EBV infection and this kind of gastric cancer has been identified as a new subtype: EBV associated Gastric cancer (EBVaGC). Furthermore, it has been reported that tumor related genes in the EBVaGC showed frequent methylation modifications compared to those in the EBV negative gastric cancer (EBVnGC). To fully understand the role of EBV in EBVaGC, we analyzed and found that 16.67% of gastric carcinoma samples showed positive EBER1 signals. Mechanically, EBV-encoded Latent membrane protein 1 (LMP1) inhibited the expression of RASSF10, and promoted tumorigenesis by recruiting DNMT1 and inducing the DNA methylation of RASSF10. Altogether, it allows us a better understanding of the possible mechanism of EBV-induced gene hypermethylation in gastric cancer genome. Targeting EBV-induced DNA methylation is a potential therapeutic modality of EBVaGC.
Collapse
Affiliation(s)
- Yingxue Gao
- Xiangya Hospital, Department of Pathology, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, 410008, China
| | - Yuxin Fu
- Xiangya Hospital, Department of Pathology, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, 410008, China
| | - Jia Wang
- Xiangya Hospital, Department of Pathology, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, 410008, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China
| | - Xiang Zheng
- Xiangya Hospital, Department of Pathology, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, 410008, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China
| | - Jianhua Zhou
- Xiangya Hospital, Department of Pathology, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, 410008, China.
| | - Jian Ma
- Xiangya Hospital, Department of Pathology, Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha, Hunan, 410008, China; Hunan Key Laboratory of Nonresolving Inflammation and Cancer, NHC Key Laboratory of Carcinogenesis, Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Changsha, Hunan, China.
| |
Collapse
|
10
|
Deutschmeyer VE, Richter AM. The ZAR1 protein in cancer; from epigenetic silencing to functional characterisation and epigenetic therapy of tumour suppressors. Biochim Biophys Acta Rev Cancer 2020; 1874:188417. [PMID: 32828887 DOI: 10.1016/j.bbcan.2020.188417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022]
Abstract
ZAR1, zygote arrest 1, is a zinc finger protein (C-terminus), which was initially identified in mouse oocytes. Later it was found that its expression is present in various human tissues e.g. lung and kidney. Interestingly, it was observed that in various tumour types the ZAR1 transcript is missing due to hypermethylation of its CpG island promoter, but not ZAR2. Since methylation of the ZAR1 promoter is described as a frequent event in tumourigenesis, ZAR1 could serve as a useful diagnostic marker in cancer screens. ZAR1 was described as a useful prognostic/diagnostic cancer marker for lung cancer, kidney cancer, melanoma and possibly liver carcinoma. Furthermore, ZAR1 was reactivated as a tumour suppressor by epigenetic therapy using CRISPR-dCas9 method. This method holds the potential to precisely target not only ZAR1 and reactivate tumour suppressors in a tailored cancer therapy. ZAR1 is highly conserved amongst vertebrates, especially its zinc finger, which is the relevant domain for its protein and RNA binding ability. ZAR1 is implicated in various cellular mechanisms including regulation of oocyte/embryo development, cell cycle control and mRNA binding, though little was known about the underlying mechanisms. ZAR1 was reported to regulate and activate translation through the binding to TCS translation control sequences in the 3'UTRs of its target mRNA the kinase WEE1. ZAR1 has a tumour suppressing function by inhibiting cell cycle progression. Here we review the current literature on ZAR1 focusing on structural, functional and epigenetic aspects. Characterising the cellular mechanisms that regulate the signalling pathways ZAR1 is involved in, could lead to a deeper understanding of tumour development and, furthermore, to new strategies in cancer treatment.
Collapse
Affiliation(s)
| | - Antje M Richter
- Institute for Genetics, University of Giessen, 35392 Giessen, Germany; Max-Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany.
| |
Collapse
|
11
|
Richter AM, Woods ML, Küster MM, Walesch SK, Braun T, Boettger T, Dammann RH. RASSF10 is frequently epigenetically inactivated in kidney cancer and its knockout promotes neoplasia in cancer prone mice. Oncogene 2020; 39:3114-3127. [PMID: 32047266 PMCID: PMC7142015 DOI: 10.1038/s41388-020-1195-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 01/21/2020] [Accepted: 01/27/2020] [Indexed: 12/22/2022]
Abstract
Kidney cancer incidences are rising globally, thereby fueling the demand for targeted therapies and precision medicine. In our previous work, we have identified and characterized the Ras-Association Domain Family encoding ten members that are often aberrantly expressed in human cancers. In this study, we created and analyzed the Rassf10 knockout mice. Here we show that Rassf10 haploinsufficiency promotes neoplasia formation in two established mouse cancer models (Rassf1A-/- and p53-/-). Haploinsufficient Rassf10 knockout mice were significantly prone to various diseases including lymphoma (Rassf1A-/- background) and thymoma (p53-/- background). Especially Rassf10-/- and p53-deficient mice exhibited threefold increased rates of kidney cysts compared with p53-/- controls. Moreover, we observed that in human kidney cancer, RASSF10 is frequently epigenetically inactivated by its CpG island promoter hypermethylation. Primary tumors of renal clear cell and papillary cell carcinoma confirmed that RASSF10 methylation is associated with decreased expression in comparison to normal kidney tissue. In independent data sets, we could validate that RASSF10 inactivation clinically correlated with decreased survival and with progressed disease state of kidney cancer patients and polycystic kidney size. Functionally, we revealed that the loss of Rassf10 was significantly associated with upregulation of KRAS signaling and MYC expression. In summary, we could show that Rassf10 functions as a haploinsufficient tumor suppressor. In combination with other markers, RASSF10 silencing can serve as diagnostic and prognostic cancer biomarker in kidney diseases.
Collapse
Affiliation(s)
- Antje M Richter
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany. .,Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.
| | - Michelle L Woods
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany
| | - Miriam M Küster
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany
| | - Sara K Walesch
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany
| | - Thomas Braun
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany.,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, 35392, Giessen, Germany
| | - Thomas Boettger
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Reinhard H Dammann
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany. .,German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, 35392, Giessen, Germany.
| |
Collapse
|
12
|
RASSF10 Is a TGFβ-Target That Regulates ASPP2 and E-Cadherin Expression and Acts as Tumor Suppressor That Is Epigenetically Downregulated in Advanced Cancer. Cancers (Basel) 2019; 11:cancers11121976. [PMID: 31817988 PMCID: PMC6966473 DOI: 10.3390/cancers11121976] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022] Open
Abstract
The Ras Association Domain Family (RASSF) encodes members of tumor suppressor genes which are frequently inactivated in human cancers. Here, the function and the regulation of RASSF10, that contains a RA (Ras-association) and two coiled domains, was investigated. We utilized mass spectrometry and immuno-precipitation to identify interaction partners of RASSF10. Additionally, we analyzed the up- and downstream pathways of RASSF10 that are involved in its tumor suppressive function. We report that RASSF10 binds ASPP1 (Apoptosis-stimulating protein of p53) and ASPP2 through its coiled-coils. Induction of RASSF10 leads to increased protein levels of ASPP2 and acts negatively on cell cycle progression. Interestingly, we found that RASSF10 is a target of the EMT (epithelial mesenchymal transition) driver TGFβ (Transforming growth factor beta) and that negatively associated genes of RASSF10 are significantly over-represented in an EMT gene set collection. We observed a positive correlation of RASSF10 expression and E-cadherin that prevents EMT. Depletion of RASSF10 by CRISPR/Cas9 technology induces the ability of lung cancer cells to proliferate and to invade an extracellular matrix after TGFβ treatment. Additionally, knockdown of RASSF10 or ASPP2 induced constitutive phosphorylation of SMAD2 (Smad family member 2). Moreover, we found that epigenetic reduction of RASSF10 levels correlates with tumor progression and poor survival in human cancers. Our study indicates that RASSF10 acts a TGFβ target gene and negatively regulates cell growth and invasion through ASPP2. This data suggests that epigenetic loss of RASSF10 contributes to tumorigenesis by promoting EMT induced by TGFβ.
Collapse
|
13
|
Deutschmeyer V, Breuer J, Walesch SK, Sokol AM, Graumann J, Bartkuhn M, Boettger T, Rossbach O, Richter AM. Epigenetic therapy of novel tumour suppressor ZAR1 and its cancer biomarker function. Clin Epigenetics 2019; 11:182. [PMID: 31801617 PMCID: PMC6894338 DOI: 10.1186/s13148-019-0774-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/29/2019] [Indexed: 12/27/2022] Open
Abstract
Background Cancer still is one of the leading causes of death and its death toll is predicted to rise further. We identified earlier the potential tumour suppressor zygote arrest 1 (ZAR1) to play a role in lung carcinogenesis through its epigenetic inactivation. Results We are the first to report that ZAR1 is epigenetically inactivated not only in lung cancer but also across cancer types, and ZAR1 methylation occurs across its complete CpG island. ZAR1 hypermethylation significantly correlates with its expression reduction in cancers. We are also the first to report that ZAR1 methylation and expression reduction are of clinical importance as a prognostic marker for lung cancer and kidney cancer. We further established that the carboxy (C)-terminally present zinc-finger of ZAR1 is relevant for its tumour suppression function and its protein partner binding associated with the mRNA/ribosomal network. Global gene expression profiling supported ZAR1's role in cell cycle arrest and p53 signalling pathway, and we could show that ZAR1 growth suppression was in part p53 dependent. Using the CRISPR-dCas9 tools, we were able to prove that epigenetic editing and reactivation of ZAR1 is possible in cancer cell lines. Conclusion ZAR1 is a novel cancer biomarker for lung and kidney, which is epigenetically silenced in various cancers by DNA hypermethylation. ZAR1 exerts its tumour suppressive function in part through p53 and through its zinc-finger domain. Epigenetic therapy can reactivate the ZAR1 tumour suppressor in cancer.
Collapse
Affiliation(s)
| | - Janina Breuer
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany.,Institute for Biochemistry, University of Giessen, 35392, Giessen, Germany
| | - Sara K Walesch
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany
| | - Anna M Sokol
- Scientific Service Group Biomolecular Mass Spectrometry, Max-Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany.,The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Max-Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Johannes Graumann
- Scientific Service Group Biomolecular Mass Spectrometry, Max-Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany.,The German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Max-Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Marek Bartkuhn
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany.,Institute for Bioinformatics, University of Giessen, 35392, Giessen, Germany
| | - Thomas Boettger
- Max-Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Oliver Rossbach
- Institute for Biochemistry, University of Giessen, 35392, Giessen, Germany
| | - Antje M Richter
- Institute for Genetics, University of Giessen, 35392, Giessen, Germany. .,Max-Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany.
| |
Collapse
|
14
|
PTBP1-mediated regulation of AXL mRNA stability plays a role in lung tumorigenesis. Sci Rep 2019; 9:16922. [PMID: 31729427 PMCID: PMC6858377 DOI: 10.1038/s41598-019-53097-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 10/23/2019] [Indexed: 11/09/2022] Open
Abstract
AXL is expressed in many types of cancer and promotes cancer cell survival, metastasis and drug resistance. Here, we focus on identifying modulators that regulate AXL at the mRNA level. We have previously observed that the AXL promoter activity is inversely correlated with the AXL expression levels, suggesting that post-transcriptional mechanisms exist that down-regulate the expression of AXL mRNA. Here we show that the RNA binding protein PTBP1 (polypyrimidine tract-binding protein) directly targets the 5′-UTR of AXL mRNA in vitro and in vivo. Moreover, we also demonstrate that PTBP1, but not PTBP2, inhibits the expression of AXL mRNA and the RNA recognition motif 1 (RRM1) of PTBP1 is crucial for this interaction. To clarify how PTBP1 regulates AXL expression at the mRNA level, we found that, while the transcription rate of AXL was not significantly different, PTBP1 decreased the stability of AXL mRNA. In addition, over-expression of AXL may counteract the PTBP1-mediated apoptosis. Knock-down of PTBP1 expression could enhance tumor growth in animal models. Finally, PTBP1 was found to be negatively correlated with AXL expression in lung tumor tissues in Oncomine datasets and in tissue micro-array (TMA) analysis. In conclusion, we have identified a molecular mechanism of AXL expression regulation by PTBP1 through controlling the AXL mRNA stability. These findings may represent new thoughts alternative to current approaches that directly inhibit AXL signaling and may eventually help to develop novel therapeutics to avoid cancer metastasis and drug resistance.
Collapse
|
15
|
Clinical and biological implications of Hippo pathway dysregulation in sarcomas. FORUM OF CLINICAL ONCOLOGY 2019. [DOI: 10.2478/fco-2018-0002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Sarcomas are mesenchymal malignant tumors with poor prognosis and limited treatment options. Hippo pathway is a recently discovered pathway normally involved in organ development and wound healing. Hippo signaling is often altered in solid tumors. The molecular elements of Hippo signaling include MST1/2 and LATS1/2 kinases which phosphorylate and regulate the activity of YAP and TAZ co-transcriptional activators. Hippo pathway cross-talks with several molecular pathways with known oncogenic function. In sarcomas Hippo signaling plays a pivotal role in tumorigenesis, evolution and resistance in chemotherapy regimens. Targeting Hippo pathway could potentially improve prognosis and outcome of sarcoma patients.
Collapse
|
16
|
Han X, Dong Q, Wu J, Luo Y, Rong X, Han Q, Zheng X, Wang E. RASSF10 suppresses lung cancer proliferation and invasion by decreasing the level of phosphorylated LRP6. Mol Carcinog 2019; 58:1168-1180. [DOI: 10.1002/mc.23000] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 02/14/2019] [Accepted: 02/18/2019] [Indexed: 01/30/2023]
Affiliation(s)
- Xu Han
- Department of Pathology; College of Basic Medical Sciences, China Medical University; Shenyang People's Republic of China
- Department of Pathology; First Hospital of China Medical University; Shenyang People's Republic of China
| | - Qianze Dong
- Department of Pathology; College of Basic Medical Sciences, China Medical University; Shenyang People's Republic of China
| | - Jingjing Wu
- Department of Pathology; College of Basic Medical Sciences, China Medical University; Shenyang People's Republic of China
| | - Yuan Luo
- Department of Pathology; College of Basic Medical Sciences, China Medical University; Shenyang People's Republic of China
| | - Xuezhu Rong
- Department of Pathology; College of Basic Medical Sciences, China Medical University; Shenyang People's Republic of China
| | - Qiang Han
- Department of Pathology; College of Basic Medical Sciences, China Medical University; Shenyang People's Republic of China
| | - Xiaoying Zheng
- Department of Pathology; College of Basic Medical Sciences, China Medical University; Shenyang People's Republic of China
| | - Enhua Wang
- Department of Pathology; College of Basic Medical Sciences, China Medical University; Shenyang People's Republic of China
- Department of Pathology; First Hospital of China Medical University; Shenyang People's Republic of China
| |
Collapse
|
17
|
Yang S, Hwang S, Jang J, Kim M, Gwak J, Jeong SM. PGC1α is required for the induction of contact inhibition by suppressing ROS. Biochem Biophys Res Commun 2018; 501:739-744. [DOI: 10.1016/j.bbrc.2018.05.059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 05/09/2018] [Indexed: 10/16/2022]
|
18
|
Abstract
Epigenetic deregulation is of importance in tumorigenesis. In particular CpG islands (CGI), are frequently hypermethylated. Here, genome-wide DNA-methylation profiles of 480,000 CpGs in lung cancer cells were generated. It was observed that intra- and intergenic CGI exhibited higher methylation compared to normal cells. The functional annotation of hypermethylated CGI revealed that the hypermethylation was associated with homeobox domain genes and targets marked by repressive histone modifications. The strongest methylation variation was observed in transitional areas of CGI, termed shores. 5'-shores of promoter-associated CGI in lung cancer cell lines were higher methylated than 3'-shores. Within two tandem-oriented genes, a significant hypermethylation of the downstream-located CGI promoters was revealed. Hypermethylation correlates with the length of the intergenic region between such tandem genes. As the RASSF1A tumor suppressor gene represents such a downstream tandem gene, its silencing was analyzed using an inducible system. It was determined that the induction of an upstream gene led to a repression of RASSF1A through a process involving histone deacetylases and CPSF1. A tumor-specific increase in expression of histone deacetylases and CPSF1 was detected in lung cancer. Our results suggest that the downstream gene could be susceptible to epigenetic silencing when organized in a tandem orientation.
Collapse
|
19
|
Richter AM, Kiehl S, Köger N, Breuer J, Stiewe T, Dammann RH. ZAR1 is a novel epigenetically inactivated tumour suppressor in lung cancer. Clin Epigenetics 2017; 9:60. [PMID: 28588743 PMCID: PMC5457737 DOI: 10.1186/s13148-017-0360-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 05/25/2017] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Lung cancer is the leading cause of cancer-related deaths with 1.8 million new cases each year and poor 5-year prognosis. Promoter hypermethylation of tumour suppressors leads to their inactivation and thereby can promote cancer development and progression. RESULTS In this study, we analysed ZAR1 (zygote arrest 1), which has been said to be a maternal-effect gene and its expression mostly limited to certain reproductive tissues. Our study shows that ZAR1 is expressed in normal lung but inactivated by promoter methylation in lung cancer. ZAR1 is hypermethylated in primary lung cancer samples (22% small cell lung carcinoma (SCLC) and 76% non-small cell lung carcinoma (NSCLC), p < 0.001) vs. normal control lung tissue (11%). In lung cancer cell lines, ZAR1 was significantly methylated in 75% of SCLC and 83% of NSCLC vs. normal tissue (p < 0.005/0.05). In matching tumours and control tissues, we observed that NSCLC primary tumour samples exhibited a tumour-specific promoter methylation of ZAR1 in comparison to the normal control lung tissue. Demethylation treatment of various lung cancer cell lines reversed ZAR1 promoter hypermethylation and subsequently re-established ZAR1 expression. In addition, we could show the growth inhibitory potential of ZAR1 in lung cancer cell lines and cancer cell lines. Exogenous expression of ZAR1 not only inhibited colony formation but also blocked cell cycle progression of cancer cell lines. CONCLUSIONS Our study shows for the first time the lung tumour-specific epigenetic inactivation of ZAR1 due to DNA methylation of its CpG island promoter. Furthermore, ZAR1 was characterised by the ability to block tumour growth through the inhibition of cell cycle progression in cancer cell lines. We propose that ZAR1 could serve as an epigenetically inactivated biomarker in lung cancer.
Collapse
Affiliation(s)
- Antje M. Richter
- Institute for Genetics, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Steffen Kiehl
- Institute for Genetics, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Nicole Köger
- Institute for Genetics, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Janina Breuer
- Institute for Genetics, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University Marburg, 35043 Marburg, Germany
- German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, 35392 Giessen, Germany
| | - Reinhard H. Dammann
- Institute for Genetics, Justus-Liebig-University Giessen, 35392 Giessen, Germany
- German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center, 35392 Giessen, Germany
| |
Collapse
|
20
|
Shimizu D, Inokawa Y, Sonohara F, Inaoka K, Nomoto S. Search for useful biomarkers in hepatocellular carcinoma, tumor factors and background liver factors. Oncol Rep 2017; 37:2527-2542. [DOI: 10.3892/or.2017.5541] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/09/2017] [Indexed: 11/06/2022] Open
|
21
|
Wang F, Li P, Feng Y, Hu YL, Liu YF, Guo YB, Jiang XL, Mao QS, Xue WJ. Low expression of RASSF10 is associated with poor survival in patients with colorectal cancer. Hum Pathol 2016; 62:108-114. [PMID: 28041974 DOI: 10.1016/j.humpath.2016.12.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/06/2016] [Accepted: 12/15/2016] [Indexed: 12/01/2022]
Abstract
The RASSF10 has been identified as a tumor suppressor in human colorectal cancer (CRC). However, the expression of RASSF10 in patients with CRC has not been evaluated for its potential use as a biomarker in the diagnosis and prognosis assessment of CRC. We analyzed the expression of RASSF10 mRNA (n=30) and protein (n=205) in CRC and matched noncancerous colon tissue samples to explore the relationships among RASSF10 expression, clinicopathological factors, and prognosis in patients with CRC. Our results showed that the expression of RASSF10 mRNA and protein in CRC-adjacent tissues was higher than that in CRC tissues. Low RASSF10 expression was associated with the T stage (P=.037, odds ratio, 0.664; 95% confidence interval, 0.452-0.975) and the N stage (P<.001, odds ratio, 0.318; 95% confidence interval, 0.184-0.549) of the tumors. In addition, univariate analysis revealed that patients with CRC with lower RASSF10 expression had poorer overall survival (OS; P<.001) and disease-free survival (DFS; P<.001). The 5-year OS and DFS rates were 48.2% and 28.3%, respectively, in patients with low RASSF10 expression and 82.2% and 62.6%, respectively, in patients with high RASSF10 expression. Multivariate Cox regression analysis revealed that the strongest predictors of OS and DFS were RASSF10 expression (P<.001 and P<.001, respectively), T stage (P=.003 and P=.009, respectively), and N stage (P=.005 and P=.026, respectively). These results demonstrate that low expression of RASSF10 in CRC tissues is significantly correlated with poor survival after curative resection and may serve as a useful biomarker predictive of CRC prognosis.
Collapse
Affiliation(s)
- Fei Wang
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China; Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Peng Li
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Ying Feng
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Yi-Lin Hu
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Yi-Fei Liu
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Yi-Bing Guo
- Department of Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China
| | - Xin-Lin Jiang
- Department of General Surgery, Hospital of Traditional Chinese Medicine of Nantong City, Nantong, Jiangsu 226001, China
| | - Qin-Sheng Mao
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China.
| | - Wan-Jiang Xue
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu 226001, China.
| |
Collapse
|
22
|
Wang F, Feng Y, Li P, Wang K, Feng L, Liu YF, Huang H, Guo YB, Mao QS, Xue WJ. RASSF10 is an epigenetically inactivated tumor suppressor and independent prognostic factor in hepatocellular carcinoma. Oncotarget 2016; 7:4279-97. [PMID: 26701853 PMCID: PMC4826205 DOI: 10.18632/oncotarget.6654] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 12/04/2015] [Indexed: 01/12/2023] Open
Abstract
Methylation of the Ras-association domain family 10 (RASSF10) promoter region correlates with clinicopathological characteristics and poor prognosis in several human cancers. Here, we examined RASSF10 expression in hepatocellular carcinoma (HCC) and its role in hepatocarcinogenesis. RASSF10 mRNA and protein levels were downregulated in both HCC cell lines and patient tissue samples. In patient tissues, low RASSF10 levels correlated with hepatocirrhosis, poor tumor differentiation, tumor thrombus and Barcelona Clinic Liver Cancer stage, and were indicative of increased tumor recurrence and reduced patient survival. Low RASSF10 expression was associated with promoter hypermethylation, which was in turn associated with polycyclic aromatic hydrocarbon and aflatoxin B1 exposure, but not DNA methyltransferase expression. Overexpression of RASSF10 in HCC cell lines suppressed cell growth and colony formation, and induced apoptosis by up- or down-regulating specific Bcl-2 family proteins. RASSF10 overexpression increased pro-apoptotic Bax and Bad levels, but decreased anti-apoptotic Bcl-2 and Bcl-xl expression. Overexpression also inhibited tumor formation in nude mice and reduced cell migration and invasion by inhibiting the epithelial-mesenchymal transition. RASSF10 knockdown promoted cell growth. Our results show that RASSF10 is frequently hypermethylated and down-regulated in HCC and can potentially serve as a useful biomarker predictive of HCC patient prognosis.
Collapse
Affiliation(s)
- Fei Wang
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Ying Feng
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Peng Li
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Kun Wang
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China.,Department of General Surgery, Changzhou Wujin People's Hospital, Changzhou, Jiangsu, China
| | - Liang Feng
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Yi-Fei Liu
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Hua Huang
- Department of Pathology, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Yi-Bing Guo
- Department of Surgical Comprehensive Laboratory, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Qin-Sheng Mao
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| | - Wan-Jiang Xue
- Department of General Surgery, Nantong University Affiliated Hospital, Nantong, Jiangsu, China
| |
Collapse
|
23
|
Candeias MM, Hagiwara M, Matsuda M. Cancer-specific mutations in p53 induce the translation of Δ160p53 promoting tumorigenesis. EMBO Rep 2016; 17:1542-1551. [PMID: 27702985 DOI: 10.15252/embr.201541956] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 09/06/2016] [Indexed: 12/29/2022] Open
Abstract
Wild-type p53 functions as a tumour suppressor while mutant p53 possesses oncogenic potential. Until now it remains unclear how a single mutation can transform p53 into a functionally distinct gene harbouring a new set of original cellular roles. Here we show that the most common p53 cancer mutants express a larger number and higher levels of shorter p53 protein isoforms that are translated from the mutated full-length p53 mRNA. Cells expressing mutant p53 exhibit "gain-of-function" cancer phenotypes, such as enhanced cell survival, proliferation, invasion and adhesion, altered mammary tissue architecture and invasive cell structures. Interestingly, Δ160p53-overexpressing cells behave in a similar manner. In contrast, an exogenous or endogenous mutant p53 that fails to express Δ160p53 due to specific mutations or antisense knock-down loses pro-oncogenic potential. Our data support a model in which "gain-of-function" phenotypes induced by p53 mutations depend on the shorter p53 isoforms. As a conserved wild-type isoform, Δ160p53 has evolved during millions of years. We thus provide a rational explanation for the origin of the tumour-promoting functions of p53 mutations.
Collapse
Affiliation(s)
- Marco M Candeias
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies Kyoto University, Kyoto, Japan .,Department of Anatomy and Developmental Biology, Graduate School of Medicine Kyoto University, Kyoto, Japan.,Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisboa, Portugal
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine Kyoto University, Kyoto, Japan.,Medical Research Support Center, Graduate School of Medicine Kyoto University, Kyoto, Japan
| | - Michiyuki Matsuda
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies Kyoto University, Kyoto, Japan.,Department of Pathology and Biology of Diseases, Graduate School of Medicine Kyoto University, Kyoto, Japan
| |
Collapse
|
24
|
Liu W, Wang J, Wang L, Qian C, Qian Y, Xuan H, Zhuo W, Li X, Yu J, Si J. Ras-association domain family 10 acts as a novel tumor suppressor through modulating MMP2 in hepatocarcinoma. Oncogenesis 2016; 5:e237. [PMID: 27348267 PMCID: PMC4945738 DOI: 10.1038/oncsis.2016.24] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 01/21/2016] [Accepted: 01/28/2016] [Indexed: 12/21/2022] Open
Abstract
Ras-Association Domain Family 10 (RASSF10) is the last identified member of the RASSF family. The functional characteristics of this new gene in human cancers remain largely unclear. Here, we examined RASSF10 for the biological functions and related molecular mechanisms in hepatocellular carcinoma (HCC). We found that RASSF10 is expressed in normal human liver tissue, but is silenced or down-regulated in 62.5% (5/8) of HCC cell lines. The mean expression level of RASSF10 was significantly lower in primary HCCs compared with their adjacent normal tissues (P<0.005, n=52). The promoter methylation contributes to the inactivation of RASSF10 as demonstrated by bisulfite genomic sequencing and demethylation treatment analyses. Transgenic expression of RASSF10 in silenced HCC cell lines suppressed cell viability, colony formation and inhibited tumor growth in nude mice (QGY7703, P<0.01; HepG2, P<0.05). Furthermore, RASSF10 was shown to induce the cell accumulation in G1 phase with the increase of p27, as well as the decrease of cyclinD1 and CDK2/CDK4. Over-expression of RASSF10 also inhibited HCC cells migration (P<0.01) or invasion (P<0.05). Adhesion genes array revealed that Matrix Metalloproteinase 2 (MMP2) was a downstream effector of RASSF10. RASSF10 acting as a tumor suppressor to inhibit HCC invasion partially mediated by Focal Adhesion Kinase or p38 MAPK to decrease the accumulation of MMP2. Our study suggests that RASSF10 acts as a tumor suppressor for HCC.
Collapse
Affiliation(s)
- W Liu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - J Wang
- Postgraduate at Institute of Gastroenterology, Zhejiang University; The First People's Hospital of Xiaoshan, Hangzhou, China
| | - L Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - C Qian
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Y Qian
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - H Xuan
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - W Zhuo
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - X Li
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - J Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - J Si
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Richter AM, Walesch SK, Dammann RH. Aberrant Promoter Methylation of the Tumour Suppressor RASSF10 and Its Growth Inhibitory Function in Breast Cancer. Cancers (Basel) 2016; 8:cancers8030026. [PMID: 26927176 PMCID: PMC4810110 DOI: 10.3390/cancers8030026] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/03/2016] [Accepted: 02/19/2016] [Indexed: 01/07/2023] Open
Abstract
Breast cancer is the most common cancer in women, with 1.7 million new cases each year. As early diagnosis and prognosis are crucial factors in cancer treatment, we investigated potential DNA methylation biomarkers of the tumour suppressor family Ras-association domain family (RASSF). Promoter hypermethylation of tumour suppressors leads to their inactivation and thereby promotes cancer development and progression. In this study we analysed the tumour suppressors RASSF1A and RASSF10. Our study shows that RASSF10 is expressed in normal breast but inactivated by methylation in breast cancer. We observed a significant inactivating promoter methylation of RASSF10 in primary breast tumours. RASSF10 is inactivated in 63% of primary breast cancer samples but only 4% of normal control breast tissue is methylated (p < 0.005). RASSF1A also shows high promoter methylation levels in breast cancer of 56% vs. 8% of normal tissue (p < 0.005). Interestingly more than 80% of breast cancer samples harboured a hypermethylation of RASSF10 and/or RASSF1A promoter. Matching samples exhibited a strong tumour specific promoter methylation of RASSF10 in comparison to the normal control breast tissue. Demethylation treatment of breast cancer cell lines MCF7 and T47D reversed RASSF10 promoter hypermethylation and re-established RASSF10 expression. In addition, we could show the growth inhibitory potential of RASSF10 in breast cancer cell lines MCF7 and T47D upon exogenous expression of RASSF10 by colony formation. We could further show, that RASSF10 induced apoptotic changes in MCF7 and T47D cells, which was verified by a significant increase in the apoptotic sub G1 fraction by 50% using flow cytometry for MCF7 cells. In summary, our study shows the breast tumour specific inactivation of RASSF10 and RASSF1A due to DNA methylation of their CpG island promoters. Furthermore RASSF10 was characterised by the ability to block growth of breast cancer cell lines by apoptosis induction.
Collapse
Affiliation(s)
- Antje M Richter
- Institute for Genetics, University of Giessen, Giessen 35392, Germany.
| | - Sara K Walesch
- Institute for Genetics, University of Giessen, Giessen 35392, Germany.
| | | |
Collapse
|
26
|
Guo J, Yang Y, Yang Y, Linghu E, Zhan Q, Brock MV, Herman JG, Zhang B, Guo M. RASSF10 suppresses colorectal cancer growth by activating P53 signaling and sensitizes colorectal cancer cell to docetaxel. Oncotarget 2016; 6:4202-13. [PMID: 25638156 PMCID: PMC4414183 DOI: 10.18632/oncotarget.2866] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Accepted: 12/07/2014] [Indexed: 01/10/2023] Open
Abstract
RASSF10 has previously been reported to be frequently methylated in a number of malignancies. To understand the importance of RASSF10 inactivation in colorectal carcinogenesis, eight colorectal cancer cell lines, 89 cases of primary colorectal cancer and 5 cases of normal colorectal mucosa were examined. Methylation specific PCR, western blot, siRNA, gene expression array and xenograft mice were employed. The expression of RASSF10 was regulated by promoter regional methylation in colorectal cancer cells. RASSF10 was methylated in 60.7% (54/89) of primary colorectal cancers and was positively associated with tumor stage (p < 0.05) and metastasis (p < 0.05). Restoration of RASSF10 led to inhibition of colorectal cancer cell proliferation in vitro and in vivo and increased apoptosis. Gene expression arrays discovered RASSF10 inhibition of MDM2 expression as a mediator of these effects, which was confirmed with RT-PCR and western blot. RASSF10 was shown to activate P53 signaling in RKO and HCT116 cells after UV exposure, and sensitized these cells to docetaxel. In conclusion, our study demonstrates RASSF10 is frequently methylated in human colorectal cancer leading to loss of expression. RASSF10 normally suppresses human colorectal cancer growth by activating P53 signaling in colorectal cancer, and restored expression sensitizes colorectal cancer to docetaxel.
Collapse
Affiliation(s)
- Jing Guo
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, Beijing 100853, P.R.China
| | - Yage Yang
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, Beijing 100853, P.R.China.,Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450003, P.R.China
| | - Yunsheng Yang
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, Beijing 100853, P.R.China
| | - Enqiang Linghu
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, Beijing 100853, P.R.China
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, P.R.China
| | - Malcolm V Brock
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland 21231, U.S.A
| | - James G Herman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland 21231, U.S.A
| | - Bingyong Zhang
- Department of Gastroenterology and Hepatology, Henan Provincial People's Hospital, Zhengzhou, Henan 450003, P.R.China
| | - Mingzhou Guo
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, Beijing 100853, P.R.China
| |
Collapse
|
27
|
Haag T, Richter AM, Schneider MB, Jiménez AP, Dammann RH. The dual specificity phosphatase 2 gene is hypermethylated in human cancer and regulated by epigenetic mechanisms. BMC Cancer 2016; 16:49. [PMID: 26833217 PMCID: PMC4736155 DOI: 10.1186/s12885-016-2087-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/27/2016] [Indexed: 12/31/2022] Open
Abstract
Background Dual specificity phosphatases are a class of tumor-associated proteins involved in the negative regulation of the MAP kinase pathway. Downregulation of the dual specificity phosphatase 2 (DUSP2) has been reported in cancer. Epigenetic silencing of tumor suppressor genes by abnormal promoter methylation is a frequent mechanism in oncogenesis. It has been shown that the epigenetic factor CTCF is involved in the regulation of tumor suppressor genes. Methods We analyzed the promoter hypermethylation of DUSP2 in human cancer, including primary Merkel cell carcinoma by bisulfite restriction analysis and pyrosequencing. Moreover we analyzed the impact of a DNA methyltransferase inhibitor (5-Aza-dC) and CTCF on the epigenetic regulation of DUSP2 by qRT-PCR, promoter assay, chromatin immuno-precipitation and methylation analysis. Results Here we report a significant tumor-specific hypermethylation of DUSP2 in primary Merkel cell carcinoma (p = 0.05). An increase in methylation of DUSP2 was also found in 17 out of 24 (71 %) cancer cell lines, including skin and lung cancer. Treatment of cancer cells with 5-Aza-dC induced DUSP2 expression by its promoter demethylation, Additionally we observed that CTCF induces DUSP2 expression in cell lines that exhibit silencing of DUSP2. This reactivation was accompanied by increased CTCF binding and demethylation of the DUSP2 promoter. Conclusions Our data show that aberrant epigenetic inactivation of DUSP2 occurs in carcinogenesis and that CTCF is involved in the epigenetic regulation of DUSP2 expression. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2087-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tanja Haag
- Institute for Genetics, Justus-Liebig-University, Heinrich-Buff-Ring 58-62, D-35392, Giessen, Germany.
| | - Antje M Richter
- Institute for Genetics, Justus-Liebig-University, Heinrich-Buff-Ring 58-62, D-35392, Giessen, Germany.
| | - Martin B Schneider
- Institute for Genetics, Justus-Liebig-University, Heinrich-Buff-Ring 58-62, D-35392, Giessen, Germany.
| | - Adriana P Jiménez
- Institute for Genetics, Justus-Liebig-University, Heinrich-Buff-Ring 58-62, D-35392, Giessen, Germany.
| | - Reinhard H Dammann
- Institute for Genetics, Justus-Liebig-University, Heinrich-Buff-Ring 58-62, D-35392, Giessen, Germany. .,Universities of Giessen and Marburg Lung Center, 35392, Giessen, Germany.
| |
Collapse
|
28
|
Deel MD, Li JJ, Crose LES, Linardic CM. A Review: Molecular Aberrations within Hippo Signaling in Bone and Soft-Tissue Sarcomas. Front Oncol 2015; 5:190. [PMID: 26389076 PMCID: PMC4557106 DOI: 10.3389/fonc.2015.00190] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 08/10/2015] [Indexed: 12/14/2022] Open
Abstract
The Hippo signaling pathway is an evolutionarily conserved developmental network vital for the regulation of organ size, tissue homeostasis, repair and regeneration, and cell fate. The Hippo pathway has also been shown to have tumor suppressor properties. Hippo transduction involves a series of kinases and scaffolding proteins that are intricately connected to proteins in developmental cascades and in the tissue microenvironment. This network governs the downstream Hippo transcriptional co-activators, YAP and TAZ, which bind to and activate the output of TEADs, as well as other transcription factors responsible for cellular proliferation, self-renewal, differentiation, and survival. Surprisingly, there are few oncogenic mutations within the core components of the Hippo pathway. Instead, dysregulated Hippo signaling is a versatile accomplice to commonly mutated cancer pathways. For example, YAP and TAZ can be activated by oncogenic signaling from other pathways, or serve as co-activators for classical oncogenes. Emerging evidence suggests that Hippo signaling couples cell density and cytoskeletal structural changes to morphogenic signals and conveys a mesenchymal phenotype. While much of Hippo biology has been described in epithelial cell systems, it is clear that dysregulated Hippo signaling also contributes to malignancies of mesenchymal origin. This review will summarize the known molecular alterations within the Hippo pathway in sarcomas and highlight how several pharmacologic compounds have shown activity in modulating Hippo components, providing proof-of-principle that Hippo signaling may be harnessed for therapeutic application in sarcomas.
Collapse
Affiliation(s)
- Michael D Deel
- Division of Hematology-Oncology, Department of Pediatrics, Duke University School of Medicine , Durham, NC , USA
| | - Jenny J Li
- Duke University School of Medicine , Durham, NC , USA
| | - Lisa E S Crose
- Division of Hematology-Oncology, Department of Pediatrics, Duke University School of Medicine , Durham, NC , USA
| | - Corinne M Linardic
- Division of Hematology-Oncology, Department of Pediatrics, Duke University School of Medicine , Durham, NC , USA ; Department of Pharmacology and Cancer Biology, Duke University School of Medicine , Durham, NC , USA
| |
Collapse
|
29
|
Ras association domain family member 10 suppresses gastric cancer growth by cooperating with GSTP1 to regulate JNK/c-Jun/AP-1 pathway. Oncogene 2015; 35:2453-64. [PMID: 26279301 DOI: 10.1038/onc.2015.300] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 06/16/2015] [Accepted: 06/28/2015] [Indexed: 11/08/2022]
Abstract
The Ras association domain family (RASSF) encodes several members with tumor-suppressive potentials. We aimed to investigate the biological function and clinical implication of RASSF10 in gastric cancer (GC). We found that RASSF10 was silenced in six of seven GC cell lines and in primary GC tissues, but was highly expressed in normal gastric tissues. The silence of RASSAF10 was mediated by promoter methylation as evaluated by bisulfite genomic sequencing. RASSF10 expression could be restored by demethylation treatment. A negative correlation between methylation and mRNA expression of RASSF10 was observed in 223 gastric samples of The Cancer Genome Atlas study (P<0.0001). Re-expression of RASSF10 in GC cell lines (AGS and MKN45) significantly suppressed cell viability, colony formation, migration and invasion, reduced cells in S phase, accumulated cells in G2 phase and induced cell apoptosis in vitro, and inhibited tumorigenicity in nude mice. These were confirmed by decreased expression of proliferation markers (proliferating cell nuclear antigen, p-CDC2 and p-CDC25) and increased apoptotic cascades (cleaved caspases-9, -8, -3 and cleaved poly (ADP-ribose) polymerase). Conversely, RASSF10 knockdown in normal gastric cell line yielded an opposing effect. Co-immunoprecipitation combined with mass spectrometry analyses were performed to reveal the downstream effectors of RASSF10. The result revealed that glutathione S-transferase Pi 1 (GSTP1) was a direct cooperator of RASSF10. The tumor-suppressive effect of RASSF10 was partially mediated by cooperating with GSTP1 to deregulate Jun N-terminal kinase (JNK)/c-Jun/AP-1 pathway. Importantly, RASSF10 methylation was detected in 56.6% (98/173) of primary GCs and is an independent risk factor for poor survival of GC patients (P=0.001). In conclusions, RASSF10 functions as a tumor suppressor by cooperating with GSTP1 to deregulate JNK/c-Jun/AP-1 pathway in GC. Promoter methylation of RASSF10 is associated with poor survival of GC patients.
Collapse
|
30
|
Claudin11 Promoter Hypermethylation Is Frequent in Malignant Melanoma of the Skin, but Uncommon in Nevus Cell Nevi. Cancers (Basel) 2015. [PMID: 26198249 PMCID: PMC4586767 DOI: 10.3390/cancers7030834] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Epigenetic inactivation of tumor-related genes is an important characteristic in the pathology of human cancers, including melanomagenesis. We analyzed the epigenetic inactivation of Claudin 11 (CLDN11) in malignant melanoma (MM) of the skin, including six melanoma cell lines, 39 primary melanoma, 41 metastases of MM and 52 nevus cell nevi (NCN). CLDN11 promoter hypermethylation was found in 19 out of 39 (49%) of the primary MM and in 21 out of 41 (51%) of the MM metastases, but only in eight out of 52 (15%) of NCN (p = 0.001 and p = 0.0003, respectively). Moreover, a significant increase in the methylation level of CLDN11 from primary melanomas to MM metastases was revealed (p = 0.003). Methylation of CLDN11 was significantly more frequent in skin metastases (79%) compared to brain metastases (31%; p = 0.007). CLDN11 methylation was also found in five out of six MM cell lines (83%) and its promoter hypermethylation correlated with a reduced expression. Treatment of MM cell lines with a DNA methylation inhibitor reactivated CLDN11 transcription by its promoter demethylation. In summary, CLDN11 proved to be an epigenetically inactivated tumor related gene in melanomagenesis, and analysis of CLDN11 methylation level represents a potential tool for assisting in the discrimination between malignant melanoma and nevus cell nevi.
Collapse
|
31
|
Jin Y, Cao B, Zhang M, Zhan Q, Herman JG, Yu M, Guo M. RASSF10 suppresses hepatocellular carcinoma growth by activating P53 signaling and methylation of RASSF10 is a docetaxel resistant marker. Genes Cancer 2015; 6:231-40. [PMID: 26124922 PMCID: PMC4482244 DOI: 10.18632/genesandcancer.67] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/28/2015] [Indexed: 01/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignances and the second leading cause of cancer related death worldwide. RASSF10 is located on chromosome 11p15.2, a region that shows frequent loss of heterozygosity (LOH) in several cancer types. Our previous study found that RASSF10 suppresses colorectal cancer growth by activating P53 signaling. To explore the epigenetic changes and the mechanism of RASSF10 in human HCC, 69 cases of primary HCC, twenty cases of normal liver tissue samples and 17 HCC cell lines were involved in this study. We found that RASSF10 was methylated in 82.6% (57/69) of human primary HCC and methylation of RASSF10 was significantly associated with tumor size (P < 0.05) and TNM stage (P < 0.05). The expression of RASSF10 was regulated by promoter region methylation. Restoration of RASSF10 expression suppressed cell proliferation, induced apoptosis and G2/M phase arrest, as well as sensitized cells to docetaxel and activated P53 signaling in HepG2 and QGY7703 cells. In conclusion, we demonstrated that RASSF10 is frequently methylated in human HCC and its methylation is a potential docetaxel resistant marker. Our data also indicate that RASSF10 suppresses human HCC growth by activating P53 signaling.
Collapse
Affiliation(s)
- Yongshuai Jin
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, Beijing, China ; Department of Interventional Radiology, Chinese PLA General Hospital, Beijing, China
| | - Baoping Cao
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, Beijing, China ; Medical College of NanKai University, Tianjin, China
| | - Meiying Zhang
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, Beijing, China ; Medical College of NanKai University, Tianjin, China
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, Cancer Institute and Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - James G Herman
- The Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Miao Yu
- Department of Interventional Radiology, Chinese PLA General Hospital, Beijing, China
| | - Mingzhou Guo
- Department of Gastroenterology & Hepatology, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
32
|
Thompson MJ, Rubbi L, Dawson DW, Donahue TR, Pellegrini M. Pancreatic cancer patient survival correlates with DNA methylation of pancreas development genes. PLoS One 2015; 10:e0128814. [PMID: 26039411 PMCID: PMC4454596 DOI: 10.1371/journal.pone.0128814] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 04/30/2015] [Indexed: 02/07/2023] Open
Abstract
DNA methylation is an epigenetic mark associated with regulation of transcription and genome structure. These markers have been investigated in a variety of cancer settings for their utility in differentiating normal tissue from tumor tissue. Here, we examine the direct correlation between DNA methylation and patient survival. We find that changes in the DNA methylation of key pancreatic developmental genes are strongly associated with patient survival.
Collapse
Affiliation(s)
- Michael J. Thompson
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, California, 90095, United States of America
| | - Liudmilla Rubbi
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, California, 90095, United States of America
| | - David W. Dawson
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, California, 90095, United States of America
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, 90095, United States of America
| | - Timothy R. Donahue
- Department of Surgery, University of California Los Angeles, Los Angeles, California, 90095, United States of America
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, 90095, United States of America
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, 90095, United States of America
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California Los Angeles, Los Angeles, California, 90095, United States of America
- * E-mail:
| |
Collapse
|
33
|
ABCB4 is frequently epigenetically silenced in human cancers and inhibits tumor growth. Sci Rep 2014; 4:6899. [PMID: 25367630 PMCID: PMC4219162 DOI: 10.1038/srep06899] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 09/18/2014] [Indexed: 12/11/2022] Open
Abstract
Epigenetic silencing through promoter hypermethylation is an important hallmark for the inactivation of tumor-related genes in carcinogenesis. Here we identified the ATP-binding cassette sub-family B member 4 (ABCB4) as a novel epigenetically silenced target gene. We investigated the epigenetic regulation of ABCB4 in 26 human lung, breast, skin, liver, head and neck cancer cells lines and in primary cancers by methylation and expression analysis. Hypermethylation of the ABCB4 CpG island promoter occurred in 16 out of 26 (62%) human cancer cell lines. Aberrant methylation of ABCB4 was also revealed in 39% of primary lung cancer and in 20% of head and neck cancer tissues. In 37% of primary lung cancer samples, ABCB4 expression was absent. For breast cancer a significant hypermethylation occurred in tumor tissues (41%) compared to matching normal samples (0%, p = 0.002). Silencing of ABCB4 was reversed by 5-aza-2'-deoxycytidine and zebularine treatments leading to its reexpression in cancer cells. Overexpression of ABCB4 significantly suppressed colony formation and proliferation of lung cancer cells. Hypermethylation of Abcb4 occurred also in murine cancer, but was not found in normal tissues. Our findings suggest that ABCB4 is a frequently silenced gene in different cancers and it may act tumor suppressivly in lung cancer.
Collapse
|
34
|
Haag T, Herkt CE, Walesch SK, Richter AM, Dammann RH. The apoptosis associated tyrosine kinase gene is frequently hypermethylated in human cancer and is regulated by epigenetic mechanisms. Genes Cancer 2014; 5:365-74. [PMID: 25352953 PMCID: PMC4209602 DOI: 10.18632/genesandcancer.28] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/18/2014] [Indexed: 11/25/2022] Open
Abstract
Epigenetic gene inactivation through promoter hypermethylation is an important aberration involved in the silencing of tumor-associated genes in cancer. Here we identified the apoptosis associated tyrosine kinase (AATK) as an epigenetically downregulated tumor related gene. We analyzed the epigenetic regulation of AATK in several human cancer cell lines and normal tissues by methylation and expression analysis. Hypermethylation of AATK was also analyzed in 25 primary lung tumors, 30 breast cancers and 24 matching breast tissues. In normal tissues the AATK CpG island promoter was unmethylated and AATK was expressed. Hypermethylation of AATK occurred frequently in 13 out of 14 (93%) human cancer cell lines. Methylation was reversed by 5-aza-2′-deoxycytidine treatment leading to re-expression of AATK in cancer cell lines. Aberrant methylation of AATK was also revealed in primary lung (40%) and breast (53%) cancers, but was found to be significantly less methylated in matching normal breast tissues (17%; p<0.01). In addition, we observed that AATK is epigenetically reactivated through the chromatin regulator CTCF. We further show that overexpression of Aatk significantly suppresses colony formation in cancer cell lines. Our findings suggest that the apoptosis associated tyrosine kinase is frequently inactivated in human cancers and acts as a tumor suppressive gene.
Collapse
Affiliation(s)
- Tanja Haag
- Institute for Genetics; Justus-Liebig-University; Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research; Giessen, Germany
| | - Christina E Herkt
- Institute for Genetics; Justus-Liebig-University; Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research; Giessen, Germany
| | - Sara K Walesch
- Institute for Genetics; Justus-Liebig-University; Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research; Giessen, Germany
| | - Antje M Richter
- Institute for Genetics; Justus-Liebig-University; Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research; Giessen, Germany
| | - Reinhard H Dammann
- Institute for Genetics; Justus-Liebig-University; Universities of Giessen and Marburg Lung Center, Member of the German Center for Lung Research; Giessen, Germany
| |
Collapse
|
35
|
Developmental pathways hijacked by osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:93-118. [PMID: 24924170 DOI: 10.1007/978-3-319-04843-7_5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cancer of any type often can be described by an arrest, alteration or disruption in the normal development of a tissue or organ, and understanding of the normal counterpart's development can aid in understanding the malignant state. This is certainly true for osteosarcoma and the normal developmental pathways that guide osteoblast development that are changed in the genesis of osteogenic sarcoma. A carefully regulated crescendo-decrescendo expression of RUNX2 accompanies the transition from mesenchymal stem cell to immature osteoblast to mature osteoblast. This pivotal role is controlled by several pathways, including bone morphogenic protein (BMP), Wnt/β-catenin, fibroblast growth factor (FGF), and protein kinase C (PKC). The HIPPO pathway and its downstream target YAP help to regulate proliferation of immature osteoblasts and their maturation into non-proliferating mature osteoblasts. This pathway also helps regulate expression of the mature osteoblast protein osteocalcin. YAP also regulates expression of MT1-MMP, a membrane-bound matrix metalloprotease responsible for remodeling the extracellular matrix surrounding the osteoblasts. YAP, in turn, can be regulated by the ERBB family protein Her-4. Osteosarcoma may be thought of as a cell held at the immature osteoblast stage, retaining some of the characteristics of that developmental stage. Disruptions of several of these pathways have been described in osteosarcoma, including BMP, Wnt/b-catenin, RUNX2, HIPPO/YAP, and Her-4. Further, PKC can be activated by several receptor tyrosine kinases implicated in osteosarcoma, including the ERBB family (EGFR, Her-2 and Her-4 in osteosarcoma), IGF1R, FGF, and others. Understanding these functions may aid in the understanding the mechanisms underpinning clinical observations in osteosarcoma, including both the lytic and blastic phenotypes of tumors, the invasiveness of the disease, and the tendency for treated tumors to ossify rather than shrink. Through a better understanding of the relationship between normal osteoblast development and osteosarcoma, we may gain insights into novel therapeutic avenues and improved outcomes.
Collapse
|
36
|
Volodko N, Gordon M, Salla M, Ghazaleh HA, Baksh S. RASSF tumor suppressor gene family: Biological functions and regulation. FEBS Lett 2014; 588:2671-84. [DOI: 10.1016/j.febslet.2014.02.041] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 02/25/2014] [Accepted: 02/25/2014] [Indexed: 01/22/2023]
|
37
|
Richter AM, Haag T, Walesch S, Herrmann-Trost P, Marsch WC, Kutzner H, Helmbold P, Dammann RH. Aberrant Promoter Hypermethylation of RASSF Family Members in Merkel Cell Carcinoma. Cancers (Basel) 2013; 5:1566-76. [PMID: 24252868 PMCID: PMC3875954 DOI: 10.3390/cancers5041566] [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: 09/11/2013] [Revised: 10/23/2013] [Accepted: 11/08/2013] [Indexed: 12/03/2022] Open
Abstract
Merkel cell carcinoma (MCC) is one of the most aggressive cancers of the skin. RASSFs are a family of tumor suppressors that are frequently inactivated by promoter hypermethylation in various cancers. We studied CpG island promoter hypermethylation in MCC of RASSF2, RASSF5A, RASSF5C and RASSF10 by combined bisulfite restriction analysis (COBRA) in MCC samples and control tissue. We found RASSF2 to be methylated in three out of 43 (7%), RASSF5A in 17 out of 39 (44%, but also 43% in normal tissue), RASSF5C in two out of 26 (8%) and RASSF10 in 19 out of 84 (23%) of the cancer samples. No correlation between the methylation status of the analyzed RASSFs or between RASSF methylation and MCC characteristics (primary versus metastatic, Merkel cell polyoma virus infection, age, sex) was found. Our results show that RASSF2, RASSF5C and RASSF10 are aberrantly hypermethylated in MCC to a varying degree and this might contribute to Merkel cell carcinogenesis.
Collapse
Affiliation(s)
- Antje M. Richter
- Institute for Genetics, University of Giessen, Giessen D-35392, Germany; E-Mails: (A.M.R.); (T.H.); (S.W.)
| | - Tanja Haag
- Institute for Genetics, University of Giessen, Giessen D-35392, Germany; E-Mails: (A.M.R.); (T.H.); (S.W.)
| | - Sara Walesch
- Institute for Genetics, University of Giessen, Giessen D-35392, Germany; E-Mails: (A.M.R.); (T.H.); (S.W.)
| | | | - Wolfgang C. Marsch
- Department of Dermatology, University of Halle, Halle D-06120, Germany; E-Mail:
| | | | - Peter Helmbold
- Department of Dermatology, University of Heidelberg, Heidelberg D-69120, Germany; E-Mail: Peter.
| | - Reinhard H. Dammann
- Institute for Genetics, University of Giessen, Giessen D-35392, Germany; E-Mails: (A.M.R.); (T.H.); (S.W.)
| |
Collapse
|
38
|
Chan JJ, Katan M. PLCɛ and the RASSF family in tumour suppression and other functions. Adv Biol Regul 2013; 53:258-279. [PMID: 23958207 DOI: 10.1016/j.jbior.2013.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 07/15/2013] [Indexed: 06/02/2023]
Abstract
Not all proteins implicated in direct binding to Ras appear to have a positive role in the generation and progression of tumours; examples include Phospholipase C epsilon (PLCɛ) and some members of the Ras-association domain family (RASSF). The RASSF family comprises of ten members, known as RASSF1 to RASSF10. PLCɛ and RASSF members carry a common Ras-association domain (RA) that can potentially bind Ras oncoproteins and mediate Ras-regulated functions. RASSF1 to RASSF6 also share a common SARAH domain that facilitates protein-protein interactions with other SARAH domain proteins. The majority of the family are frequently downregulated by epigenetic silencing in cancers. They are implicated in various important biological processes including apoptosis, microtubule stabilisation and cell cycle regulation. Recent studies have reinforced the tumour suppressive properties of the RASSF family, with new evidence of emerging pathways and novel functions that suggest a wider role for these proteins. This review will first describe an emerging role of PLCɛ in tumour suppression and then focus on and summarise the new findings on the RASSF family in the last five years to consolidate their well-established functions, and highlight the new regulatory roles of specific RASSF members.
Collapse
Affiliation(s)
- Jia Jia Chan
- Institute of Structural and Molecular Biology, Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | | |
Collapse
|