1
|
Shi CJ, Xue ZH, Zeng WQ, Deng LQ, Pang FX, Zhang FW, Fu WM, Zhang JF. LncRNA-NEF suppressed oxaliplatin resistance and epithelial-mesenchymal transition in colorectal cancer through epigenetically inactivating MEK/ERK signaling. Cancer Gene Ther 2023:10.1038/s41417-023-00595-1. [PMID: 36782047 DOI: 10.1038/s41417-023-00595-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 01/06/2023] [Accepted: 02/01/2023] [Indexed: 02/15/2023]
Abstract
A major cause of oxaliplatin chemoresistance in colorectal cancer (CRC) is acquired epithelial-mesenchymal transition (EMT) in cancer cells, making the cancer cells easy to metastasis and recurrence. LncRNA Neighboring Enhancer of FOXA2 (lncRNA-NEF) has been characterized as a tumor suppressor to mediate cancer metastasis in multiple cancer types. However, whether it mediated the drug resistance remains unknown. In the present study, an oxaliplatin-resistant CRC cell line (SW620R) was established and lncRNA-NEF was obviously down-regulated in this resistant cell line. The further loss and gain-of-function studies demonstrated that this lncRNA suppressed oxaliplatin resistance as well as EMT programme in vitro and inhibited metastasis in vivo. Mechanistically, lncRNA-NEF epigenetically promoted the expression of DOK1 (Downstream of Tyrosine kinase 1), a negative regulator of MEK/ERK signaling, by disrupting DNA methyltransferases (DNMTs)-mediated DNA methylation. DOK1, in turn, induced the inactivation of MEK/ERK signaling, forming the lncRNA-NEF/DOK1/MEK/ERK regulatory axis to mediate oxaliplatin resistance in CRC. Collectively, our work reveals the critical function of lncRNA-NEF in mediating the oxaliplatin chemotherapy resistance in CRC, and provides a promising therapeutic strategy for CRC patients with oxaliplatin resistance.
Collapse
Affiliation(s)
- Chuan-Jian Shi
- Cancer center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong, PR China.,Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Zhi-He Xue
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Wei-Qiang Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Li-Qiang Deng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China
| | - Feng-Xiang Pang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Feng-Wei Zhang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Wei-Ming Fu
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, PR China.
| | - Jin-Fang Zhang
- Cancer center, Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, 518000, Guangdong, PR China.
| |
Collapse
|
2
|
Boström M, Larsson E. Somatic mutation distribution across tumour cohorts provides a signal for positive selection in cancer. Nat Commun 2022; 13:7023. [PMID: 36396655 PMCID: PMC9671924 DOI: 10.1038/s41467-022-34746-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer gene discovery is reliant on distinguishing driver mutations from a multitude of passenger mutations in tumour genomes. While driver genes may be revealed based on excess mutation recurrence or clustering, there is a need for orthogonal principles. Here, we take advantage of the fact that non-cancer genes, containing only passenger mutations under neutral selection, exhibit a likelihood of mutagenesis in a given tumour determined by the tumour's mutational signature and burden. This relationship can be disrupted by positive selection, leading to a difference in the distribution of mutated cases across a cohort for driver and passenger genes. We apply this principle to detect cancer drivers independently of recurrence in large pan-cancer cohorts, and show that our method (SEISMIC) performs comparably to traditional approaches and can provide resistance to known confounding mutational phenomena. Being based on a different principle, the approach provides a much-needed complement to existing methods for detecting signals of selection.
Collapse
Affiliation(s)
- Martin Boström
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Erik Larsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, SE-405 30, Gothenburg, Sweden.
| |
Collapse
|
3
|
Guan Y, Li M, Qiu Z, Xu J, Zhang Y, Hu N, Zhang X, Guo W, Yuan J, Shi Q, Wang W. Comprehensive analysis of DOK family genes expression, immune characteristics, and drug sensitivity in human tumors. J Adv Res 2022; 36:73-87. [PMID: 35127166 PMCID: PMC8799871 DOI: 10.1016/j.jare.2021.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
The expression of DOK family genes is related to overall survival (OS), clinical stage, tumor mutation, methylation, CNV, and SNV. DOK family genes are significantly associated with poor prognosis of UVM. DOK1-DOK3 has obvious correlation with tumor immunity and tumor microenvironment. DOK family gene is significantly related to tumor stemness and drug sensitivity. The expression of DOK family genes is related to the activation of EMT and hormone ER pathways, and is related to the inhibition of DNA damage response, cell cycle, and hormone AR pathways. DOK1 and DOK3, DOK2 and DOK3 have the significant correlation.
Introduction Objectives Methods Results Conclusions
Collapse
|
4
|
Development and Validation of Tumor Immunogenicity Based Gene Signature for Skin Cancer Risk Stratification. Int J Mol Sci 2021; 22:ijms222112025. [PMID: 34769455 PMCID: PMC8584987 DOI: 10.3390/ijms222112025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Melanoma is one of the most aggressive types of skin cancer, with significant heterogeneity in overall survival. Currently, tumor-node-metastasis (TNM) staging is insufficient to provide accurate survival prediction and appropriate treatment decision making for several types of tumors, such as those in melanoma patients. Therefore, the identification of more reliable prognosis biomarkers is urgently essential. Recent studies have shown that low immune cells infiltration is significantly associated with unfavorable clinical outcome in melanoma patients. Here we constructed a prognostic-related gene signature for melanoma risk stratification by quantifying the levels of several cancer hallmarks and identify the Wnt/β-catenin activation pathway as a primary risk factor for low tumor immunity. A series of bioinformatics and statistical methods were combined and applied to construct a Wnt-immune-related prognosis gene signature. With this gene signature, we computed risk scores for individual patients that can predict overall survival. To evaluate the robustness of the result, we validated the signature in multiple independent GEO datasets. Finally, an overall survival-related nomogram was established based on the gene signature and clinicopathological features. The Wnt-immune-related prognostic risk score could better predict overall survival compared with standard clinicopathological features. Our results provide a comprehensive map of the oncogene-immune-related gene signature that can serve as valuable biomarkers for better clinical decision making.
Collapse
|
5
|
Al-Yozbaki M, Jabre I, Syed NH, Wilson CM. Targeting DNA methyltransferases in non-small-cell lung cancer. Semin Cancer Biol 2021; 83:77-87. [PMID: 33486076 DOI: 10.1016/j.semcancer.2021.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 01/14/2021] [Accepted: 01/17/2021] [Indexed: 12/30/2022]
Abstract
Despite the advances in treatment using chemotherapy or targeted therapies, due to static survival rates, non-small cell lung cancer (NSCLC) is the major cause of cancer-related deaths worldwide. Epigenetic-based therapies have been developed for NSCLC by targeting DNA methyltransferases (DNMTs) and histone-modifying enzymes. However, treatment using single epigenetic agents on solid tumours has been inadequate; whereas, treatment with a combination of DNMTs inhibitors with chemotherapy and immunotherapy has shown great promise. Dietary sources of phytochemicals could also inhibit DNMTs and cancer stem cells, representing a novel and promising way to prevent and treat cancer. Herein, we will discuss the different DNMTs, DNA methylation profiling in NSCLC as well as current demethylating agents in ongoing clinical trials. Therefore, providing a concise overview of future developments in the field of epigenetic therapy in NSCLC.
Collapse
Affiliation(s)
- Minnatallah Al-Yozbaki
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| | - Ibtissam Jabre
- Dept. of Microbial Sciences, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7XH, UK
| | - Naeem H Syed
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK
| | - Cornelia M Wilson
- Canterbury Christ Church University, School of Human and Life Sciences, Life Sciences Industry Liaison Lab, Sandwich, UK; University of Liverpool, Institute of Translation Medicine, Dept of Molecular & Clinical Cancer Medicine, UK.
| |
Collapse
|
6
|
Abstract
Breast cancer, which is the most common type of cancer among women, is a heterogenous disease. It results from progressive accumulation of genetic and epigenetic alterations in different genes. The Dok1 protein has been identified as the major substrate of protein tyrosine kinases in hematopoietic cells. It is considered as a tumor suppressor due to the reports which describe its inhibitory effect on major oncogenic signaling pathways such as Mek/Erk/PI3k/Akt and Wnt/β-catenin. In this study, we investigated the mutation frequency of the DOK1 gene in 118 breast tumors using Sanger sequencing and DOK1 mRNA expression level in 63 breast cancer samples using qRT-PCR methods. Although the mutation frequency was low DOK1 mRNA expression levels were significantly reduced (63.5%) in the tumors compared to adjacent non-cancerous tissue. We also correlated expression changes with clinicopathological characteristics. Low mRNA levels correlated with age (p = 0.01) and c-erbB-2 (p = 0.05). In most of the previous reports, down-regulation of DOK1 mRNA expression has been associated with promoter methylation. We identified four different coding sequence alterations in 5.1% (6/118) of the tumor samples. However, all of these alterations were located in the functional domains of the protein. Therefore, these mutations may affect the function and/or cellular localization of the protein and contribute to cancer progression by this way. In conclusion our data indicate that DOK1 acts as a tumor suppressor in breast cancer and association of Dok1 with the c-erbB-2 mediated mechanism of action in breast cancer needs to be investigated.
Collapse
|
7
|
Cheng YY, Rath EM, Linton A, Yuen ML, Takahashi K, Lee K. The Current Understanding Of Asbestos-Induced Epigenetic Changes Associated With Lung Cancer. LUNG CANCER (AUCKLAND, N.Z.) 2020; 11:1-11. [PMID: 32021524 PMCID: PMC6955579 DOI: 10.2147/lctt.s186843] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 11/08/2019] [Indexed: 12/19/2022]
Abstract
Asbestos is a naturally occurring mineral consisting of extremely fine fibres that can become trapped in the lungs after inhalation. Occupational and environmental exposures to asbestos are linked to development of lung cancer and malignant mesothelioma, a cancer of the lining surrounding the lung. This review discusses the factors that are making asbestos-induced lung cancer a continuing problem, including the extensive historic use of asbestos and decades long latency between exposure and disease development. Genomic mutations of DNA nucleotides and gene rearrangements driving lung cancer are well-studied, with biomarkers and targeted therapies already in clinical use for some of these mutations. The genes involved in these mutation biomarkers and targeted therapies are also involved in epigenetic mechanisms and are discussed in this review as it is hoped that identification of epigenetic aberrations in these genes will enable the same gene biomarkers and targeted therapies to be used. Currently, understanding of how asbestos fibres trapped in the lungs leads to epigenetic changes and lung cancer is incomplete. It has been shown that oxidoreduction reactions on fibre surfaces generate reactive oxygen species (ROS) which in turn damage DNA, leading to genetic and epigenetic alterations that reduce the activity of tumour suppressor genes. Epigenetic DNA methylation changes associated with lung cancer are summarised in this review, and some of these changes will be due to asbestos exposure. So far, little research has been carried out to separate the asbestos driven epigenetic changes from those due to non-asbestos causes of lung cancer. Asbestos-associated lung cancers exhibit less methylation variability than lung cancers in general, and in a large proportion of samples variability has been found to be restricted to promoter regions. Epigenetic aberrations in cancer are proving to be promising biomarkers for diagnosing cancers. It is hoped that further understanding of epigenetic changes in lung cancer can result in useful asbestos-associated lung cancer biomarkers to guide treatment. Research is ongoing into the detection of lung cancer epigenetic alterations using non-invasive samples of blood and sputum. These efforts hold the promise of non-invasive cancer diagnosis in the future. Efforts to reverse epigenetic aberrations in lung cancer by epigenetic therapies are ongoing but have not yet yielded success.
Collapse
Affiliation(s)
- Yuen Yee Cheng
- Asbestos Disease Research Institute, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Emma M Rath
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Anthony Linton
- Asbestos Disease Research Institute, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| | - Man Lee Yuen
- Asbestos Disease Research Institute, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Ken Takahashi
- Asbestos Disease Research Institute, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Kenneth Lee
- Asbestos Disease Research Institute, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- Concord Repatriation General Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
8
|
Liljedahl ER, Wahlberg K, Lidén C, Albin M, Broberg K. Genetic variants of filaggrin are associated with occupational dermal exposure and blood DNA alterations in hairdressers. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 653:45-54. [PMID: 30399560 DOI: 10.1016/j.scitotenv.2018.10.328] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/11/2018] [Accepted: 10/24/2018] [Indexed: 06/08/2023]
Abstract
Hairdressers are exposed to high levels of chemicals, including possible carcinogens. For dermal exposure, the skin protects against the uptake of chemicals and the protein filaggrin (encoded by FLG) has a key role in skin barrier function. This study investigated if variants of FLG previously linked to impaired skin barrier function, i.e. null mutations and copy number variation (CNV) alleles (CNV10), are associated with cancer-related DNA changes. Blood and questionnaire data were collected from hairdressers (n = 295) and controls (n = 92). Exposure to aromatic amines was measured as hemoglobin adducts by gas chromatography tandem mass spectrometry. DNA from peripheral blood was used to test for FLG null mutations and CNV (10, 11, or 12 repeats), telomere length, and methylation of selected cancer-related genes. Hairdressers had a lower frequency of FLG null mutations (4.1 vs. 7.6%, P = 0.18) and CNV10 (43.2 vs. 56%, P = 0.0032) than controls. In hairdressers, CNV10 carriers had a decreased risk of high ortho-toluidine adducts in blood compared with non-carriers (odds ratio, OR = 0.49, 95% CI = 0.30-0.81). Further, telomere length was shorter for carriers of any FLG null allele (β = -0.18, 95% CI = -0.31 to -0.044) and CNV10 carriers (β = -0.054, 95% CI = -0.11 to -0.00051, linear regression adjusted for age, passive smoking, residence, and education) compared to non-carriers. Carriers of any FLG null allele showed higher methylation of the cyclin-dependent kinase inhibitor 2A gene CDKN2A (OR = 6.26, CI = 1.13-34.7), but not of the other genes analyzed. These associations were not found among the controls. Our study showed that the frequency of FLG CNV10 was lower among hairdressers than controls, which may indicate a healthy worker selection. Moreover, FLG null and CNV10 were associated with cancer-related DNA changes in hairdressers, which may influence their risk of cancer.
Collapse
Affiliation(s)
- Emelie Rietz Liljedahl
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Karin Wahlberg
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Carola Lidén
- Unit of Work Environment Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maria Albin
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden; Unit of Occupational Medicine, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Karin Broberg
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden; Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
9
|
He P, Xu Z, Zhou J, Li X, Zhang W, Wu D, Zhang Z, Lian X, Yao X, Deng Z, Lin J, Qian J. Methylation‐associated
DOK1
and
DOK2
down‐regulation: Potential biomarkers for predicting adverse prognosis in acute myeloid leukemia. J Cell Physiol 2018; 233:6604-6614. [PMID: 29150948 DOI: 10.1002/jcp.26271] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/25/2017] [Accepted: 11/06/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Pin‐Fang He
- Laboratory CenterAffiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuP.R. China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang CityZhenjiangJiangsuP.R. China
| | - Zi‐Jun Xu
- Laboratory CenterAffiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuP.R. China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang CityZhenjiangJiangsuP.R. China
| | - Jing‐Dong Zhou
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang CityZhenjiangJiangsuP.R. China
- Department of HematologyAffiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuP.R. China
| | - Xi‐Xi Li
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang CityZhenjiangJiangsuP.R. China
- Department of HematologyAffiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuP.R. China
| | - Wei Zhang
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang CityZhenjiangJiangsuP.R. China
- Department of HematologyAffiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuP.R. China
| | - De‐Hong Wu
- Department of HematologyThe Third People's Hospital of KunShan CityKunshanJiangsuP.R. China
| | - Zhi‐Hui Zhang
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang CityZhenjiangJiangsuP.R. China
- Department of HematologyAffiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuP.R. China
| | - Xin‐Yue Lian
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang CityZhenjiangJiangsuP.R. China
- Department of HematologyAffiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuP.R. China
| | - Xin‐Yu Yao
- School of medicineJiangsu UniversityZhenjiangJiangsuP.R. China
| | - Zhao‐Qun Deng
- Laboratory CenterAffiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuP.R. China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang CityZhenjiangJiangsuP.R. China
| | - Jiang Lin
- Laboratory CenterAffiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuP.R. China
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang CityZhenjiangJiangsuP.R. China
| | - Jun Qian
- The Key Lab of Precision Diagnosis and Treatment of Zhenjiang CityZhenjiangJiangsuP.R. China
- Department of HematologyAffiliated People's Hospital of Jiangsu UniversityZhenjiangJiangsuP.R. China
| |
Collapse
|
10
|
DOK1/PPARgamma pathway mediates anti-tumor ability of all-trans retinoic acid in breast cancer MCF-7 cells. Biochem Biophys Res Commun 2017; 487:189-193. [DOI: 10.1016/j.bbrc.2017.04.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 04/05/2017] [Indexed: 01/15/2023]
|
11
|
Falco M, Palma G, Rea D, De Biase D, Scala S, D'Aiuto M, Facchini G, Perdonà S, Barbieri A, Arra C. Tumour biomarkers: homeostasis as a novel prognostic indicator. Open Biol 2016; 6:160254. [PMID: 27927793 PMCID: PMC5204124 DOI: 10.1098/rsob.160254] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/10/2016] [Indexed: 12/15/2022] Open
Abstract
The term 'personalized medicine' refers to a medical procedure that consists in the grouping of patients based on their predicted individual response to therapy or risk of disease. In oncologic patients, a 'tailored' therapeutic approach may potentially improve their survival and well-being by not only reducing the tumour, but also enhancing therapeutic response and minimizing the adverse effects. Diagnostic tests are often used to select appropriate and optimal therapies that rely both on patient genome and other molecular/cellular analysis. Several studies have shown that lifestyle and environmental factors can influence the epigenome and that epigenetic events may be involved in carcinogenesis. Thus, in addition to traditional biomarkers, epigenetic factors are raising considerable interest, because they could potentially be used as an excellent tool for cancer diagnosis and prognosis. In this review, we summarize the role of conventional cancer genetic biomarkers and their association with epigenomics. Furthermore, we will focus on the so-called 'homeostatic biomarkers' that result from the physiological response to cancer, emphasizing the concept that an altered 'new' homeostasis influence not only tumour environment, but also the whole organism.
Collapse
Affiliation(s)
- Michela Falco
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Giuseppe Palma
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Domenica Rea
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Davide De Biase
- Department of Veterinary Medicine and Animal Production, University of Naples 'Federico II', Via Delpino 1, 80137 Naples, Italy
| | - Stefania Scala
- Molecular lmmunology and Immuneregulation, Istituto Nazionale per lo Studio e la Cura dei Tumori, IRCCS Naples 'Fondazione G. Pascale', Naples, italy, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Massimiliano D'Aiuto
- Division of Breast Surgery, Department of Breast Disease, National Cancer Institute, IRCCS, 'Fondazione Pascale', Naples, Italy
| | - Gaetano Facchini
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, , Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione G. Pascale', IRCCS, 80131 Naples, Italy
| | - Sisto Perdonà
- Department of Urology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione G. Pascale', IRCCS, 80131 Naples, Italy
| | - Antonio Barbieri
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| | - Claudio Arra
- Struttura Semplice Dipartimentale Sperimentazione Animale, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Via Mariano Semmola, 80131 Naples, Italy
| |
Collapse
|
12
|
Spatial intratumoral heterogeneity and temporal clonal evolution in esophageal squamous cell carcinoma. Nat Genet 2016; 48:1500-1507. [PMID: 27749841 PMCID: PMC5127772 DOI: 10.1038/ng.3683] [Citation(s) in RCA: 176] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 08/31/2016] [Indexed: 12/14/2022]
Abstract
Esophageal squamous cell carcinoma (ESCC) is among the most common malignancies, but little is known about its spatial intratumor heterogeneity (ITH) and temporal clonal evolutionary processes. To address this, we performed multiregion whole-exome sequencing on 51 tumor regions from 13 ESCCs, and multiregion global methylation profiling on three of these 13 cases. We found an average of 35.8% heterogeneous somatic mutations with strong evidence of ITH. Half of driver mutations located on the branches targeted oncogenes, including PIK3CA, NFE2L2, MTOR, etc. By contrast, the majority of truncal and clonal driver mutations occurred in tumor suppressor genes, including TP53, KMT2D, ZNF750, etc. Interestingly, the phyloepigenetic trees robustly recapitulated the topologic structures of the phylogenetic ones, indicating the possible relationship between genetic and epigenetic alterations. Our integrated investigations of the spatial ITH and clonal evolution provide an important molecular foundation for enhanced understanding of the tumorigenesis and progression of ESCC.
Collapse
|
13
|
Parroche P, Roblot G, Le Calvez-Kelm F, Tout I, Marotel M, Malfroy M, Durand G, McKay J, Ainouze M, Carreira C, Allatif O, Traverse-Glehen A, Mendiola M, Pozo-Kreilinger JJ, Caux C, Tommasino M, Goutagny N, Hasan UA. TLR9 re-expression in cancer cells extends the S-phase and stabilizes p16(INK4a) protein expression. Oncogenesis 2016; 5:e244. [PMID: 27454079 PMCID: PMC4972902 DOI: 10.1038/oncsis.2016.49] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/12/2016] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptor 9 (TLR9) recognizes bacterial, viral or cell damage-associated DNA, which initiates innate immune responses. We have previously shown that TLR9 expression is downregulated in several viral induced cancers including HPV16-induced cervical neoplasia. Findings supported that downregulation of TLR9 expression is involved in loss of anti-viral innate immunity allowing an efficient viral replication. Here we investigated the role of TLR9 in altering the growth of transformed epithelial cells. Re-introducing TLR9 under the control of an exogenous promoter in cervical or head and neck cancer patient-derived cells reduced cell proliferation, colony formation and prevented independent growth of cells under soft agar. Neither TLR3, 7, nor the TLR adapter protein MyD88 expression had any effect on cell proliferation, indicating that TLR9 has a unique role in controlling cell growth. The reduction of cell growth was not due to apoptosis or necrosis, yet we observed that cells expressing TLR9 were slower in entering the S-phase of the cell cycle. Microarray-based gene expression profiling analysis highlighted a strong interferon (IFN) signature in TLR9-expressing head and neck cancer cells, with an increase in IFN-type I and IL-29 expression (IFN-type III), yet neither IFN-type I nor IL-29 production was responsible for the block in cell growth. We observed that the protein half-life of p16(INK4a) was increased in TLR9-expressing cells. Taken together, these data show for the first time that TLR9 affects the cell cycle by regulating p16(INK4a) post-translational modifications and highlights the role of TLR9 in the events that lead to carcinogenesis.
Collapse
Affiliation(s)
- P Parroche
- CIRI, INSERM U1111, Ecole Normale Supérieure, Université de Lyon, Lyon, France
| | - G Roblot
- CIRI, INSERM U1111, Ecole Normale Supérieure, Université de Lyon, Lyon, France
| | - F Le Calvez-Kelm
- IARC-International Agency for Research on Cancer 150 Cours Albert Thomas, Lyon, France
| | - I Tout
- CIRI, INSERM U1111, Ecole Normale Supérieure, Université de Lyon, Lyon, France
| | - M Marotel
- CIRI, INSERM U1111, Ecole Normale Supérieure, Université de Lyon, Lyon, France
| | - M Malfroy
- CRCL, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard, Lyon France
| | - G Durand
- IARC-International Agency for Research on Cancer 150 Cours Albert Thomas, Lyon, France
| | - J McKay
- IARC-International Agency for Research on Cancer 150 Cours Albert Thomas, Lyon, France
| | - M Ainouze
- CIRI, INSERM U1111, Ecole Normale Supérieure, Université de Lyon, Lyon, France
| | - C Carreira
- IARC-International Agency for Research on Cancer 150 Cours Albert Thomas, Lyon, France
| | - O Allatif
- CIRI, INSERM U1111, Ecole Normale Supérieure, Université de Lyon, Lyon, France
| | | | - M Mendiola
- Molecular Pathology and Therapeutic Targets Group, Research Insitute (IdiPAZ), La Paz University Hospital, Madrid, Spain and Molecular Pathology Diagnostics Unit, Institute of Medical and Molecular Genetics (INGEMM), La Paz University Hospital, Madrid, Spain
| | | | - C Caux
- CRCL, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard, Lyon France
| | - M Tommasino
- IARC-International Agency for Research on Cancer 150 Cours Albert Thomas, Lyon, France
| | - N Goutagny
- CRCL, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard, Lyon France
| | - U A Hasan
- CIRI, INSERM U1111, Ecole Normale Supérieure, Université de Lyon, Lyon, France
| |
Collapse
|
14
|
Subcellular compartmentalization of docking protein-1 contributes to progression in colorectal cancer. EBioMedicine 2016; 8:159-172. [PMID: 27428427 PMCID: PMC4919572 DOI: 10.1016/j.ebiom.2016.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/19/2016] [Accepted: 05/04/2016] [Indexed: 12/20/2022] Open
Abstract
Full-length (FL) docking protein-1 (DOK1) is an adapter protein which inhibits growth factor and immune response pathways in normal tissues, but is frequently lost in human cancers. Small DOK1 variants remain in cells of solid tumors and leukemias, albeit, their functions are elusive. To assess the so far unknown role of DOK1 in colorectal cancer (CRC), we generated DOK1 mutants which mimic the domain structure and subcellular distribution of DOK1 protein variants in leukemia patients. We found that cytoplasmic DOK1 activated peroxisome-proliferator-activated-receptor-gamma (PPARγ) resulting in inhibition of the c-FOS promoter and cell proliferation, whereas nuclear DOK1 was inactive. PPARγ-agonist increased expression of endogenous DOK1 and interaction with PPARγ. Forward translation of this cell-based signaling model predicted compartmentalization of DOK1 in patients. In a large series of CRC patients, loss of DOK1 protein was associated with poor prognosis at early tumor stages (*p = 0.001; n = 1492). In tumors with cytoplasmic expression of DOK1, survival was improved, whereas nuclear localization of DOK1 correlated with poor outcome, indicating that compartmentalization of DOK1 is critical for CRC progression. Thus, DOK1 was identified as a prognostic factor for non-metastatic CRC, and, via its drugability by PPARγ-agonist, may constitute a potential target for future cancer treatments. Forward translation of a cell-based signaling model predicted clinical relevance for DOK1 in colorectal cancer (CRC). DOK1 is an independent prognostic factor in CRC patients, and its loss associated with poor survival. Cancer cell growth inhibition by DOK1 was increased (“drugable”) by PPARγ-agonist. Poor survival due to failure to respond to clinical therapies prevents effective treatment of cancer. Thus, there is a high medical need for novel drug targets and biomarkers. DOK1 blocks pro-cancer signaling in the healthy body, but is often lost in tumors. We show that colorectal cancer patients who are positive for DOK1 have a better survival outcome than patients who are negative. Anti-diabetic drugs up-regulated DOK1 and promoted its protective actions against tumor cells. Our study therefore suggests DOK1 as a marker for good prognosis and as a potential drug target for therapy of colorectal cancer.
Collapse
|
15
|
Coppin E, De Grandis M, Pandolfi PP, Arcangeli ML, Aurrand-Lions M, Nunès JA. Dok1 and Dok2 Proteins Regulate Cell Cycle in Hematopoietic Stem and Progenitor Cells. THE JOURNAL OF IMMUNOLOGY 2016; 196:4110-21. [DOI: 10.4049/jimmunol.1501037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 03/11/2016] [Indexed: 01/27/2023]
|
16
|
Araújo OC, Rosa AS, Fernandes A, Niel C, Villela-Nogueira CA, Pannain V, Araujo NM. RASSF1A and DOK1 Promoter Methylation Levels in Hepatocellular Carcinoma, Cirrhotic and Non-Cirrhotic Liver, and Correlation with Liver Cancer in Brazilian Patients. PLoS One 2016; 11:e0153796. [PMID: 27078152 PMCID: PMC4831787 DOI: 10.1371/journal.pone.0153796] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 04/04/2016] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the second most common cause of cancer mortality worldwide. Most cases of HCC are associated with cirrhosis related to chronic hepatitis B virus or hepatitis C virus infections. Hypermethylation of promoter regions is the main epigenetic mechanism of gene silencing and has been involved in HCC development. The aim of this study was to determine whether aberrant methylation of RASSF1A and DOK1 gene promoters is associated with the progression of liver disease in Brazilian patients. Methylation levels were measured by pyrosequencing in 41 (20 HCC, 9 cirrhotic, and 12 non-cirrhotic) liver tissue samples. Mean rates of methylation in RASSF1A and DOK1 were 16.2% and 12.0% in non-cirrhotic, 26.1% and 19.6% in cirrhotic, and 59.1% and 56.0% in HCC tissues, respectively, showing a gradual increase according to the progression of the disease, with significantly higher levels in tumor tissues. In addition, hypermethylation of RASSF1A and DOK1 was found in the vast majority (88%) of the HCC cases. Interestingly, DOK1 methylation levels in HCC samples were significantly higher in the group of younger (<40 years) patients, and higher in moderately differentiated than in poorly differentiated tumors (p < 0.05). Our results reinforce the hypothesis that hypermethylation of RASSF1A and DOK1 contributes to hepatocarcinogenesis and is associated to clinicopathological characteristics. RASSF1A and DOK1 promoter hypermethylation may be a valuable biomarker for early diagnosis of HCC and a potential molecular target for epigenetic-based therapy.
Collapse
Affiliation(s)
- Oscar C Araújo
- Laboratory of Molecular Virology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Agatha S Rosa
- Laboratory of Molecular Virology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Arlete Fernandes
- Department of Pathology, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christian Niel
- Laboratory of Molecular Virology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Cristiane A Villela-Nogueira
- Hepatology Division, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vera Pannain
- Department of Pathology, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia M Araujo
- Laboratory of Molecular Virology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Phosphorylation of Dok1 by Abl family kinases inhibits CrkI transforming activity. Oncogene 2014; 34:2650-9. [PMID: 25043303 PMCID: PMC4302068 DOI: 10.1038/onc.2014.210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 06/13/2014] [Accepted: 06/15/2014] [Indexed: 12/11/2022]
Abstract
The Crk SH2/SH3 adaptor and the Abl nonreceptor tyrosine kinase were first identified as oncoproteins, and both can induce tumorigenesis when overexpressed or mutationally activated. We previously reported the surprising finding that inhibition or knockdown of Abl family kinases enhanced transformation of mouse fibroblasts by CrkI. Abl family inhibitors are currently used or are being tested for treatment of human malignancies, and our finding raised concerns that such inhibitors might actually promote the growth of tumors overexpressing CrkI. Here, we identify the Dok1 adaptor as the key effector for the enhancement of CrkI transformation by Abl inhibition. We show that phosphorylation of tyrosines 295 and 361 of Dok1 by Abl family kinases suppresses CrkI transforming activity, and that upon phosphorylation these tyrosines bind the SH2 domains of the Ras inhibitor p120 RasGAP. Knockdown of RasGAP resulted in a similar enhancement of CrkI transformation, consistent with a critical role for Ras activity. Imaging studies using a FRET sensor of Ras activation revealed alterations in the localization of activated Ras in CrkI-transformed cells. Our results support a model in which Dok1 phosphorylation normally suppresses localized Ras pathway activity in Crk-transformed cells via recruitment and/or activation of RasGAP, and that preventing this negative feedback mechanism by inhibiting Abl family kinases leads to enhanced transformation by Crk.
Collapse
|
18
|
Epstein-Barr virus down-regulates tumor suppressor DOK1 expression. PLoS Pathog 2014; 10:e1004125. [PMID: 24809689 PMCID: PMC4014463 DOI: 10.1371/journal.ppat.1004125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 04/01/2014] [Indexed: 12/29/2022] Open
Abstract
The DOK1 tumor suppressor gene encodes an adapter protein that acts as a negative regulator of several signaling pathways. We have previously reported that DOK1 expression is up-regulated upon cellular stress, via the transcription factor E2F1, and down-regulated in a variety of human malignancies due to aberrant hypermethylation of its promoter. Here we show that Epstein Barr virus (EBV) infection of primary human B-cells leads to the down-regulation of DOK1 gene expression via the viral oncoprotein LMP1. LMP1 alone induces recruitment to the DOK1 promoter of at least two independent inhibitory complexes, one containing E2F1/pRB/DNMT1 and another containing at least EZH2. These events result in tri-methylation of histone H3 at lysine 27 (H3K27me3) of the DOK1 promoter and gene expression silencing. We also present evidence that the presence of additional EBV proteins leads to further repression of DOK1 expression with an additional mechanism. Indeed, EBV infection of B-cells induces DNA methylation at the DOK1 promoter region including the E2F1 responsive elements that, in turn, lose the ability to interact with E2F complexes. Treatment of EBV-infected B-cell-lines with the methyl-transferase inhibitor 5-aza-2′-deoxycytidine rescues DOK1 expression. In summary, our data show the deregulation of DOK1 gene expression by EBV and provide novel insights into the regulation of the DOK1 tumor suppressor in viral-related carcinogenesis. Many oncogenic viruses exhibit cellular transforming properties, often involving oncogenes activation and tumor suppressor genes inactivation. The DOK1 gene is a newly identified tumor suppressor gene with altered expression via hypermethylation of its promoter in a variety of human cancers, including head and neck, lung, gastric and others. In addition, a correlation has been reported between DOK1 aberrant hypermethylation and the presence of oncogenic viruses such as hepatitis B virus (HBV) in hepatocellular carcinoma (HCC) and Epstein-Barr virus (EBV) in Burkitt's lymphoma-derived cell lines. Here we demonstrate for the first time that EBV is directly involved in the inhibition of DOK1 expression in B-cells. We show that EBV leads to epigenetic repression of DOK1 through increased DNA methylation of its promoter and H3K27 tri-methylation. The LMP1 oncoprotein plays a key role in the repression of DOK1 expression. It promotes the formation and the recruitment to the DOK1 promoter of transcriptionally inhibitory complexes composed of E2F1/pRB/DNMT1 and of EZH2 which is part of the polycomb repressive complex 2. Interestingly, one or more additional EBV protein(s) cooperate(s) with LMP1 in inducing massive DNA methylation at the DOK1 promoter, leading to the loss of E2F1 complexes recruitment and even stronger repression of DOK1 expression.
Collapse
|
19
|
Zhang H, Nan X, Li X, Chen Y, Zhang J, Sun L, Han W, Li T. CMTM5 exhibits tumor suppressor activity through promoter methylation in oral squamous cell carcinoma. Biochem Biophys Res Commun 2014; 447:304-10. [DOI: 10.1016/j.bbrc.2014.03.158] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 03/28/2014] [Indexed: 12/29/2022]
|
20
|
Miah S, Goel RK, Dai C, Kalra N, Beaton-Brown E, Bagu ET, Bonham K, Lukong KE. BRK targets Dok1 for ubiquitin-mediated proteasomal degradation to promote cell proliferation and migration. PLoS One 2014; 9:e87684. [PMID: 24523872 PMCID: PMC3921129 DOI: 10.1371/journal.pone.0087684] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 01/02/2014] [Indexed: 12/13/2022] Open
Abstract
Breast tumor kinase (BRK), also known as protein tyrosine kinase 6 (PTK6), is a non-receptor tyrosine kinase overexpressed in more that 60% of human breast carcinomas. The overexpression of BRK has been shown to sensitize mammary epithelial cells to mitogenic signaling and to promote cell proliferation and tumor formation. The molecular mechanisms of BRK have been unveiled by the identification and characterization of BRK target proteins. Downstream of tyrosine kinases 1 or Dok1 is a scaffolding protein and a substrate of several tyrosine kinases. Herein we show that BRK interacts with and phosphorylates Dok1 specifically on Y362. We demonstrate that this phosphorylation by BRK significantly downregulates Dok1 in a ubiquitin-proteasome-mediated mechanism. Together, these results suggest a novel mechanism of action of BRK in the promotion of tumor formation, which involves the targeting of tumor suppressor Dok1 for degradation through the ubiquitin proteasomal pathway.
Collapse
Affiliation(s)
- Sayem Miah
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Raghuveera Kumar Goel
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Chenlu Dai
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Natasha Kalra
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Erika Beaton-Brown
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Cancer Research Unit, Health Research Division, Saskatchewan Cancer Agency, and Division of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Edward T. Bagu
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Cancer Research Unit, Health Research Division, Saskatchewan Cancer Agency, and Division of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Keith Bonham
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
- Cancer Research Unit, Health Research Division, Saskatchewan Cancer Agency, and Division of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Kiven E. Lukong
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
21
|
Fleischhacker M, Dietrich D, Liebenberg V, Field JK, Schmidt B. The role of DNA methylation as biomarkers in the clinical management of lung cancer. Expert Rev Respir Med 2014; 7:363-83. [DOI: 10.1586/17476348.2013.814397] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
22
|
Bracken J, Ghanem T, Kasem A, Jiang WG, Mokbel K. Evidence for Tumour Suppressor Function of DOK7 in Human Breast Cancer. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/jct.2014.51009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
23
|
Abstract
BACKGROUND A remarkable correspondence exists between the cytogenetic locations of the known fragile sites and frequently reported sites of hypermethylation. The best-known features of fragile sites are sequence motifs that are prone to the spontaneous formation of a non-B DNA structure. These facts, coupled with the known enzymological specificities of DNA methyltransferase 1 (DNMT1), the ATP-dependent and actin-dependent helicases, and the ten-eleven translocation (TET) dioxygenases, suggest that these enzymes are involved in an epigenetic cycle that maintains the unmethylated state at these sites by resolving non-B structure, preventing both the sequestration of DNA methyltransferases (DNMTs) and hypermethylation in normal cells. PRESENTATION OF THE HYPOTHESIS The innate tendency of DNA sequences present at fragile sites to form non-B DNA structures results in de novo methylation of DNA at these sites that is held in check in normal cells by the action of ATP-dependent and actin-dependent helicases coupled with the action of TET dioxygenases. This constitutes a previously unrecognized epigenetic repair cycle in which spontaneously forming non-B DNA structures formed at fragile sites are methylated by DNMTs as they are removed by the action of ATP-dependent and actin-dependent helicases, with the resulting nascent methylation rendered non-transmissible by TET dioxygenases. TESTING THE HYPOTHESIS A strong prediction of the hypothesis is that knockdown of ATP-dependent and actin-dependent helicases will result in enhanced bisulfite sensitivity and hypermethylation at non-B structures in multiple fragile sites coupled with global hypomethylation. IMPLICATIONS OF THE HYPOTHESIS A key implication of the hypothesis is that helicases, like the lymphoid-specific helicase and alpha thalassemia/mental retardation syndrome X-linked helicase, passively promote accurate maintenance of DNA methylation by preventing the sequestration of DNMTs at sites of unrepaired non-B DNA structure. When helicase action is blocked due to mutation or downregulation of the respective genes, DNMTs stall at unrepaired non-B structures in fragile sites after methylating them and are unable to methylate other sites in the genome, resulting in hypermethylation at non-B DNA-forming sites, along with hypomethylation elsewhere.
Collapse
Affiliation(s)
- Steven S Smith
- City of Hope, 1500 East Duarte Road, Duarte, CA 91010, USA.
| |
Collapse
|
24
|
Goel RK, Miah S, Black K, Kalra N, Dai C, Lukong KE. The unique N-terminal region of SRMS regulates enzymatic activity and phosphorylation of its novel substrate docking protein 1. FEBS J 2013; 280:4539-59. [PMID: 23822091 DOI: 10.1111/febs.12420] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 06/20/2013] [Accepted: 06/25/2013] [Indexed: 01/07/2023]
Abstract
SRMS (Src-related tyrosine kinase lacking C-terminal regulatory tyrosine and N-terminal myristoylation sites) belongs to a family of nonreceptor tyrosine kinases, which also includes breast tumour kinase and Fyn-related kinase. SRMS, similar to breast tumour kinase and Fyn-related kinase, harbours a Src homology 3 and Src homology 2, as well as a protein kinase domain. However, unlike breast tumour kinase and Fyn-related kinase, SRMS lacks a C-terminal regulatory tail but distinctively possesses an extended N-terminal region. Both breast tumour kinase and Fyn-related kinase play opposing roles in cell proliferation and signalling. SRMS, however, is an understudied member of this family. Although cloned in 1994, information on the biochemical, cellular and physiological roles of SRMS remains unreported. The present study is the first to explore the expression pattern of SRMS in breast cancers, its enzymatic activity and autoregulatory elements, and the characterization of docking protein 1 as its first bonafide substrate. We found that, similar to breast tumour kinase, SRMS is highly expressed in most breast cancers compared to normal mammary cell lines and tissues. We generated a series of SRMS point and deletion mutants and assessed enzymatic activity, subcellular localization and substrate recognition. We report for the first time that ectopically-expressed SRMS is constitutively active and that its N-terminal region regulates the enzymatic activity of the protein. Finally, we present evidence indicating that docking protein 1 is a direct substrate of SRMS. Our data demonstrate that, unlike members of the Src family, the enzymatic activity of SRMS is regulated by the intramolecular interactions involving the N-terminus of the enzyme and that docking protein 1 is a bona fide substrate of SRMS.
Collapse
Affiliation(s)
- Raghuveera K Goel
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
The expression of the tumor suppressor DOK1 is repressed in a variety of human tumors as a result of hypermethylation of its promoter region. However, the molecular mechanisms by which DOK1 expression is regulated have been poorly investigated. Here, we show that the expression of DOK1 is regulated mainly by the transcription factor E2F1. We identified three putative E2F1 response elements (EREs) in the DOK1 promoter region. E2F1 had a relatively higher binding affinity for the ERE located between bp -498 and -486 compared with the other two EREs. E2F1 gene silencing strongly inhibited DOK1 expression. E2F1-driven DOK1 transcription occurred in the presence of cellular stresses, such as accumulation of DNA damage induced by etoposide. DOK1 silencing promoted cell proliferation and protected against etoposide-induced apoptosis, indicating that DOK1 acts as a key mediator of cellular stress-induced cell death. Most importantly, we observed that DNA methylation of the DOK1 core promoter region found in head and neck cancer cell lines hampered the recruitment of E2F1 to the DOK1 promoter and compromised DOK1 expression. In summary, our data show that E2F1 is a key factor in DOK1 expression and provide novel insights into the regulation of these events in cancer cells.
Collapse
|
26
|
Mani S, Szymańska K, Cuenin C, Zaridze D, Balassiano K, Lima SCS, Matos E, Daudt A, Koifman S, Filho VW, Menezes AMB, Curado MP, Ferro G, Vaissière T, Sylla BS, Tommasino M, Pinto LFR, Boffetta P, Hainaut P, Brennan P, Herceg Z. DNA methylation changes associated with risk factors in tumors of the upper aerodigestive tract. Epigenetics 2012; 7:270-7. [PMID: 22430803 PMCID: PMC3335950 DOI: 10.4161/epi.7.3.19306] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 01/05/2012] [Accepted: 01/09/2012] [Indexed: 12/20/2022] Open
Abstract
Cancers of the upper aerodigestive tract (UADT) are common forms of malignancy associated with tobacco and alcohol exposures, although human papillomavirus and nutritional deficiency are also important risk factors. While somatically acquired DNA methylation changes have been associated with UADT cancers, what triggers these events and precise epigenetic targets are poorly understood. In this study, we applied quantitative profiling of DNA methylation states in a panel of cancer-associated genes to a case-control study of UADT cancers. Our analyses revealed a high frequency of aberrant hypermethylation of several genes, including MYOD1, CHRNA3 and MTHFR in UADT tumors, whereas CDKN2A was moderately hypermethylated. Among differentially methylated genes, we identified a new gene (the nicotinic acetycholine receptor gene) as target of aberrant hypermethylation in UADT cancers, suggesting that epigenetic deregulation of nicotinic acetycholine receptors in non-neuronal tissues may promote the development of UADT cancers. Importantly, we found that sex and age is strongly associated with the methylation states, whereas tobacco smoking and alcohol intake may also influence the methylation levels in specific genes. This study identifies aberrant DNA methylation patterns in UADT cancers and suggests a potential mechanism by which environmental factors may deregulate key cellular genes involved in tumor suppression and contribute to UADT cancers.
Collapse
Affiliation(s)
- Samson Mani
- International Agency for Research on Cancer (IARC); Lyon, France
| | | | - Cyrille Cuenin
- International Agency for Research on Cancer (IARC); Lyon, France
| | | | - Karen Balassiano
- International Agency for Research on Cancer (IARC); Lyon, France
| | - Sheila CS Lima
- International Agency for Research on Cancer (IARC); Lyon, France
- Divisão de Genética; Instituto Nacional de Câncer; Rio de Janeiro, Brazil
| | - Elena Matos
- Institut of Oncology Angel H. Roffo; University of Buenos Aires; Buenos Aires, Argentina
| | | | - Sergio Koifman
- Escola Nacional de Saude Publica; Rio de Janeiro, Brazil
| | | | | | | | - Gilles Ferro
- International Agency for Research on Cancer (IARC); Lyon, France
| | - Thomas Vaissière
- International Agency for Research on Cancer (IARC); Lyon, France
| | - Bakary S Sylla
- International Agency for Research on Cancer (IARC); Lyon, France
| | | | - Luis Felipe Ribeiro Pinto
- Divisão de Genética; Instituto Nacional de Câncer; Rio de Janeiro, Brazil
- Departamento de Bioquímica; Universidade do Estado do Rio de Janeiro; Instituto de Biologia Roberto Alcantara Gomes; Rio de Janeiro, Brazil
| | - Paolo Boffetta
- The International Prevention Research Institute; Lyon, France
| | - Pierre Hainaut
- International Agency for Research on Cancer (IARC); Lyon, France
| | - Paul Brennan
- International Agency for Research on Cancer (IARC); Lyon, France
| | - Zdenko Herceg
- International Agency for Research on Cancer (IARC); Lyon, France
| |
Collapse
|
27
|
Liu L, Ling X, Liang H, Gao Y, Yang H, Shao J, Tang H. Hypomethylation mediated by decreased DNMTs involves in the activation of proto-oncogene MPL in TK6 cells treated with hydroquinone. Toxicol Lett 2012; 209:239-45. [PMID: 22245671 DOI: 10.1016/j.toxlet.2011.12.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2011] [Revised: 12/28/2011] [Accepted: 12/29/2011] [Indexed: 12/15/2022]
Abstract
Hydroquinone (HQ), one of the most important metabolites derived from benzene, is known to be associated with acute myelogenous leukemia (AML) risk, however, its carcinogenic mechanism remains unclear. In this study, the epigenetic mechanism of HQ exposure was investigated. We characterized the epigenomic response of TK6 cells to HQ exposure, and examined the mRNA expression of DNA methyltransferases (DNMTs) including DNMT1, DNMT3a and DNMT3b, methyl-CpG-binding domain protein 2 (MBD2) and six proto-oncogenes (MPL, RAF1, MYB, MYC, ERBB2 and BRAF). Compared to the control cells, HQ exposure (2.5, 5.0, 10.0 and 20.0 μM for 48 h) resulted in the decrease of DNMTs and MBD2 expression, the global hypomethylation and increase of MPL at mRNA level. Meanwhile, most of these changes were in dose-dependent manner. Moreover, inhibition of DNMTs induced by 5-aza-2'-deoxycytidine (5-AZA), an identified DNMT inhibitor, caused more induction of MPL expression at mRNA level compared to the HQ (10.0 μM) pre-treated group. Furthermore, treatment of HQ potentially led to MPL itself hypomethylation (10.0 and 20.0 μM reduced by 47% and 44%, respectively), further revealing that the activation of proto-oncogene MPL was related to hypomethylation in its DNA sequences. In conclusion, hypomethylation, including global and specific hypomethylation, might be involved in the activation of MPL, and the hypomethylation could be induced by decreased DNMTs in TK6 cells exposed to HQ.
Collapse
Affiliation(s)
- Linhua Liu
- Department of Environmental and Occupational Health, School of Public Health, Guangdong Medical College, Dongguan 523808, PR China
| | | | | | | | | | | | | |
Collapse
|