1
|
Zhang X, Li P, Gan Y, Xiang S, Gu L, Zhou J, Zhou X, Wu P, Zhang B, Deng D. Driving effect of P16 methylation on telomerase reverse transcriptase-mediated immortalization and transformation of normal human fibroblasts. Chin Med J (Engl) 2024:00029330-990000000-00975. [PMID: 38420748 DOI: 10.1097/cm9.0000000000003004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND P16 inactivation is frequently accompanied by telomerase reverse transcriptase (TERT) amplification in human cancer genomes. P16 inactivation by DNA methylation often occurs automatically during immortalization of normal cells by TERT. However, direct evidence remains to be obtained to support the causal effect of epigenetic changes, such as P16 methylation, on cancer development. This study aimed to provide experimental evidence that P16 methylation directly drives cancer development. METHODS A zinc finger protein-based P16-specific DNA methyltransferase (P16-Dnmt) vector containing a "Tet-On" switch was used to induce extensive methylation of P16 CpG islands in normal human fibroblast CCD-18Co cells. Battery assays were used to evaluate cell immortalization and transformation throughout their lifespan. Cell subcloning and DNA barcoding were used to track the diversity of cell evolution. RESULTS Leaking P16-Dnmt expression (without doxycycline-induction) could specifically inactivate P16 expression by DNA methylation. P16 methylation only promoted proliferation and prolonged lifespan but did not induce immortalization of CCD-18Co cells. Notably, cell immortalization, loss of contact inhibition, and anchorage-independent growth were always prevalent in P16-Dnmt&TERT cells, indicating cell transformation. In contrast, almost all TERT cells died in the replicative crisis. Only a few TERT cells recovered from the crisis, in which spontaneous P16 inactivation by DNA methylation occurred. Furthermore, the subclone formation capacity of P16-Dnmt&TERT cells was two-fold that of TERT cells. DNA barcoding analysis showed that the diversity of the P16-Dnmt&TERT cell population was much greater than that of the TERT cell population. CONCLUSION P16 methylation drives TERT-mediated immortalization and transformation of normal human cells that may contribute to cancer development.
Collapse
Affiliation(s)
- Xuehong Zhang
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Paiyun Li
- Division of Etiology, Beijing Cancer Hospital, Beijing 100142, China
- Radiation Oncology Department, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ying Gan
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Shengyan Xiang
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Liankun Gu
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jing Zhou
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Xiaorui Zhou
- Department of Biomedical Engineering, Peking University Cancer Hospital and Institute, Beijing 100871, China
| | - Peihuang Wu
- Department of Biomedical Engineering, Peking University Cancer Hospital and Institute, Beijing 100871, China
| | - Baozhen Zhang
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
- Division of Etiology, Beijing Cancer Hospital, Beijing 100142, China
| | - Dajun Deng
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| |
Collapse
|
2
|
Raczkowska J, Bielska A, Krętowski A, Niemira M. Extracellular circulating miRNAs as potential non-invasive biomarkers in non-small cell lung cancer patients. Front Oncol 2023; 13:1209299. [PMID: 37546401 PMCID: PMC10401434 DOI: 10.3389/fonc.2023.1209299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/28/2023] [Indexed: 08/08/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) comprises 85% of all lung cancers and is a malignant condition resistant to advanced-stage treatment. Despite the advancement in detection and treatment techniques, the disease is taking a deadly toll worldwide, being the leading cause of cancer death every year. Current diagnostic methods do not ensure the detection of the disease at an early stage, nor can they predict the risk of its development. There is an urgent need to identify biomarkers that can help predict an individual's risk of developing NSCLC, distinguish NSCLC subtype, allow monitor disease and treatment progression which can improve patient survival. Micro RNAs (miRNAs) represent the class of small and non-coding RNAs involved in gene expression regulation, influencing many biological processes such as proliferation, differentiation, and carcinogenesis. Research reports significant differences in miRNA profiles between healthy and neoplastic tissues in NSCLC. Its abundant presence in biofluids, such as serum, blood, urine, and saliva, makes them easily detectable and does not require invasive collection techniques. Many studies support miRNAs' importance in detecting, predicting, and prognosis of NSCLC, indicating their utility as a promising biomarker. In this work, we reviewed up-to-date research focusing on biofluid miRNAs' role as a diagnostic tool in NSCLC cases. We also discussed the limitations of applying miRNAs as biomarkers and highlighted future areas of interest.
Collapse
Affiliation(s)
- Justyna Raczkowska
- Clinical Research Centre, Medical University of Białystok, Białystok, Poland
| | - Agnieszka Bielska
- Clinical Research Centre, Medical University of Białystok, Białystok, Poland
| | - Adam Krętowski
- Clinical Research Centre, Medical University of Białystok, Białystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Białystok, Białystok, Poland
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
3
|
Pourasghariazar M, Zarredar H, Asadi M, Caner A, Akhgari A, Valizadeh H, Bornehdeli S, Hashemzadeh S, Raeisi M. Comparative evaluation of ZMYND-8 and RARβ2 genes promoters’ methylation changes in tumor and tumor margin tissues of patients with lung cancer. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2023. [DOI: 10.1186/s43042-023-00399-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2023] Open
Abstract
Abstract
Background
Lung cancer remains one of the most lethal carcinomas worldwide because of its late diagnosis. One of the DNA modifications is methylation, one of the primary alterations of tumor development, consisting of fascinating indicators for cancer diagnosis. This study investigated ZMYND-8 and RARβ2 gene methylation in NSCLC as a new epigenetic tool.
Methods
First, to find out the potential diagnostic capability of ZMYND-8 and RARβ2 genes methylation, we entirely surfed DNA methylation microarrays from the Cancer Genome Atlas (TCGA) data of NSCLC samples. Additionally, we took advantage of using q-MSP in several pieces comprising NSCLC tumors and neighboring normal tissues; ZMYND-8 and RARβ2 genes methylation grades were acquired.
Results
Our finding displayed significant hypomethylation of ZMYND-8 and hypermethylation of RARβ2 in NSCLC samples compared to neighboring standard specimens, which significantly correlated with the clinical stage of malignancy. In addition, the incredible precision of ZMYND-8 and RARβ2 methylations as reliable cancer diagnosis indicators in NSCLC was confirmed, drawing the ROC curve analysis with an AUC value of 0.751 and 0.8676, respectively, for ZMYND-8 and RARβ2. Additional studies of other dominant cancer entities in TCGA displayed that RARβ2’s higher methylation degree and ZMYND-8 lower methylation degree are prevalent changes in tumor evolution which could be possibly considered as a potential diagnostic biomarkers for lung cancer.
Conclusion
Based on this study, ZMYND-8 and RARβ2 methylation are reliable biomarkers for lung cancer.
Collapse
|
4
|
MAYLINA L, KAMBAYASHI S, BABA K, IGASE M, MIZUNO T, OKUDA M. Decreased sensitivity of cyclin-dependent kinase 4/6 inhibitors, palbociclib and abemaciclib to canine lymphoma cells with high p16 protein expression and low retinoblastoma protein phosphorylation. J Vet Med Sci 2023; 85:99-104. [PMID: 36450591 PMCID: PMC9887221 DOI: 10.1292/jvms.22-0498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Canine lymphoma/leukemia cell lines with p16 protein expressions: high (17-71 and GL-1) and low (CLBL-1, CLC, Nody-1, and UL-1) were treated in vitro with cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors, palbociclib or abemaciclib. Cell proliferation decreased as a result, with higher IC50 levels observed in the high p16 (17-71 and GL-1) and one low p16 (UL-1) cell lines compared with the low p16 cells (CLBL-1, CLC, and Nody-1). As expected, palbociclib and abemaciclib treatment reduced pRb phosphorylation in a dose-dependent manner, especially in cells with low p16. These results suggest that CDK4/6 inhibitors have potential as new chemotherapeutic agents for canine lymphoma and high p16 protein expression may be used as a biomarker for resistance to CDK4/6 inhibitor therapy.
Collapse
Affiliation(s)
- Leni MAYLINA
- Laboratory of Veterinary Internal Medicine, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan,Division of Veterinary Internal Medicine, Department of Clinic, Reproduction and Pathology, School of Veterinary Medicine and Biomedical Sciences, IPB
University, Bogor, Indonesia
| | - Satoshi KAMBAYASHI
- Laboratory of Veterinary Internal Medicine, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan,Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Kenji BABA
- Laboratory of Veterinary Internal Medicine, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan,Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masaya IGASE
- Laboratory of Molecular Diagnosis and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan,Laboratory of Molecular Diagnosis and Therapeutics, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Takuya MIZUNO
- Laboratory of Molecular Diagnosis and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan,Laboratory of Molecular Diagnosis and Therapeutics, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan
| | - Masaru OKUDA
- Laboratory of Veterinary Internal Medicine, Joint Graduate School of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan,Laboratory of Veterinary Internal Medicine, Joint Faculty of Veterinary Medicine, Yamaguchi University, Yamaguchi, Japan,Correspondence to: Okuda M: , Laboratory of Veterinary Internal Medicine,
Joint Faculty of Veterinary Medicine, Yamaguchi University, Yoshida 1677-1, Yamaguchi 753-8515, Japan
| |
Collapse
|
5
|
Li P, Liu S, Du L, Mohseni G, Zhang Y, Wang C. Liquid biopsies based on DNA methylation as biomarkers for the detection and prognosis of lung cancer. Clin Epigenetics 2022; 14:118. [PMID: 36153611 PMCID: PMC9509651 DOI: 10.1186/s13148-022-01337-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 09/16/2022] [Indexed: 11/27/2022] Open
Abstract
Lung cancer (LC) is the main cause of cancer-related mortality. Most LC patients are diagnosed in an advanced stage when the symptoms are obvious, and the prognosis is quite poor. Although low-dose computed tomography (LDCT) is a routine clinical examination for early detection of LC, the false-positive rate is over 90%. As one of the intensely studied epigenetic modifications, DNA methylation plays a key role in various diseases, including cancer and other diseases. Hypermethylation in tumor suppressor genes or hypomethylation in oncogenes is an important event in tumorigenesis. Remarkably, DNA methylation usually occurs in the very early stage of malignant tumors. Thus, DNA methylation analysis may provide some useful information about the early detection of LC. In recent years, liquid biopsy has developed rapidly. Liquid biopsy can detect and monitor both primary and metastatic malignant tumors and can reflect tumor heterogeneity. Moreover, it is a minimally invasive procedure, and it causes less pain for patients. This review summarized various liquid biopsies based on DNA methylation for LC. At first, we briefly discussed some emerging technologies for DNA methylation analysis. Subsequently, we outlined cell-free DNA (cfDNA), sputum, bronchoalveolar lavage fluid, bronchial aspirates, and bronchial washings DNA methylation-based liquid biopsy for the early detection of LC. Finally, the prognostic value of DNA methylation in cfDNA and sputum and the diagnostic value of other DNA methylation-based liquid biopsies for LC were also analyzed.
Collapse
|
6
|
Non-Invasive Biomarkers for Early Lung Cancer Detection. Cancers (Basel) 2022; 14:cancers14235782. [PMID: 36497263 PMCID: PMC9739091 DOI: 10.3390/cancers14235782] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 11/27/2022] Open
Abstract
Worldwide, lung cancer (LC) is the most common cause of cancer death, and any delay in the detection of new and relapsed disease serves as a major factor for a significant proportion of LC morbidity and mortality. Though invasive methods such as tissue biopsy are considered the gold standard for diagnosis and disease monitoring, they have several limitations. Therefore, there is an urgent need to identify and validate non-invasive biomarkers for the early diagnosis, prognosis, and treatment of lung cancer for improved patient management. Despite recent progress in the identification of non-invasive biomarkers, currently, there is a shortage of reliable and accessible biomarkers demonstrating high sensitivity and specificity for LC detection. In this review, we aim to cover the latest developments in the field, including the utility of biomarkers that are currently used in LC screening and diagnosis. We comment on their limitations and summarise the findings and developmental stages of potential molecular contenders such as microRNAs, circulating tumour DNA, and methylation markers. Furthermore, we summarise research challenges in the development of biomarkers used for screening purposes and the potential clinical applications of newly discovered biomarkers.
Collapse
|
7
|
Leng S, Picchi MA, Meek PM, Jiang M, Bayliss SH, Zhai T, Bayliyev RI, Tesfaigzi Y, Campen MJ, Kang H, Zhu Y, Lan Q, Sood A, Belinsky SA. Wood smoke exposure affects lung aging, quality of life, and all-cause mortality in New Mexican smokers. Respir Res 2022; 23:236. [PMID: 36076291 PMCID: PMC9454202 DOI: 10.1186/s12931-022-02162-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/27/2022] [Indexed: 11/10/2022] Open
Abstract
Background The role of wood smoke (WS) exposure in the etiology of chronic obstructive pulmonary disease (COPD), lung cancer (LC), and mortality remains elusive in adults from countries with low ambient levels of combustion-emitted particulate matter. This study aims to delineate the impact of WS exposure on lung health and mortality in adults age 40 and older who ever smoked. Methods We assessed health impact of self-reported “ever WS exposure for over a year” in the Lovelace Smokers Cohort using both objective measures (i.e., lung function decline, LC incidence, and deaths) and two health related quality-of-life questionnaires (i.e., lung disease-specific St. George's Respiratory Questionnaire [SGRQ] and the generic 36-item short-form health survey). Results Compared to subjects without WS exposure, subjects with WS exposure had a more rapid decline of FEV1 (− 4.3 ml/s, P = 0.025) and FEV1/FVC ratio (− 0.093%, P = 0.015), but not of FVC (− 2.4 ml, P = 0.30). Age modified the impacts of WS exposure on lung function decline. WS exposure impaired all health domains with the increase in SGRQ scores exceeding the minimal clinically important difference. WS exposure increased hazard for incidence of LC and death of all-cause, cardiopulmonary diseases, and cancers by > 50% and shortened the lifespan by 3.5 year. We found no evidence for differential misclassification or confounding from socioeconomic status for the health effects of WS exposure. Conclusions We identified epidemiological evidence supporting WS exposure as an independent etiological factor for the development of COPD through accelerating lung function decline in an obstructive pattern. Time-to-event analyses of LC incidence and cancer-specific mortality provide human evidence supporting the carcinogenicity of WS exposure. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02162-y.
Collapse
Affiliation(s)
- Shuguang Leng
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA. .,Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA. .,Lung Cancer Program, Lovelace Biomedical Research Institute, Albuquerque, NM, 87108, USA.
| | - Maria A Picchi
- Lung Cancer Program, Lovelace Biomedical Research Institute, Albuquerque, NM, 87108, USA
| | - Paula M Meek
- College of Nursing, University of Utah, Salt Lake City, UT, 84112, USA
| | - Menghui Jiang
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Samuel H Bayliss
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Ting Zhai
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA.,Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Ruslan I Bayliyev
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Yohannes Tesfaigzi
- Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 01255, USA
| | - Matthew J Campen
- Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA.,College of Pharmacy, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Huining Kang
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA.,Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA
| | - Yiliang Zhu
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Akshay Sood
- Department of Internal Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Steven A Belinsky
- Cancer Control and Population Sciences, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM, 87131, USA.,Lung Cancer Program, Lovelace Biomedical Research Institute, Albuquerque, NM, 87108, USA
| |
Collapse
|
8
|
Frattini M, Froesch P, Epistolio S. Overview of recent advances in molecular analysis for diagnosing early stage lung cancer nodules. Transl Lung Cancer Res 2022; 10:4303-4307. [PMID: 35004258 PMCID: PMC8674592 DOI: 10.21037/tlcr-21-802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/13/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Milo Frattini
- Laboratory of Molecular Pathology, Institute of Pathology (ICP), Cantonal Hospital (EOC), Locarno, Switzerland
| | - Patrizia Froesch
- Oncology Institute of Southern Switzerland (IOSI), Cantonal Hospital (EOC), Bellinzona, Switzerland
| | - Samantha Epistolio
- Laboratory of Molecular Pathology, Institute of Pathology (ICP), Cantonal Hospital (EOC), Locarno, Switzerland
| |
Collapse
|
9
|
Si H, Du D, Li W, Li Q, Li J, Zhao D, Li L, Tang B. Sputum-Based Tumor Fluid Biopsy: Isolation and High-Throughput Single-Cell Analysis of Exfoliated Tumor Cells for Lung Cancer Diagnosis. Anal Chem 2021; 93:10477-10486. [PMID: 34292723 DOI: 10.1021/acs.analchem.1c00833] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Timely and effective diagnosis is of great significance for improving the survival rate of lung cancer patients. Although histopathology is the main diagnostic tool among the existing methods for lung cancer diagnosis, it is not suitable for high-risk groups, early lung cancer patients, patients with advanced-stage disease, and other situations wherein tumor tissues cannot be obtained. In view of this, we proposed an innovative lung cancer diagnosis method employing for the first time a microfluidic technology for high-efficiency isolation and high-throughput single-cell analysis of exfoliated tumor cells (ETCs) in sputum. This method fully combines the advantages of traditional sputum cytology and microfluidic technology and realizes the diagnosis of lung cancer by using a small amount of repeatable ETCs instead of the tumor tissue. This method is expected to provide a practical strategy for the non-invasive detection of lung cancer patients and lung cancer screening for high-risk groups.
Collapse
Affiliation(s)
- Haibin Si
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China
| | - Dexin Du
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China
| | - Wenbo Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China
| | - Qingling Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Dongbo Zhao
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250117, P. R. China
| | - Lu Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, P. R. China
| |
Collapse
|
10
|
Li N, Dhilipkannah P, Jiang F. High-Throughput Detection of Multiple miRNAs and Methylated DNA by Droplet Digital PCR. J Pers Med 2021; 11:jpm11050359. [PMID: 33946992 PMCID: PMC8146424 DOI: 10.3390/jpm11050359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/14/2021] [Accepted: 04/22/2021] [Indexed: 01/04/2023] Open
Abstract
Altered miRNA expression and DNA methylation have highly active and diverse roles in carcinogenesis. Simultaneous detection of the molecular aberrations may have a synergistic effect on the diagnosis of malignancies. Herein, we develop a high-throughput assay for detecting multiple miRNAs and DNA methylation using droplet digital PCR (ddPCR) coupled with a 96-microwell plate. The microplate-based ddPCR could absolutely and reproducibly quantify 15 miRNAs and 14 DNA methylation sites with a high sensitivity (one copy/µL and 0.1%, respectively). Analyzing sputum and plasma of 40 lung cancer patients and 36 cancer-free smokers by this approach identified an integrated biomarker panel consisting of two sputum miRNAs (miRs-31-5p and 210-3p), one sputum DNA methylation (RASSF1A), and two plasma miRNAs (miR-21-5p and 126) for the diagnosis of lung cancer with higher sensitivity and specificity compared with a single type of biomarker. The diagnostic value of the integrated biomarker panel for the early detection of lung cancer was confirmed in a different cohort of 36 lung cancer patients and 39 cancer-free smokers. The high-throughput assay for quantification of multiple molecular aberrations across sputum and plasma could improve the early detection of lung cancer.
Collapse
|
11
|
Baszuk P, Janasik B, Pietrzak S, Marciniak W, Reszka E, Białkowska K, Jabłońska E, Muszyńska M, Lesicka M, Derkacz R, Grodzki T, Wójcik J, Wojtyś M, Dębniak T, Cybulski C, Gronwald J, Kubisa B, Wójcik N, Pieróg J, Gajić D, Waloszczyk P, Scott RJ, Wąsowicz W, Jakubowska A, Lubiński J, Lener MR. Lung Cancer Occurrence-Correlation with Serum Chromium Levels and Genotypes. Biol Trace Elem Res 2021; 199:1228-1236. [PMID: 32648197 PMCID: PMC7886837 DOI: 10.1007/s12011-020-02240-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 06/08/2020] [Indexed: 12/19/2022]
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Exposure to environmental and occupational carcinogens is an important cause of lung cancer. One of these substances is chromium, which is found ubiquitously across the planet. The International Agency for Research on Cancer has classified chromium(VI) as a human carcinogen. The aim of this study was to assess whether serum chromium levels, as well as DNA variants in selected genes involved in carcinogenesis, xenobiotic-metabolism, and oxidative stress could be helpful in the detection of lung cancer. We conducted a study using 218 lung cancer patients and 218 matched healthy controls. We measured serum chromium levels and genotyped ten genetic variants in ERCC2, XRCC1, MT1B, GSTP1, ABCB1, NQ01, CRTC3, GPX1, SOD2 and CAT. The odds ratios of being diagnosed with lung cancer were calculated using conditional logistic regression with respect to serum chromium level and genotypes. The odds ratio for the occurrence of lung cancer increased with increasing serum chromium levels. The difference between the quartiles with the lowest vs. highest chromium level was more than fourfold in the entire group (OR 4.52, CI 2.17-9.42, p < 0.01). This correlation was significantly increased by more than twice when specific genotypes were taken into consideration (ERCC-rs12181 TT, OR 12.34, CI 1.17-130.01, p = 0.04; CRTC3-rs12915189 non GG, OR 9.73, CI 1.58-60.10, p = 0.01; GSTP1-rs1695 non AA, OR 9.47, CI 2.06-43.49, p = < 0.01; CAT-rs1001179 non CC, OR 9.18, CI 1.64-51.24, p = 0.01). Total serum chromium levels > 0.1 μg/L were correlated with 73% (52/71) of lung cancers diagnosed with stage I disease. Our findings support the role of chromium and the influence of key proteins on lung cancer burden in the general population.
Collapse
Affiliation(s)
- Piotr Baszuk
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252, Szczecin, Poland
| | - Beata Janasik
- Biological and Environment Monitoring Department, Nofer Institute of Occupational Medicine, ul.św. Teresy od dzieciątka Jezus 8, 91-348, Łódź, Poland
| | - Sandra Pietrzak
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252, Szczecin, Poland
| | - Wojciech Marciniak
- Read-Gene, Grzepnica, ul. Alabastrowa 8, 72-003, Grzepnica, Dobra(Szczecińska), Poland
| | - Edyta Reszka
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, ul.św. Teresy od dzieciątka Jezus 8, 91-348, Łódź, Poland
| | - Katarzyna Białkowska
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252, Szczecin, Poland
| | - Ewa Jabłońska
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, ul.św. Teresy od dzieciątka Jezus 8, 91-348, Łódź, Poland
| | - Magdalena Muszyńska
- Read-Gene, Grzepnica, ul. Alabastrowa 8, 72-003, Grzepnica, Dobra(Szczecińska), Poland
| | - Monika Lesicka
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, ul.św. Teresy od dzieciątka Jezus 8, 91-348, Łódź, Poland
| | - Róża Derkacz
- Read-Gene, Grzepnica, ul. Alabastrowa 8, 72-003, Grzepnica, Dobra(Szczecińska), Poland
| | - Tomasz Grodzki
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891, Szczecin, Poland
| | - Janusz Wójcik
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891, Szczecin, Poland
| | - Małgorzata Wojtyś
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891, Szczecin, Poland
| | - Tadeusz Dębniak
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252, Szczecin, Poland
| | - Cezary Cybulski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252, Szczecin, Poland
| | - Jacek Gronwald
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252, Szczecin, Poland
| | - Bartosz Kubisa
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891, Szczecin, Poland
| | - Norbert Wójcik
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891, Szczecin, Poland
| | - Jarosław Pieróg
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891, Szczecin, Poland
| | - Darko Gajić
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891, Szczecin, Poland
| | - Piotr Waloszczyk
- Independent Laboratory of Pathology, Zdunomed, ul. Energetyków 2, 70-656, Szczecin, Poland
| | - Rodney J Scott
- Priority Research Centre for Cancer Research, Innovation and Translation, Hunter Medical Research Institute, New Lambton Heights, Australia
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, Australia
- Division of Molecular Medicine, Pathology North, John Hunter Hospital, New Lambton, NSW, 2305, Australia
| | - Wojciech Wąsowicz
- Biological and Environment Monitoring Department, Nofer Institute of Occupational Medicine, ul.św. Teresy od dzieciątka Jezus 8, 91-348, Łódź, Poland
| | - Anna Jakubowska
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252, Szczecin, Poland
- Read-Gene, Grzepnica, ul. Alabastrowa 8, 72-003, Grzepnica, Dobra(Szczecińska), Poland
| | - Jan Lubiński
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252, Szczecin, Poland
- Read-Gene, Grzepnica, ul. Alabastrowa 8, 72-003, Grzepnica, Dobra(Szczecińska), Poland
| | - Marcin R Lener
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252, Szczecin, Poland.
| |
Collapse
|
12
|
Lener MR, Reszka E, Marciniak W, Lesicka M, Baszuk P, Jabłońska E, Białkowska K, Muszyńska M, Pietrzak S, Derkacz R, Grodzki T, Wójcik J, Wojtyś M, Dębniak T, Cybulski C, Gronwald J, Kubisa B, Pieróg J, Waloszczyk P, Scott RJ, Jakubowska A, Narod SA, Lubiński J. Blood cadmium levels as a marker for early lung cancer detection. J Trace Elem Med Biol 2021; 64:126682. [PMID: 33249371 DOI: 10.1016/j.jtemb.2020.126682] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/16/2020] [Accepted: 11/06/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND We assessed whether blood cadmium levels were associated with incident lung cancer and could be used in the context of a screening program for early-stage lung cancer. MATERIAL AND METHODS We measured blood cadmium levels among 205 lung cancer patients and 205 matched controls. Cases and controls were matched for sex, age and smoking history (total pack-years, years since cessation for former smokers). RESULTS The odds ratio for those in the highest quartile of cadmium level (versus lowest) was four-fold (OR = 4.41, 95 % CI:2.01-9.67, p < 0.01). The association was present in former smokers (OR = 16.8, 95 % CI:3.96-71.2, p < 0.01), but not in current smokers (OR = 1.23, 95 % CI: 0.34-4.38) or in never smokers (OR not defined). Among former smokers, the association was present in both early- and late-stage lung cancer. CONCLUSION Blood cadmium levels may be a marker to help with the early detection of lung cancer among former smokers.
Collapse
Affiliation(s)
- Marcin R Lener
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | - Edyta Reszka
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, ul.św. Teresy od Dzieciątka Jezus 8, 91-348 Łódź, Poland
| | - Wojciech Marciniak
- Read-Gene, Grzepnica, ul. Alabastrowa 8, 72-003 Dobra (Szczecińska), Poland
| | - Monika Lesicka
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, ul.św. Teresy od Dzieciątka Jezus 8, 91-348 Łódź, Poland
| | - Piotr Baszuk
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | - Ewa Jabłońska
- Department of Molecular Genetics and Epigenetics, Nofer Institute of Occupational Medicine, ul.św. Teresy od Dzieciątka Jezus 8, 91-348 Łódź, Poland
| | - Katarzyna Białkowska
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | | | - Sandra Pietrzak
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | - Róża Derkacz
- Read-Gene, Grzepnica, ul. Alabastrowa 8, 72-003 Dobra (Szczecińska), Poland
| | - Tomasz Grodzki
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891 Szczecin, Poland
| | - Janusz Wójcik
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891 Szczecin, Poland
| | - Małgorzata Wojtyś
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891 Szczecin, Poland
| | - Tadeusz Dębniak
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | - Cezary Cybulski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252 Szczecin, Poland; Read-Gene, Grzepnica, ul. Alabastrowa 8, 72-003 Dobra (Szczecińska), Poland
| | - Jacek Gronwald
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252 Szczecin, Poland; Read-Gene, Grzepnica, ul. Alabastrowa 8, 72-003 Dobra (Szczecińska), Poland
| | - Bartosz Kubisa
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891 Szczecin, Poland
| | - Jarosław Pieróg
- Department of Thoracic Surgery and Transplantation, Pomeranian Medical University in Szczecin, ul. A. Sokołowskiego 11, 70-891 Szczecin, Poland
| | - Piotr Waloszczyk
- Independent Laboratory of Pathology, Zdunomed, ul. Energetyków 2, 70-656 Szczecin, Poland
| | - Rodney J Scott
- Medical Genetics, Hunter Medical Research Institute, Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Pathology North, John Hunter Hospital, Cnr King and Auckland Streets, Newcastle NSW 2300 Australia
| | - Anna Jakubowska
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | - Steven A Narod
- Women's College Research Institute, Toronto, Ontario, Canada; Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Jan Lubiński
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, ul. Unii Lubelskiej 1, 71-252 Szczecin, Poland; Read-Gene, Grzepnica, ul. Alabastrowa 8, 72-003 Dobra (Szczecińska), Poland.
| |
Collapse
|
13
|
Farooq M, Herman JG. Noninvasive Diagnostics for Early Detection of Lung Cancer: Challenges and Potential with a Focus on Changes in DNA Methylation. Cancer Epidemiol Biomarkers Prev 2020; 29:2416-2422. [PMID: 33148791 PMCID: PMC11559093 DOI: 10.1158/1055-9965.epi-20-0704] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/20/2020] [Accepted: 10/13/2020] [Indexed: 11/16/2022] Open
Abstract
Lung cancer remains the leading cause of cancer deaths in the United States and the world. Early detection of this disease can reduce mortality, as demonstrated for low-dose computed tomography (LDCT) screening. However, there remains a need for improvements in lung cancer detection to complement LDCT screening and to increase adoption of screening. Molecular changes in the tumor, and the patient's response to the presence of the tumor, have been examined as potential biomarkers for diagnosing lung cancer. There are significant challenges to developing an effective biomarker with sufficient sensitivity and specificity for the early detection of lung cancer, particularly the detection of circulating tumor DNA, which is present in very small quantities. We will review approaches to develop biomarkers for the early detection of lung cancer, with special consideration to detection of rare tumor events, focus on the use of DNA methylation-based detection in plasma and sputum, and discuss the promise and challenges of lung cancer early detection. Plasma-based detection of lung cancer DNA methylation may provide a simple cost-effective method for the early detection of lung cancer.See all articles in this CEBP Focus section, "NCI Early Detection Research Network: Making Cancer Detection Possible."
Collapse
Affiliation(s)
- Maria Farooq
- Department of Medicine, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - James G Herman
- Department of Medicine, The University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
- UPMC Hillman Comprehensive Cancer Center, Pittsburgh, Pennsylvania
| |
Collapse
|
14
|
Liu B, Ricarte Filho J, Mallisetty A, Villani C, Kottorou A, Rodgers K, Chen C, Ito T, Holmes K, Gastala N, Valyi-Nagy K, David O, Gaba RC, Ascoli C, Pasquinelli M, Feldman LE, Massad MG, Wang TH, Jusue-Torres I, Benedetti E, Winn RA, Brock MV, Herman JG, Hulbert A. Detection of Promoter DNA Methylation in Urine and Plasma Aids the Detection of Non-Small Cell Lung Cancer. Clin Cancer Res 2020; 26:4339-4348. [PMID: 32430478 PMCID: PMC7442601 DOI: 10.1158/1078-0432.ccr-19-2896] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/16/2020] [Accepted: 05/14/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Low-dose CT screening can reduce lung cancer-related mortality. However, CT screening has an FDR of nearly 96%. We sought to assess whether urine samples can be a source for DNA methylation-based detection of non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN This nested case-control study of subjects with suspicious nodules on CT imaging obtained plasma and urine samples preoperatively. Cases (n = 74) had pathologic confirmation of NSCLC. Controls (n = 27) had a noncancer diagnosis. We detected promoter methylation in plasma and urine samples using methylation on beads and quantitative methylation-specific real-time PCR for cancer-specific genes (CDO1, TAC1, HOXA7, HOXA9, SOX17, and ZFP42). RESULTS DNA methylation at cancer-specific loci was detected in both plasma and urine, and was more frequent in patients with cancer compared with controls for all six genes in plasma and in CDO1, TAC1, HOXA9, and SOX17 in urine. Univariate and multivariate logistic regression analysis showed that methylation detection in each one of six genes in plasma and CDO1, TAC1, HOXA9, and SOX17 in urine were significantly associated with the diagnosis of NSCLC, independent of age, race, and smoking pack-years. When methylation was detected for three or more genes in both plasma and urine, the sensitivity and specificity for lung cancer diagnosis were 73% and 92%, respectively. CONCLUSIONS DNA methylation-based biomarkers in plasma and urine could be useful as an adjunct to CT screening to guide decision-making regarding further invasive procedures in patients with pulmonary nodules.
Collapse
Affiliation(s)
- Bin Liu
- Cancer Center, University of Illinois at Chicago, Chicago, Illinois
| | | | - Apurva Mallisetty
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Cassandra Villani
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Anastasia Kottorou
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Clinical and Molecular Oncology Laboratory, Medical School, University of Patras, Patras, Greece
| | - Kristen Rodgers
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chen Chen
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Tomoaki Ito
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Surgery, Juntendo University Shizuoka Hospital, Juntendo University School of Medicine, Shizuoka, Japan
| | - Kyla Holmes
- Cancer Center, University of Illinois at Chicago, Chicago, Illinois
| | - Nicole Gastala
- Department of Family Medicine, Mile Square Health Center, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Klara Valyi-Nagy
- Department of Pathology, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Odile David
- Department of Pathology, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Ron C Gaba
- Department of Radiology, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Christian Ascoli
- Department of Pulmonary, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Mary Pasquinelli
- Department of Pulmonary, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Lawrence E Feldman
- Department of Hematology and Oncology, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Malek G Massad
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Tza-Huei Wang
- Department of Biomedical Engineering and Institute for Nano Biotechnology, The Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - Ignacio Jusue-Torres
- Department of Neurological Surgery, Loyola University Stritch School of Medicine, Maywood, Illinois
| | - Enrico Benedetti
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Robert A Winn
- Cancer Center, University of Illinois at Chicago, Chicago, Illinois
- Department of Pulmonary, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| | - Malcolm V Brock
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Cancer Center, Department of Oncology, The Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - James G Herman
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Alicia Hulbert
- Cancer Center, University of Illinois at Chicago, Chicago, Illinois.
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, Illinois
| |
Collapse
|
15
|
Epigenetic Silencing of LMX1A Contributes to Cancer Progression in Lung Cancer Cells. Int J Mol Sci 2020; 21:ijms21155425. [PMID: 32751497 PMCID: PMC7432919 DOI: 10.3390/ijms21155425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/16/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022] Open
Abstract
Epigenetic modification is considered a major mechanism of the inactivation of tumor suppressor genes that finally contributes to carcinogenesis. LIM homeobox transcription factor 1α (LMX1A) is one of the LIM-homeobox-containing genes that is a critical regulator of growth and differentiation. Recently, LMX1A was shown to be hypermethylated and functioned as a tumor suppressor in cervical cancer, ovarian cancer, and gastric cancer. However, its role in lung cancer has not yet been clarified. In this study, we used public databases, methylation-specific PCR (MSP), reverse transcription PCR (RT-PCR), and bisulfite genomic sequencing to show that LMX1A was downregulated or silenced due to promoter hypermethylation in lung cancers. Treatment of lung cancer cells with the demethylating agent 5-aza-2'-deoxycytidine restored LMX1A expression. In the lung cancer cell lines H23 and H1299, overexpression of LMX1A did not affect cell proliferation but suppressed colony formation and invasion. These suppressive effects were reversed after inhibition of LMX1A expression in an inducible expression system in H23 cells. The quantitative RT-PCR (qRT-PCR) data showed that LMX1A could modulate epithelial mesenchymal transition (EMT) through E-cadherin (CDH1) and fibronectin (FN1). NanoString gene expression analysis revealed that all aberrantly expressed genes were associated with processes related to cancer progression, including angiogenesis, extracellular matrix (ECM) remodeling, EMT, cancer metastasis, and hypoxia-related gene expression. Taken together, these data demonstrated that LMX1A is inactivated through promoter hypermethylation and functions as a tumor suppressor. Furthermore, LMX1A inhibits non-small cell lung cancer (NSCLC) cell invasion partly through modulation of EMT, angiogenesis, and ECM remodeling.
Collapse
|
16
|
Roncarati R, Lupini L, Miotto E, Saccenti E, Mascetti S, Morandi L, Bassi C, Rasio D, Callegari E, Conti V, Rinaldi R, Lanza G, Gafà R, Papi A, Frassoldati A, Sabbioni S, Ravenna F, Casoni GL, Negrini M. Molecular testing on bronchial washings for the diagnosis and predictive assessment of lung cancer. Mol Oncol 2020; 14:2163-2175. [PMID: 32441866 PMCID: PMC7463327 DOI: 10.1002/1878-0261.12713] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/04/2020] [Accepted: 05/15/2020] [Indexed: 12/17/2022] Open
Abstract
Cytopathological analyses of bronchial washings (BWs) collected during fibre‐optic bronchoscopy are often inconclusive for lung cancer diagnosis. To address this issue, we assessed the suitability of conducting molecular analyses on BWs, with the aim to improve the diagnosis and outcome prediction of lung cancer. The methylation status of RASSF1A, CDH1, DLC1 and PRPH was analysed in BW samples from 91 lung cancer patients and 31 controls, using a novel two‐colour droplet digital methylation‐specific PCR (ddMSP) technique. Mutations in ALK, BRAF, EGFR, ERBB2, KRAS, MAP2K1, MET, NRAS, PIK3CA, ROS1 and TP53 and gene fusions of ALK, RET and ROS1 were also investigated, using next‐generation sequencing on 73 lung cancer patients and 14 tumour‐free individuals. Our four‐gene methylation panel had significant diagnostic power, with 97% sensitivity and 74% specificity (relative risk, 7.3; odds ratio, 6.1; 95% confidence interval, 12.7–127). In contrast, gene mutation analysis had a remarkable value for predictive, but not for diagnostic, purposes. Actionable mutations in EGFR, HER2 and ROS1 as well as in other cancer genes (KRAS, PIK3CA and TP53) were detected. Concordance with gene mutations uncovered in tumour biopsies was higher than 90%. In addition, bronchial‐washing analyses permitted complete patient coverage and the detection of additional actionable mutations. In conclusion, BWs are a useful material on which to perform molecular tests based on gene panels: aberrant gene methylation and mutation analyses could be performed as approaches accompanying current diagnostic and predictive assays during the initial workup phase. This study establishes the grounds for further prospective investigation.
Collapse
Affiliation(s)
- Roberta Roncarati
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy.,CNR, Institute of Genetics and Biomedical Research, National Research Council of Italy, Milano, Italy
| | - Laura Lupini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy
| | - Elena Miotto
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy
| | - Elena Saccenti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy
| | - Susanna Mascetti
- Azienda Ospedaliero-Universitaria di Ferrara, Division of Respiratory Endoscopy, S. Anna Hospital, Cona, Italy
| | - Luca Morandi
- Azienda Ospedaliero-Universitaria di Ferrara, Division of Respiratory Endoscopy, S. Anna Hospital, Cona, Italy
| | - Cristian Bassi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy.,Laboratorio per le Tecnologie delle Terapie Avanzate, Tecnopolo, University of Ferrara, Italy
| | - Debora Rasio
- Department of Clinical and Molecular Medicine, Sant' Andrea Hospital, University "La Sapienza", Rome, Italy
| | - Elisa Callegari
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy
| | - Valentina Conti
- Pneumology Division, State Hospital, San Marino, Republic of San Marino
| | - Rosa Rinaldi
- Division of Anatomic Pathology, Carlo Poma Hospital, Mantova, Italy
| | - Giovanni Lanza
- Azienda Ospedaliero-Universitaria di Ferrara, Division of Anatomic Pathology, S. Anna Hospital, Cona, Italy.,Department of Medical Sciences, University of Ferrara, Italy
| | - Roberta Gafà
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy.,Azienda Ospedaliero-Universitaria di Ferrara, Division of Anatomic Pathology, S. Anna Hospital, Cona, Italy
| | - Alberto Papi
- Azienda Ospedaliero-Universitaria di Ferrara, Division of Respiratory Endoscopy, S. Anna Hospital, Cona, Italy.,Department of Medical Sciences, University of Ferrara, Italy
| | - Antonio Frassoldati
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy.,Azienda Ospedaliero-Universitaria di Ferrara, Medical Oncology Unit, S. Anna Hospital, Cona, Italy
| | - Silvia Sabbioni
- Laboratorio per le Tecnologie delle Terapie Avanzate, Tecnopolo, University of Ferrara, Italy.,Department of Life Sciences and Biotechnology, University of Ferrara, Italy
| | - Franco Ravenna
- Division of Pneumology and Intensive Respiratory Unit, Carlo Poma Hospital, Mantova, Italy
| | - Gian L Casoni
- Azienda Ospedaliero-Universitaria di Ferrara, Division of Respiratory Endoscopy, S. Anna Hospital, Cona, Italy
| | - Massimo Negrini
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy.,Laboratorio per le Tecnologie delle Terapie Avanzate, Tecnopolo, University of Ferrara, Italy
| |
Collapse
|
17
|
Tessema M, Tassew DD, Yingling CM, Do K, Picchi MA, Wu G, Petersen H, Randell S, Lin Y, Belinsky SA, Tesfaigzi Y. Identification of novel epigenetic abnormalities as sputum biomarkers for lung cancer risk among smokers and COPD patients. Lung Cancer 2020; 146:189-196. [PMID: 32559455 DOI: 10.1016/j.lungcan.2020.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/09/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Smoking is a common risk factor for chronic obstructive pulmonary disease (COPD) and lung cancer. Although COPD patients have higher risk of lung cancer compared to non-COPD smokers, the molecular links between these diseases are not well-defined. This study aims to identify genes that are downregulated by cigarette smoke and commonly repressed in COPD and lung cancer. MATERIALS AND METHODS Primary human airway epithelial cells (HAEC) were exposed to cigarette-smoke-extract (CSE) for 10-weeks and significantly suppressed genes were identified by transcriptome array. Epigenetic abnormalities of these genes in lung adenocarcinoma (LUAD) from patients with or without COPD were determined using genome-wide and gene-specific assays and by in vitro treatment of cell lines with trichostatin-A or 5-aza-2-deoxycytidine. RESULTS The ten most commonly downregulated genes following chronic CSE exposure of HAEC and show promoter hypermethylation in LUAD were selected. Among these, expression of CCNA1, SNCA, and ZNF549 was significantly reduced in lung tissues from COPD compared with non-COPD cases while expression of CCNA1 and SNCA was further downregulated in tumors with COPD. The promoter regions of all three genes were hypermethylated in LUAD but not normal or COPD lungs. The reduced expression and aberrant promoter hypermethylation of these genes in LUAD were independently validated using data from the Cancer Genome Atlas project. Importantly, SNCA and ZNF549 methylation detected in sputum DNA from LUAD (52% and 38%) cases were more prevalent compared to cancer-free smokers (26% and 15%), respectively (p < 0.02). CONCLUSIONS Our data show that suppression of CCNA1, SNCA, and ZNF549 in lung cancer and COPD occurs with or without promoter hypermethylation, respectively. Detecting methylation of these and previously identified genes in sputum of cancer-free smokers may serve as non-invasive biomarkers for early detection of lung cancer among high risk smokers including COPD patients.
Collapse
Affiliation(s)
- Mathewos Tessema
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, M, USA.
| | - Dereje D Tassew
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA; Currently, Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Christin M Yingling
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, M, USA
| | - Kieu Do
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, M, USA
| | - Maria A Picchi
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, M, USA
| | - Guodong Wu
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, M, USA
| | - Hans Petersen
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Scott Randell
- Department of Cell and Molecular Physiology, The University of North Carolina, Chapel Hill, NC, USA
| | - Yong Lin
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, M, USA
| | - Steven A Belinsky
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, M, USA
| | - Yohannes Tesfaigzi
- COPD Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA; Currently, Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
18
|
Septin 9 Methylation in Nasopharyngeal Swabs: A Potential Minimally Invasive Biomarker for the Early Detection of Nasopharyngeal Carcinoma. DISEASE MARKERS 2020; 2020:7253531. [PMID: 32454907 PMCID: PMC7232724 DOI: 10.1155/2020/7253531] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 01/17/2020] [Accepted: 03/28/2020] [Indexed: 02/06/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is highly prevalent in Southeast Asia, and an unfavorable outcome is usually attributed to advanced stage NPC. Current methods for the early diagnosis of NPC have limitations in clinical practice. The aim of this study was to investigate the diagnostic ability of Septin 9 methylation for NPC. A quantitative methylation-sensitive PCR (qMS-PCR) assay was developed to measure the methylation status and levels of Septin 9 in nasopharyngeal tissues and paired swabs from patients with NPC, chronic nasopharyngitis, and healthy donors. Methylated Septin 9 was detected in 92% (23/25) of NPC tissues and 25% (4/16) of nasopharyngitis controls (p < 0.05). High-frequency hypermethylation with decreased mRNA expression of Septin 9 in NPC was also identified. Further, Septin 9 methylation was identified in 90.5% (19/21) of NPC biopsies and 71.4% (15/21) of paired swabs, indicating a good concordance between the two sample types. In addition, methylated Septin 9 was found in 16 (72.7%) nasal swabs from 22 NPC patients, 2 of 19 (10.5%) nasopharyngitis, but not in any of the healthy controls (p < 0.01). The methylation score in nasal swabs of the NPC group was also significantly higher than that of non-NPC controls (p < 0.001). Moreover, receiver operating characteristic (ROC) curve analysis showed an area under the curve (AUC) of 0.882 of Septin 9 methylation tests to discriminate NPC from non-NPC subjects. Our study demonstrated that frequent methylation of Septin 9 was present in NPC. Its detection in nasopharyngeal swabs may provide a minimally invasive and informative method for identifying early NPC cases.
Collapse
|
19
|
Gupta C, Su J, Zhan M, Stass SA, Jiang F. Sputum long non-coding RNA biomarkers for diagnosis of lung cancer. Cancer Biomark 2020; 26:219-227. [PMID: 31450489 DOI: 10.3233/cbm-190161] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Analysis of molecular changes in sputum may help diagnose lung cancer. Long non-coding RNAs (lncRNAs) play vital roles in various biological processes, and their dysregulations contribute to the development and progression of lung tumorigenesis. Herein, we determine whether aberrant lncRNAs could be used as potential sputum biomarkers for lung cancer. METHODS Using reverse transcription PCR, we measure expressions of lung cancer-associated lncRNAs in sputum of a discovery cohort of 67 lung cancer patients and 65 cancer-free smokers with benign diseases and a validation cohort of 59 lung cancer patients and 60 cancer-free smokers with benign diseases. RESULTS In the discovery cohort, four of the lncRNAs displayed a significantly different level in sputum of lung cancer patients vs.cancer-free smokers with benign diseases (all P< 0.001). From the four lncRNAs, three lncRNAs (SNHG1, H19, and HOTAIR) are identified as a biomarker panel, producing 82.09% sensitivity and 89.23% specificity for diagnosis of lung cancer. Furthermore, the biomarker panel has a higher sensitivity (82.09% vs. 52.24%, P= 0.02) and a similar specificity compared with sputum cytology (89.23% vs. 90.77%, P= 0.45). In addition, the lncRNA biomarker panel had a higher sensitivity (87.50% vs. 70.07%, p= 0.03) for diagnosis of squamous cell carcinoma compared with adenocarcinoma of the lung, while maintaining the same specificity (89.23%). The potential of the sputum lncRNA biomarkers for lung cancer detection is confirmed in the validation cohort. CONCLUSION We have for the first time shown that the analysis of lncRNAs in sputum might be a noninvasive approach for diagnosis of lung cancer.
Collapse
Affiliation(s)
- Chhavi Gupta
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jian Su
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Min Zhan
- Departments of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sanford A Stass
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Feng Jiang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
20
|
Sharma M, Surani S. Exploring Novel Technologies in Lung Cancer Diagnosis: Do We Have Room for Improvement? Cureus 2020; 12:e6828. [PMID: 32181072 PMCID: PMC7051117 DOI: 10.7759/cureus.6828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related death worldwide. Preventive strategies, mainly smoking cessation have a big impact on the reduction of lung cancer-related mortality. Screening with low dose computed tomography (LDCT) has proven to be beneficial in reducing the mortality related to lung cancer mainly based on early detection of cancer and timely initiation of treatment. Despite its beneficial effects, guideline-directed LDCT screening could lead to high false positive results, subjecting patients to harmful radiation, increase cost of healthcare and induce anxiety amongst the patients. Thus, it is imperative to look beyond the prevailing modalities of lung cancer screening and diagnosis to achieve better yield and mitigate the existent drawbacks.
Collapse
Affiliation(s)
- Munish Sharma
- Internal Medicine, Corpus Christi Medical Center, Corpus Christi, USA
| | - Salim Surani
- Internal Medicine, Texas A&M Health Science Center, Bryan, USA
| |
Collapse
|
21
|
Liao J, Shen J, Leng Q, Qin M, Zhan M, Jiang F. MicroRNA-based biomarkers for diagnosis of non-small cell lung cancer (NSCLC). Thorac Cancer 2020; 11:762-768. [PMID: 31994346 PMCID: PMC7049510 DOI: 10.1111/1759-7714.13337] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 01/09/2020] [Accepted: 01/12/2020] [Indexed: 11/27/2022] Open
Abstract
Background The development of biomarkers for the early detection of non‐small cell lung cancer (NSCLC) is clinically important. We have developed miRNA biomarkers in sputum and plasma, respectively, for NSCLC. Herein, we evaluate whether integrated analysis of the miRNAs across the different types of specimens could improve the early detection of NSCLC. Methods Using reverse transcription PCR, we determined expressions of two miRNAs (miRs‐31‐5p and 210‐3p) in sputum and three miRNAs (miRs‐21‐5p, 210‐3p, and 486‐5p) in plasma of a training cohort of 76 NSCLC patients and 72 cancer‐free smokers. The results were validated in a testing cohort of 56 NSCLC patients and 55 cancer‐free smokers. Results The panels of two sputum miRNAs and three plasma miRNAs had 65.8–75.0% sensitivities and 83.3–87.5% specificities for diagnosis of NSCLC in the training cohort. The individual sputum or plasma miRNA panel had a higher sensitivity for squamous cell carcinoma or adenocarcinoma of the lung, respectively. From the miRNAs, we optimized an integrated panel of biomarkers consisting of two sputum miRNAs (miRs‐31‐5p and 210‐3p) and one plasma miRNA (miR‐21‐5p) that had higher sensitivity (85.5%) and specificity (91.7%) for diagnosis of NSCLC compared with the individual panels alone. Furthermore, the performance of the integrated panel of biomarkers was independent of histology and stage of NSCLC, and patients' age, sex, and ethnicity. The performance of the integrated panel of biomarkers was confirmed in the testing cohort. Conclusions Integrating biomarkers across different body fluids would synergistically improve the early detection of NSCLC. Key points Lung cancer is a heterogeneous disease and develops from complex aberrations. Integrating sputum and plasma miRNAs has higher accuracy than when they are used alone
Collapse
Affiliation(s)
- Jipei Liao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jun Shen
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Qixin Leng
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Meng Qin
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Min Zhan
- Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Feng Jiang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
22
|
Abstract
Despite the introduction of low-dose computed tomography (LDCT) and implementation of lung cancer screening programs, lung cancer still maintains the leading cause of cancer-specific death all around the world in terms of morbidity and mortality. Many studies demonstrated that the methylation status of selected genes may act as prognostic biomarkers for lung cancer patients. Recently, the development of high-throughput sequencing for methylation would help researchers better understand the role of methylation in the tumorigenicity or metastasis of lung cancer. This chapter reviews the progress of DNA methylation in lung cancer.
Collapse
|
23
|
Li P, Gan Y, Qin S, Han X, Cui C, Liu Z, Zhou J, Gu L, Lu ZM, Zhang B, Deng D. DNA hydroxymethylation increases the susceptibility of reactivation of methylated P16 alleles in cancer cells. Epigenetics 2019; 15:618-631. [PMID: 31790633 DOI: 10.1080/15592294.2019.1700004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
It is well established that 5-methylcytosine (5mC) in genomic DNA of mammalian cells can be oxidized into 5-hydroxymethylcytosine (5hmC) and other derivates by DNA dioxygenase TETs. While conversion of 5mC to 5hmC plays an important role in active DNA demethylation through further oxidation steps, a certain proportion of 5hmCs remain in the genome. Although 5hmCs contribute to the flexibility of chromatin and protect bivalent promoters from hypermethylation, the direct effect of 5hmCs on gene transcription is unknown. In this present study, we have engineered a zinc-finger protein-based P16-specific DNA dioxygenase (P16-TET) to induce P16 hydroxymethylation and demethylation in cancer cells. Our results demonstrate, for the first time, that although the hydroxymethylated P16 alleles retain transcriptionally inactive, hydroxymethylation could increase the susceptibility of reactivation of methylated P16 alleles.
Collapse
Affiliation(s)
- Paiyun Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| | - Ying Gan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| | - Sisi Qin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| | - Xiao Han
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| | - Chenghua Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| | - Zhaojun Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| | - Jing Zhou
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| | - Liankun Gu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| | - Zhe-Ming Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| | - Baozhen Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| | - Dajun Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute , Beijing, China
| |
Collapse
|
24
|
Wu W, Wang Z, Zhou Z. Role of the human papillomavirus in malignant transformation of oral leukoplakia distinct from oropharyngeal squamous cell carcinoma: A study of 76 patients with internal-control specimens. Oral Surg Oral Med Oral Pathol Oral Radiol 2019; 128:273-279. [DOI: 10.1016/j.oooo.2019.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 12/22/2018] [Accepted: 01/03/2019] [Indexed: 10/27/2022]
|
25
|
Impact of catechol-O-methyltransferase gene variants on methylation status of P16 and MGMT genes and their downregulation in colorectal cancer. Eur J Cancer Prev 2019; 28:68-75. [PMID: 30379684 DOI: 10.1097/cej.0000000000000485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Globally, colorectal cancer (CRC) is the third most commonly diagnosed cancer in males and the second most commonly diagnosed cancer in females, with 1.4 million new cases and almost 694 000 deaths estimated to have occurred in 2012. The development and progression of CRC is dictated by a series of alterations in diverse genes mostly proto-oncogenes and tumor suppressor genes. In this dreadful disease disturbances different from mutations called as epigenetic regulations are also taken into consideration and are thoroughly investigated. The present study was designed to analyze the promoter hypermethylation of CpG (cytosine, followed by guanine nucleotide) islands of cyclin-dependent kinase inhibitor 2A (P16) and O-methylguanine-DNA methyltransferase (MGMT) genes and its subsequent effect on the protein expression in CRC. The impact of the common functional polymorphism of the catechol-O-methyltransferase (COMT) gene, Val158Met, on promoter hypermethylation of P16 and MGMT genes in CRC was also investigated. The study included 200 CRC cases and equal numbers of normal samples. DNA was extracted using the kit method and methylation specific-PCR was performed for analysis of the promoter hypermethylation status. Total protein was isolated form all CRC cases and western blotting was performed for P16 and MGMT proteins. The COMT Val158Met polymorphism was analyzed by a PCR-restriction fragment length polymorphism assay. Epigenetic analysis showed that unlike other high-risk regions, the Kashmiri population has a different promoter hypermethylation profile of both P16 and MGMT genes, with frequent and significant promoter hypermethylation of both in CRC. The frequency of promoter hypermethylation of both genes was significantly higher in males and was insignificantly found to be higher in stage III/IV. The degree of P16 and MGMT promoter hypermethylation increased significantly with increasing severity of the lesion. We also found a significant correlation between P16 and MGMT promoter hypermethylation and loss of protein expression in CRC. A significant association was found between COMT polymorphism (homozygous variant) and P16 methylation status. Similar results were also found for MGMT hypermethylated cases.
Collapse
|
26
|
Shankar A, Saini D, Dubey A, Roy S, Bharati SJ, Singh N, Khanna M, Prasad CP, Singh M, Kumar S, Sirohi B, Seth T, Rinki M, Mohan A, Guleria R, Rath GK. Feasibility of lung cancer screening in developing countries: challenges, opportunities and way forward. Transl Lung Cancer Res 2019; 8:S106-S121. [PMID: 31211111 DOI: 10.21037/tlcr.2019.03.03] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lung cancer is the leading cause of all cancer deaths worldwide, comprising 18.4% of all cancer deaths. Low-dose computed tomography (LDCT) has shown mortality benefit in various trials and now a standard tool for lung cancer screening. Most researches have been carried out in developed countries where lung cancer incidence and mortality is very high. There is an increasing trend in lung cancer incidence in developing countries attributed to tobacco smoking and various environmental and occupational risk factors. Implementation of lung cancer screening is challenging, so organised lung cancer screening is practically non-existent. There are numerous challenges in implementing such programs ranging from infrastructure, trained human resources, referral algorithm to cost and psychological trauma due to over-diagnosis. Pulmonary tuberculosis and other chest infections are important issues to be addressed while planning for lung cancer screening in developing countries. Burden of these diseases is very high and can lead to over-diagnosis in view of cut off of lung nodule size in various studies. Assessment of high risk cases for lung cancer is difficult as various forms of smoking make quantification non-uniform and difficult. Lung cancer screening targets only high risk population unlike screening programs for other cancers where entire population is targeted. There is a need of lung cancer screening for high risk cases as it saves life. Tobacco control and smoking cessation remain the most important long term intervention to decrease morbidity and mortality from lung cancer in developing countries. There is no sufficient evidence supporting the introduction of population-based screening for lung cancer in public health services.
Collapse
Affiliation(s)
- Abhishek Shankar
- Preventive Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| | - Deepak Saini
- Indian Society of Clinical Oncology, Delhi, India
| | - Anusha Dubey
- Indian Society of Clinical Oncology, Delhi, India
| | - Shubham Roy
- Indian Society of Clinical Oncology, Delhi, India
| | - Sachidanand Jee Bharati
- Oncoanaesthesia and Palliative Medicine, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| | - Navneet Singh
- Pulmonary Medicine, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | | | - Chandra Prakash Prasad
- Medical Oncology (Lab), Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| | - Mayank Singh
- Medical Oncology (Lab), Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| | - Sunil Kumar
- Surgical Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| | - Bhawna Sirohi
- Medical Oncology, Max Institute of Cancer Care, Delhi, India
| | - Tulika Seth
- Clinical Hematology, All India Institute of Medical Sciences, Delhi, India
| | - Minakshi Rinki
- Biotechnology, Swami Shraddhanand College, Delhi University, Delhi, India
| | - Anant Mohan
- Pulmonary Medicine & Sleep Disorders, All India Institute of Medical Sciences, Delhi, India
| | - Randeep Guleria
- Pulmonary Medicine & Sleep Disorders, All India Institute of Medical Sciences, Delhi, India
| | - Goura Kishor Rath
- Radiation Oncology, Dr BR Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, Delhi, India
| |
Collapse
|
27
|
Shende P, Augustine S, Prabhakar B, Gaud RS. Advanced multimodal diagnostic approaches for detection of lung cancer. Expert Rev Mol Diagn 2019; 19:409-417. [DOI: 10.1080/14737159.2019.1607299] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Pravin Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, Shri Vile Parle Kelavani Mandal’S Narsee Monjee Institute of Management Studies University, Mumbai, India
| | - Steffi Augustine
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, Shri Vile Parle Kelavani Mandal’S Narsee Monjee Institute of Management Studies University, Mumbai, India
| | - Bala Prabhakar
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, Shri Vile Parle Kelavani Mandal’S Narsee Monjee Institute of Management Studies University, Mumbai, India
| | - R. S. Gaud
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, Shri Vile Parle Kelavani Mandal’S Narsee Monjee Institute of Management Studies University, Mumbai, India
| |
Collapse
|
28
|
Sandanger TM, Nøst TH, Guida F, Rylander C, Campanella G, Muller DC, van Dongen J, Boomsma DI, Johansson M, Vineis P, Vermeulen R, Lund E, Chadeau-Hyam M. DNA methylation and associated gene expression in blood prior to lung cancer diagnosis in the Norwegian Women and Cancer cohort. Sci Rep 2018; 8:16714. [PMID: 30425263 PMCID: PMC6233189 DOI: 10.1038/s41598-018-34334-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022] Open
Abstract
The majority of lung cancer is caused by tobacco smoking, and lung cancer-relevant epigenetic markers have been identified in relation to smoking exposure. Still, smoking-related markers appear to mediate little of the effect of smoking on lung cancer. Thus in order to identify disease-relevant markers and enhance our understanding of pathways, a wide search is warranted. Through an epigenome-wide search within a case-control study (131 cases, 129 controls) nested in a Norwegian prospective cohort of women, we found 25 CpG sites associated with lung cancer. Twenty-three were classified as associated with smoking (LC-AwS), and two were classified as unassociated with smoking (LC-non-AwS), as they remained associated with lung cancer after stringent adjustment for smoking exposure using the comprehensive smoking index (CSI): cg10151248 (PC, CSI-adjusted odds ratio (OR) = 0.34 [0.23-0.52] per standard deviation change in methylation) and cg13482620 (B3GNTL1, CSI-adjusted OR = 0.33 [0.22-0.50]). Analysis among never smokers and a cohort of smoking-discordant twins confirmed the classification of the two LC-non-AwS CpG sites. Gene expression profiles demonstrated that the LC-AwS CpG sites had different enriched pathways than LC-non-AwS sites. In conclusion, using blood-derived DNA methylation and gene expression profiles from a prospective lung cancer case-control study in women, we identified 25 CpG lung cancer markers prior to diagnosis, two of which were LC-non-AwS markers and related to distinct pathways.
Collapse
Affiliation(s)
- Torkjel Manning Sandanger
- Department of Community Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway.
| | - Therese Haugdahl Nøst
- Department of Community Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Florence Guida
- MRC/PHE Centre for Environmental Health, Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Genetic Epidemiology Group, International Agency for Research on Cancer, Lyon, France
| | - Charlotta Rylander
- Department of Community Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Gianluca Campanella
- MRC/PHE Centre for Environmental Health, Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - David C Muller
- MRC/PHE Centre for Environmental Health, Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
| | - Jenny van Dongen
- Netherlands Twin Register, Vrije Universiteit, Department of Biological Psychology, Amsterdam, The Netherlands
| | - Dorret I Boomsma
- Netherlands Twin Register, Vrije Universiteit, Department of Biological Psychology, Amsterdam, The Netherlands
| | - Mattias Johansson
- Genetic Epidemiology Group, International Agency for Research on Cancer, Lyon, France
| | - Paolo Vineis
- MRC/PHE Centre for Environmental Health, Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Italian Institute for Genomic Medicine (IIGM), Turin, Italy
| | - Roel Vermeulen
- MRC/PHE Centre for Environmental Health, Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Institute for Risk Assessment Sciences (IRAS), Division of Environmental Epidemiology, Utrecht University, Utrecht, The Netherlands
| | - Eiliv Lund
- Department of Community Medicine, Faculty of Health Sciences, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Marc Chadeau-Hyam
- MRC/PHE Centre for Environmental Health, Department of Epidemiology and Biostatistics, Imperial College London, London, United Kingdom
- Institute for Risk Assessment Sciences (IRAS), Division of Environmental Epidemiology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
29
|
A multi-parameterized artificial neural network for lung cancer risk prediction. PLoS One 2018; 13:e0205264. [PMID: 30356283 PMCID: PMC6200229 DOI: 10.1371/journal.pone.0205264] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 09/21/2018] [Indexed: 01/20/2023] Open
Abstract
The objective of this study is to train and validate a multi-parameterized artificial neural network (ANN) based on personal health information to predict lung cancer risk with high sensitivity and specificity. The 1997-2015 National Health Interview Survey adult data was used to train and validate our ANN, with inputs: gender, age, BMI, diabetes, smoking status, emphysema, asthma, race, Hispanic ethnicity, hypertension, heart diseases, vigorous exercise habits, and history of stroke. We identified 648 cancer and 488,418 non-cancer cases. For the training set the sensitivity was 79.8% (95% CI, 75.9%-83.6%), specificity was 79.9% (79.8%-80.1%), and AUC was 0.86 (0.85-0.88). For the validation set sensitivity was 75.3% (68.9%-81.6%), specificity was 80.6% (80.3%-80.8%), and AUC was 0.86 (0.84-0.89). Our results indicate that the use of an ANN based on personal health information gives high specificity and modest sensitivity for lung cancer detection, offering a cost-effective and non-invasive clinical tool for risk stratification.
Collapse
|
30
|
Liu H, Liu Z, Liu XW, Xu S, Wang L, Liu Y, Zhou J, Gu L, Gao Y, Liu XY, Shi H, Sun Z, Deng D. A similar effect of P16 hydroxymethylation and true-methylation on the prediction of malignant transformation of oral epithelial dysplasia: observation from a prospective study. BMC Cancer 2018; 18:918. [PMID: 30249192 PMCID: PMC6154879 DOI: 10.1186/s12885-018-4787-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 08/31/2018] [Indexed: 01/18/2023] Open
Abstract
Background Total P16 methylation (P16M), including P16 hydroxymethylation (P16H) and true-P16M, correlates with malignant transformation of oral epithelial dysplasia (OED). Both true-P16M and P16H are early events in carcinogenesis. The aim of this study is to prospectively determine if discrimination of true-P16M from P16H is necessary for prediction of cancer development from OEDs. Methods Patients (n = 265) with mild or moderate OED were recruited into the double blind two-center cohort. Total-P16M and P16H were analyzed using the 115-bp MethyLight, TET-assisted bisulfite (TAB) methylation-specific PCR (MSP), and TAB-sequencing. Total-P16M-positive and P16H-negative samples were defined as true-P16M-positive. Progression of OEDs was monitored for a minimum 24 months follow-up period. Results P16H was detected in 23 of 73 (31.5%) total-P16M-positive OEDs. Follow-up information was obtained from 247 patients with an ultimate compliance rate of 93.2%. OED-derived squamous cell carcinomas were observed in 13.0% (32/247) patients during follow-up (median, 41.0 months). The cancer progression rate for total-P16M-positive patients was significantly increased when compared to total-P16M-negative patients [23.3% vs 8.6%; adjusted odds ratio = 2.67 (95% CI: 1.19–5.99)]. However, the cancer progression rates were similar between P16H- and true-P16M-positive OEDs [26.1% (6/23) vs 22.0% (11/50); odds ratio = 0.80 (95% CI: 0.22–2.92)]. The cancer-free survival was also similar for these patients. Conclusion P16H and true-P16M are similar biomarkers for determining malignant potential of OEDs. Discrimination of P16H from true-P16M, at least in OED, may be not necessary in clinical applications. Trial registration This study is registered prospectively in the U.S. National Institutes of Health Clinical Trials Protocol Registration System (trial number NCT02967120, available at https://ClinicalTrials.gov/ct2/show/NCT02967120). Electronic supplementary material The online version of this article (10.1186/s12885-018-4787-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hongwei Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University School of Stomatology, Beijing, 100081, China
| | - Zhaojun Liu
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Aetiology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xue-Wei Liu
- Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Si Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University School of Stomatology, Beijing, 100081, China
| | - Lei Wang
- Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Yang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University School of Stomatology, Beijing, 100081, China
| | - Jing Zhou
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Aetiology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Liankun Gu
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Aetiology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yan Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University School of Stomatology, Beijing, 100081, China
| | - Xiao-Yong Liu
- Capital Medical University School of Stomatology, Beijing, 100050, China
| | - Huidong Shi
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Zheng Sun
- Capital Medical University School of Stomatology, Beijing, 100050, China.
| | - Dajun Deng
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Aetiology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
31
|
Xiong JX, Wang YS, Sheng J, Xiang D, Huang TX, Tan BB, Zeng CM, Li HH, Yang J, Meltzer SJ, Mori Y, Qin YR, Guan XY, Fu L. Epigenetic alterations of a novel antioxidant gene SLC22A3 predispose susceptible individuals to increased risk of esophageal cancer. Int J Biol Sci 2018; 14:1658-1668. [PMID: 30416380 PMCID: PMC6216027 DOI: 10.7150/ijbs.28482] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/24/2018] [Indexed: 01/29/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) occurs with the highest frequency in China, especially in the high-risk Northern Chinese. Recent studies have reported that SLC22A3 is significantly downregulated in non-tumor (NT) esophageal tissues from familial ESCC patients compared with those from sporadic ESCC. However, the mechanism of how SLC22A3 regulates familial ESCC remains unknown. In this study, post hoc genome-wide association studies (GWAS) in 496 cases with a family history of upper gastrointestinal tract cancers and 1056 controls were performed and the results revealed that SLC22A3 is a novel susceptibility gene for familial ESCC. Reduced expression of SLC22A3 in NT esophageal tissues from familial ESCC patients significantly correlates with its promoter hypermethylation. Moreover, case-control study of Chinese descendants from different risk areas of China revealed that the methylation of the SLC22A3 gene in peripheral blood leukocyte (PBL) DNA samples could be a risk factor for developing ESCC in this high-risk population. Functional studies showed that SLC22A3 is a novel antioxidant gene, and deregulation of SLC22A3 facilitates heat stress-induced oxidative DNA damage and formation of γ-H2AX foci in normal esophageal epithelial cells. Collectively, we show that epigenetic alterations of SLC22A3 predispose susceptible individuals to increased risk of esophageal cancer.
Collapse
Affiliation(s)
- Ji-Xian Xiong
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen 518039, China
| | - Yan-Song Wang
- Department of Stomatology, Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Jingyi Sheng
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong
- Shenzhen Huarui Translational Research Institute, Shenzhen, China
| | - Di Xiang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen 518039, China
| | - Tu-Xiong Huang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen 518039, China
| | - Bin-Bin Tan
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen 518039, China
| | - Cui-Mian Zeng
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen 518039, China
| | - Hua-Hui Li
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen 518039, China
| | - Jiao Yang
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen 518039, China
| | - Stephen J. Meltzer
- Department of Medicine and Oncology, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Yuriko Mori
- Department of Medicine and Oncology, The Johns Hopkins University School of Medicine and Sidney Kimmel Comprehensive Cancer Center, Baltimore, Maryland, USA
| | - Yan-Ru Qin
- Department of Clinical Oncology, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, University of Hong Kong, Hong Kong
| | - Li Fu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Carson International Cancer Center, Shenzhen University School of Medicine, Shenzhen 518039, China
| |
Collapse
|
32
|
Leng S, Diergaarde B, Picchi MA, Wilson DO, Gilliland FD, Yuan JM, Siegfried JM, Belinsky SA. Gene Promoter Hypermethylation Detected in Sputum Predicts FEV 1 Decline and All-Cause Mortality in Smokers. Am J Respir Crit Care Med 2018; 198:187-196. [PMID: 29437466 PMCID: PMC6058990 DOI: 10.1164/rccm.201708-1659oc] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 02/12/2018] [Indexed: 02/07/2023] Open
Abstract
RATIONALE Gene promoter hypermethylation detected in sputum assesses the extent of field cancerization and predicts lung cancer (LC) risk in ever-smokers. A rapid decline of FEV1 is a major driver for development of airway obstruction. OBJECTIVES To assess the effects of methylation of 12 genes on FEV1 decline and of FEV1 decline on subsequent LC incidence using two independent, longitudinal cohorts (i.e., LSC [Lovelace Smokers Cohort] and PLuSS [Pittsburgh Lung Screening Study]). METHODS Gene methylation was measured in sputum using two-stage nested methylation-specific PCR. The linear mixed effects model was used to assess the effects of studied variables on FEV1 decline. MEASUREMENTS AND MAIN RESULTS A dose-dependent relationship between number of genes methylated and FEV1 decline was identified, with smokers with three or more methylated genes having 27.8% and 10.3% faster FEV1 decline than smokers with zero to two methylated genes in the LSC and PLuSS cohort, respectively (all P < 0.01). High methylation in sputum was associated with a shorter latency for LC incidence (log-rank P = 0.0048) and worse all-cause mortality (log-rank P < 0.0001). Smokers with subsequent LC incidence had a more rapid annual decline of FEV1 (by 5.2 ml, P = 0.038) than smoker control subjects. CONCLUSIONS Gene methylation detected in sputum predicted FEV1 decline, LC incidence, and all-cause mortality in smokers. Rapid FEV1 decline may be a risk factor for LC incidence in smokers, which may explain a greater prevalence of airway obstruction seen in patients with LC.
Collapse
Affiliation(s)
- Shuguang Leng
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Cancer Control Research Program and
- School of Public Health, Qingdao University, Qingdao, Shandong, China
| | - Brenda Diergaarde
- Department of Human Genetics and
- Cancer Epidemiology and Prevention Program
- Lung Cancer Program, and
| | - Maria A. Picchi
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - David O. Wilson
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Frank D. Gilliland
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California; and
| | - Jian-Min Yuan
- Department of Epidemiology, Graduate School of Public Health, and
- Division of Cancer Control and Population Sciences, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | | | - Steven A. Belinsky
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
- Cancer Control Research Program and
- Cancer Genetics and Epigenetics Program, University of New Mexico Comprehensive Cancer Center, Albuquerque, New Mexico
| |
Collapse
|
33
|
Su Y, Fang HB, Jiang F. An epigenetic classifier for early stage lung cancer. Clin Epigenetics 2018; 10:68. [PMID: 29796119 PMCID: PMC5964676 DOI: 10.1186/s13148-018-0502-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Accepted: 05/14/2018] [Indexed: 12/19/2022] Open
Abstract
Background Methylated genes detected in sputum are promise biomarkers for lung cancer. Yet the current PCR technologies for quantification of DNA methylation and diagnostic value of the sputum biomarkers are not sufficient to be used for lung cancer early detection. The emerging droplet digital PCR (ddPCR) is a straightforward means for precise, direct, and absolute quantification of nucleic acids. Here, we investigate whether ddPCR can sensitively and robustly quantify DNA methylation in sputum for more precise diagnosis of lung cancer. Results First, the analytic performance of methylation-specific ddPCR (ddMSP) and quantitative methylation-specific PCR (qMSP) is determined in methylated and unmethylated DNA samples. Second, 29 genes, previously proposed as potential sputum biomarkers for lung cancer, are analyzed by using ddMSP in a training set of 127 lung cancer patients and 159 controls. ddMSP has higher sensitivity, precision, and reproducibility for quantification of methylation compared with qMSP (all p < 0.05). A classifier comprising four sputum methylation biomarkers for lung cancer is developed by using ddMSP, producing 86.6% sensitivity and 90.6% specificity, independent of stage and histology of lung cancer (all p > 0.05). The classifier has higher accuracy compared with sputum cytology (88.8 vs. 70.6%, p < 0.01). The diagnostic performance is confirmed in a testing set of 89 cases and 107 controls. Conclusions ddMSP is a robust tool for reliable quantification of DNA methylation in sputum, and the epigenetic classifier could help diagnose lung cancer at the early stage.
Collapse
Affiliation(s)
- Yun Su
- 1Department of Surgery, Jiangsu Province Hospital of Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023 China
| | - Hong Bin Fang
- 2Department of Biostatistics, Bioinformatics and Biomathematics, Georgetown University Medical Center, 4000 Reservoir Road, N.W, Washington D.C., 20057 USA
| | - Feng Jiang
- 3Department of Pathology, University of Maryland School of Medicine, Baltimore, MD USA
| |
Collapse
|
34
|
de Antonellis P, De Luca A, Normanno N. Gene methylation in liquid biopsy and risk of recurrence in lung cancer. J Thorac Dis 2018; 10:1286-1289. [PMID: 29707279 DOI: 10.21037/jtd.2018.02.84] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Pasqualino de Antonellis
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| |
Collapse
|
35
|
Lin CC, Hsieh TC, Wu LSH. Long-term use of valproic acid and the prevalence of cancers in bipolar disorder patients in a Taiwanese population: An association analysis using the National Health Insurance Research Database (NHIRD). J Affect Disord 2018; 232:103-108. [PMID: 29481993 DOI: 10.1016/j.jad.2018.02.047] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/24/2018] [Accepted: 02/16/2018] [Indexed: 10/18/2022]
Abstract
BACKGROUND Epigenetic events play a major role in the carcinogenesis of many cancers. A retrospective cohort study had been performed to evaluate the effects of exposure to the anticonvulsant agent valproic acid (VPA), a histone deacetylase inhibitor, on the risk of developing cancers. METHODS The study was based on the 1998 through 2009 National Health Insurance Research Database (NHIRD), provided by the Taiwan National Health Research Institute. Patients with a diagnosis of bipolar disorder (ICD-9-CM codes 296.0, 296.1, 296.4-8) from 1998 to 2009 were identified. VPA and lithium were the primary index drugs. Patients treated with anticonvulsants who did not use VPA or lithium were selected as the control group. Competing risk regression analysis were used to estimate hazards ratios (HR) and 95% confidence intervals (95% CI) reflecting the association between use of VPA and cancer incidence. RESULTS The cancer incidence of bipolar disorder patients treated with VPA was no significant difference than treated with lithium and other anticonvulsants. In subgroup analysis, VPA associated to higher risk of genitourinary cancer in the duration < 1 year group (HR: 3.49; 95%CI: 1.04, 11.67). No significant differences in other cancers incidence in any duration of VPA treatment. LIMITATIONS The cancer prevalence in selected bipolar disorder patients was still low. The sample size was not enough for some types of cancer. CONCLUSIONS A role of VPA in cancer prevention was not found in this study. An increased subgroup risk of genitourinary cancer was observed.
Collapse
Affiliation(s)
- Cheng Chia Lin
- Department of Urology, Chang Gung Memorial Hospital, Keelung Division Taiwan, ROC
| | - Tsung-Cheng Hsieh
- Institute of Medical Sciences, Tzu Chi University, #701, Zhongyang Road, Section 3, Hualien 97004, Taiwan, ROC
| | - Lawrence Shih-Hsin Wu
- Graduate Institute of Biomedical Sciences, China Medical University, #91 Hsueh-Shih Road, Taichung, Taiwan, ROC.
| |
Collapse
|
36
|
Strzelczyk JK, Krakowczyk Ł, Owczarek AJ. Aberrant DNA methylation of the p16, APC, MGMT, TIMP3 and CDH1 gene promoters in tumours and the surgical margins of patients with oral cavity cancer. J Cancer 2018; 9:1896-1904. [PMID: 29896273 PMCID: PMC5995944 DOI: 10.7150/jca.24477] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/10/2018] [Indexed: 12/15/2022] Open
Abstract
Oral cavity cancer is a type of head and neck squamous cell carcinoma (HNSCC) and contributes to significant morbidity and mortality each year. An epigenetic pathway of transcriptional inactivation for many genes has been described in various cancers, including HNSCC. For our study, we selected genes for which silencing caused by hypermethylation can promote cancer development. In 75 primary HNSCC tumours and paired surgical margins, we investigated the methylation status of the p16, APC, MGMT, TIMP3 and CDH1 gene promoters by methylation-specific PCR after bisulphite treatment. The promoter methylation rates of p16, APC, MGMT, TIMP3 and CDH1 in tumours were 58.67%, 49.33%, 58.67%, 50.67%, and 57.33% and 50.67%, 41.33%, 37.33%, 42.67%, and 25.33% in the surgical margin, respectively. Our observations confirm the presence of epigenetic changes not only in the cancer cells, but also in the surrounding mucosa and represent a basis for further analysis to unravel these complicated issues. Appropriate cancer risk assessment based on epigenetic alterations in surgical margins may influence a patient's diagnosis and cure.
Collapse
Affiliation(s)
- Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, School of Medicine with the Division of Dentistry in Zabrze, Jordana 19 Str., 41-808 Zabrze, Medical University of Silesia in Katowice, Zabrze, Poland
| | - Łukasz Krakowczyk
- Clinic of Oncological and Reconstructive Surgery, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Gliwice Branch, Wybrzeże Armii Krajowej 15 Str., 44-101 Gliwice, Poland
| | - Aleksander Jerzy Owczarek
- Department of Statistics, Department of Instrumental Analysis, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Ostrogórska 30 Str., 41-200 Sosnowiec, Medical University of Silesia in Katowice, Sosnowiec, Poland
| |
Collapse
|
37
|
Epigenetic Modifications as Biomarkers of Tumor Development, Therapy Response, and Recurrence across the Cancer Care Continuum. Cancers (Basel) 2018; 10:cancers10040101. [PMID: 29614786 PMCID: PMC5923356 DOI: 10.3390/cancers10040101] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 02/06/2023] Open
Abstract
Aberrant epigenetic modifications are an early event in carcinogenesis, with the epigenetic landscape continuing to change during tumor progression and metastasis—these observations suggest that specific epigenetic modifications could be used as diagnostic and prognostic biomarkers for many cancer types. DNA methylation, post-translational histone modifications, and non-coding RNAs are all dysregulated in cancer and are detectable to various degrees in liquid biopsies such as sputum, urine, stool, and blood. Here, we will focus on the application of liquid biopsies, as opposed to tissue biopsies, because of their potential as non-invasive diagnostic tools and possible use in monitoring therapy response and progression to metastatic disease. This includes a discussion of septin-9 (SEPT9) DNA hypermethylation for detecting colorectal cancer, which is by far the most developed epigenetic biomarker assay. Despite their potential as prognostic and diagnostic biomarkers, technical issues such as inconsistent methodology between studies, overall low yield of epigenetic material in samples, and the need for improved histone and non-coding RNA purification methods are limiting the use of epigenetic biomarkers. Once these technical limitations are overcome, epigenetic biomarkers could be used to monitor cancer development, disease progression, therapeutic response, and recurrence across the entire cancer care continuum.
Collapse
|
38
|
Jiang CL, He SW, Zhang YD, Duan HX, Huang T, Huang YC, Li GF, Wang P, Ma LJ, Zhou GB, Cao Y. Air pollution and DNA methylation alterations in lung cancer: A systematic and comparative study. Oncotarget 2018; 8:1369-1391. [PMID: 27901495 PMCID: PMC5352062 DOI: 10.18632/oncotarget.13622] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 11/09/2016] [Indexed: 12/20/2022] Open
Abstract
The lung cancer incidence in the Xuanwei and neighboring region, Yunnan, China, is among the highest in China and is attributed to severe air pollution with high benzo(a)pyrene levels. We systematically and comparatively analyzed DNA methylation alterations at genome and gene levels in Xuanwei lung cancer tissues and cell lines, as well as benzo(a)pyrene-treated cells and mouse samples. We obtained a comprehensive dataset of genome-wide cytosine-phosphate-guanine island methylation in air pollution-related lung cancer samples. Benzo(a)pyrene exposure induced multiple alterations in DNA methylation and in mRNA expressions of DNA methyltransferases and ten-11 translocation proteins; these alterations partially occurred in Xuanwei lung cancer. Furthermore, benzo(a)pyrene-induced DKK2 and EN1 promoter hypermethylation and LPAR2 promoter hypomethylation led to down-regulation and up-regulation of the genes, respectively; the down-regulation of DKK2 and EN1 promoted the cellular proliferation. Thus, DNA methylation alterations induced by benzo(a)pyrene contribute partially to abnormal DNA methylation in air pollution-related lung cancer, and these DNA methylation alterations may affect the development and progression of lung cancer. Additionally, vitamin C and B6 can reduce benzo(a)pyrene-induced DNA methylation alterations and may be used as chemopreventive agents for air pollution-related lung cancer.
Collapse
Affiliation(s)
- Cheng-Lan Jiang
- Laboratory of Molecular and Experimental Pathology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.,Kunming College of Life Sciences, University of Chinese Academy of Sciences, Kunming 650223, China
| | - Shui-Wang He
- Laboratory of Molecular and Experimental Pathology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Yun-Dong Zhang
- Laboratory of Molecular and Experimental Pathology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.,School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - He-Xian Duan
- Laboratory of Molecular and Experimental Pathology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yun-Chao Huang
- Department of Thoracic and Cardiovascular Surgery, The Third Affiliated Hospital of Kunming Medical University, (Yunnan Tumor Hospital), Kunming 650106, China
| | - Gao-Feng Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Kunming Medical University, (Yunnan Tumor Hospital), Kunming 650106, China
| | - Ping Wang
- Department of Thoracic Surgery, The First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Li-Ju Ma
- Clinical Medicine Research Center, The First Affiliated Hospital of Kunming Medical University, Kunming 650332, China
| | - Guang-Biao Zhou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yi Cao
- Laboratory of Molecular and Experimental Pathology, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China
| |
Collapse
|
39
|
Zeng Y, Zhu J, Shen D, Qin H, Lei Z, Li W, Liu Z, Huang JA. MicroRNA-205 targets SMAD4 in non-small cell lung cancer and promotes lung cancer cell growth in vitro and in vivo. Oncotarget 2018; 8:30817-30829. [PMID: 28199217 PMCID: PMC5458170 DOI: 10.18632/oncotarget.10339] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Accepted: 06/13/2016] [Indexed: 12/25/2022] Open
Abstract
Despite advances in diagnosis and treatment, the survival of non-small cell lung cancer (NSCLC) patients remains poor; therefore, improved understanding of the disease mechanism and novel treatment strategies are needed. Downregulation of SMAD4 and dysregulated expression of miR-205 have been reported. However, the relationship between them remains unclear. We investigated the effect of microRNA (miR)-205 on the expression of SMAD4 in NSCLC. Knockdown and overexpression of SMAD4 promoted or suppressed cellular viability and proliferation, and accelerated or inhibited the cell cycle in NSCLC cells, respectively. The 3′-untranslated region (3′-UTR) of SMAD4 was predicted as a target of miR-205. Luciferase assays validated that miR-205 binds directly to the SMAD4 3′-UTR. Protein and mRNA expression analyses confirmed that miR-205 overexpression in NSCLC cells inhibited the expression of SMAD4 mRNA and protein. In human NSCLC tissues, increased miR-205 expression was observed frequently and was inversely correlated with decreased SMAD4 expression. Ectopic expression of miR-205 in NSCLC cells suppressed cellular viability and proliferation, accelerated the cell cycle, and promoted tumor growth of lung carcinoma xenografts in nude mice. Our study showed that miR-205 decreased SMAD4 expression, thus promoting NSCLC cell growth. Our findings highlighted the therapeutic potential of targeting miR-205 in NSCLC treatment.
Collapse
Affiliation(s)
- Yuanyuan Zeng
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China.,Institute of Respiratory Diseases, Soochow University, Suzhou, P. R. China
| | - Jianjie Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China.,Institute of Respiratory Diseases, Soochow University, Suzhou, P. R. China
| | - Dan Shen
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China
| | - Hualong Qin
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China
| | - Zhe Lei
- Suzhou Key Laboratory for Molecular Cancer Genetics, Suzhou, P. R. China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China
| | - Zeyi Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China.,Institute of Respiratory Diseases, Soochow University, Suzhou, P. R. China
| | - Jian-An Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, P. R. China.,Institute of Respiratory Diseases, Soochow University, Suzhou, P. R. China
| |
Collapse
|
40
|
Su J, Leng Q, Lin Y, Ma J, Jiang F, Lee CJ, Fang H, Jiang F. Integrating Circulating Immunological and Sputum Biomarkers for the Early Detection of Lung Cancer. BIOMARKERS IN CANCER 2018; 10:1179299X18759297. [PMID: 29467585 PMCID: PMC5815414 DOI: 10.1177/1179299x18759297] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 01/23/2018] [Indexed: 01/02/2023]
Abstract
We have demonstrated that assessments of microRNA (miRNA) expressions in circulating peripheral blood mononucleated cell (PBMC) and sputum specimens, respectively, may help diagnose lung cancer. To assess the individual and combined analysis of the miRNAs across the different body fluids for lung cancer early detection, we analyse a panel of 3 sputum miRNAs (miRs-21, 31, and 210) and a panel of 2 PBMC miRNAs (miRs-19b-3p and 29b-3p) in a discovery cohort of 68 patients with lung cancer and 66 cancer-free smokers. We find that integrating 2 sputum miRNAs (miRs-31 and 210) and 1 PBMC miRNA (miR-19b-3p) has higher sensitivity (86.8%) and specificity (92.4%) compared with the individual panels. The synergistic value of the integrated panel of 3 biomarkers is confirmed in a validation cohort, independent of stage and histological type of lung cancer, and patients' age, sex, and ethnicity. Integrating circulating immunological and sputum biomarkers could improve the early detection of lung cancer.
Collapse
Affiliation(s)
- Jian Su
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Qixin Leng
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yanli Lin
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jie Ma
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Fangran Jiang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Cheng-Ju Lee
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - HongBin Fang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Feng Jiang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
41
|
Ningaraj N, Salimath B, Sankpal U, Perera R, Vats T. Targeted Brain Tumor Treatment-Current Perspectives. Drug Target Insights 2017. [DOI: 10.1177/117739280700200008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Affiliation(s)
- N.S. Ningaraj
- Department of Pediatric Neurooncology and Molecular Pharmacology, Hoskins Center, Curtis and Elizabeth Anderson Cancer Institute, Memorial Health University Medical Center, Mercer University Medical School, 4700 Waters Avenue, Savannah, GA 31404, U.S.A
| | - B.P. Salimath
- Department of Biotechnology, University of Mysore, Mysore 570006, Karnataka, India
| | - U.T. Sankpal
- Department of Pediatric Neurooncology and Molecular Pharmacology, Hoskins Center, Curtis and Elizabeth Anderson Cancer Institute, Memorial Health University Medical Center, Mercer University Medical School, 4700 Waters Avenue, Savannah, GA 31404, U.S.A
| | - R Perera
- Department of Pediatric Neurooncology and Molecular Pharmacology, Hoskins Center, Curtis and Elizabeth Anderson Cancer Institute, Memorial Health University Medical Center, Mercer University Medical School, 4700 Waters Avenue, Savannah, GA 31404, U.S.A
| | - T Vats
- Department of Pediatric Neurooncology and Molecular Pharmacology, Hoskins Center, Curtis and Elizabeth Anderson Cancer Institute, Memorial Health University Medical Center, Mercer University Medical School, 4700 Waters Avenue, Savannah, GA 31404, U.S.A
| |
Collapse
|
42
|
Leng S, Picchi MA, Kang H, Wu G, Filipczak PT, Juri DE, Zhang X, Gauderman WJ, Gilliland FD, Belinsky SA. Dietary Nutrient Intake, Ethnicity, and Epigenetic Silencing of Lung Cancer Genes Detected in Sputum in New Mexican Smokers. Cancer Prev Res (Phila) 2017; 11:93-102. [PMID: 29118161 DOI: 10.1158/1940-6207.capr-17-0196] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/19/2017] [Accepted: 10/30/2017] [Indexed: 12/17/2022]
Abstract
Lung cancer gene methylation detected in sputum assesses field cancerization and predicts lung cancer incidence. Hispanic smokers have higher lung cancer susceptibility compared with non-Hispanic whites (NHW). We aimed to identify novel dietary nutrients affecting lung cancer gene methylation and determine the degree of ethnic disparity in methylation explained by diet. Dietary intakes of 139 nutrients were assessed using a validated Harvard food frequency questionnaire in 327 Hispanics and 1,502 NHWs from the Lovelace Smokers Cohort. Promoter methylation of 12 lung cancer genes was assessed in sputum DNA. A global association was identified between dietary intake and gene methylation (Ppermutation = 0.003). Seventeen nutrient measurements were identified with magnitude of association with methylation greater than that seen for folate. A stepwise approach identified B12, manganese, sodium, and saturated fat as the minimally correlated set of nutrients whose optimal intakes could reduce the methylation by 36% (Ppermutation < 0.001). Six protective nutrients included vitamin D, B12, manganese, magnesium, niacin, and folate. Approximately 42% of ethnic disparity in methylation was explained by insufficient intake of protective nutrients in Hispanics compared with NHWs. Functional validation of protective nutrients showed an enhanced DNA repair capacity toward double-strand DNA breaks, a mechanistic biomarker strongly linked to acquisition of lung cancer gene methylation in smokers. Dietary intake is a major modifiable factor for preventing promoter methylation of lung cancer genes in smokers' lungs. Complex dietary supplements could be developed on the basis of these protective nutrients for lung cancer chemoprevention in smokers. Hispanic smokers may benefit the most from this complex for reducing their lung cancer susceptibility. Cancer Prev Res; 11(2); 93-102. ©2017 AACR.
Collapse
Affiliation(s)
- Shuguang Leng
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico. .,Cancer Control (CaC) Research Program, University of New Mexico Cancer Center, Albuquerque, New Mexico
| | - Maria A Picchi
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Huining Kang
- Department of Internal Medicine and UNM Comprehensive Cancer Center, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Guodong Wu
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Piotr T Filipczak
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Daniel E Juri
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Xiequn Zhang
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - W James Gauderman
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Frank D Gilliland
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Steven A Belinsky
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico. .,Cancer Control (CaC) Research Program, University of New Mexico Cancer Center, Albuquerque, New Mexico
| |
Collapse
|
43
|
Lissa D, Robles AI. Sputum-based DNA methylation biomarkers to guide lung cancer screening decisions. J Thorac Dis 2017; 9:4308-4310. [PMID: 29268498 DOI: 10.21037/jtd.2017.10.78] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Delphine Lissa
- Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, USA
| | - Ana I Robles
- Laboratory of Human Carcinogenesis, National Cancer Institute, NIH, Bethesda, USA
| |
Collapse
|
44
|
Fusco N, Fumagalli C, Guerini-Rocco E. Looking for sputum biomarkers in lung cancer secondary prevention: where are we now? J Thorac Dis 2017; 9:4277-4279. [PMID: 29268490 DOI: 10.21037/jtd.2017.10.22] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Nicola Fusco
- Division of Pathology, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy.,Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | | | - Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
45
|
An YQC, Goettel W, Han Q, Bartels A, Liu Z, Xiao W. Dynamic Changes of Genome-Wide DNA Methylation during Soybean Seed Development. Sci Rep 2017; 7:12263. [PMID: 28947812 PMCID: PMC5613027 DOI: 10.1038/s41598-017-12510-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/08/2017] [Indexed: 02/06/2023] Open
Abstract
Seed development is programmed by expression of many genes in plants. Seed maturation is an important developmental process to soybean seed quality and yield. DNA methylation is a major epigenetic modification regulating gene expression. However, little is known about the dynamic nature of DNA methylation and its effects on gene expression during plant development. Through whole-genome bisulfite sequencing, we showed that DNA methylation went through dynamic changes during seed maturation. An average of 66% CG, 45% CHG and 9% CHH contexts was methylated in cotyledons. CHH methylation levels in cotyledons changed greatly from 6% at the early stage to 11% at the late stage. Transcribed genes were approximately two-fold more likely to be differentially methylated than non-transcribed genes. We identified 40, 66 and 2136 genes containing differentially methylated regions (DMRs) with negative correlation between their expression and methylation in the CG, CHG and CHH contexts, respectively. The majority of the DMR genes in the CHH context were transcriptionally down-regulated as seeds mature: 99% of them during early maturation were down-regulated, and preferentially associated with DNA replication and cell division. The results provide novel insights into the dynamic nature of DNA methylation and its relationship with gene regulation in seed development.
Collapse
Affiliation(s)
- Yong-Qiang Charles An
- US Department of Agriculture, Agricultural Research Service, Midwest Area, Plant Genetics Research Unit, Donald Danforth Plant Science Center, St. Louis, MO, 63132, USA.
| | - Wolfgang Goettel
- US Department of Agriculture, Agricultural Research Service, Midwest Area, Plant Genetics Research Unit, Donald Danforth Plant Science Center, St. Louis, MO, 63132, USA
| | - Qiang Han
- Department of Biology, Saint Louis University, St. Louis, MO, 63103, USA
| | - Arthur Bartels
- Department of Biology, Saint Louis University, St. Louis, MO, 63103, USA
| | - Zongrang Liu
- US Department of Agriculture, Agricultural Research Service, Appalachian Fruit Research Station, Kearneysville, WV, 25430, USA
| | - Wenyan Xiao
- Department of Biology, Saint Louis University, St. Louis, MO, 63103, USA.
| |
Collapse
|
46
|
Duruisseaux M, Esteller M. Lung cancer epigenetics: From knowledge to applications. Semin Cancer Biol 2017; 51:116-128. [PMID: 28919484 DOI: 10.1016/j.semcancer.2017.09.005] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/12/2017] [Accepted: 09/13/2017] [Indexed: 12/17/2022]
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Advances in our understanding of the genomics of lung cancer have led to substantial progress in the treatment of specific molecular subsets. Immunotherapy also emerges as a major breakthrough in lung cancer treatment. However, challenges remain as a consensual approach for early lung cancer detection remains elusive while primary or secondary drug resistance eventually leads to treatment failure in all patients with advanced disease. Furthermore, a large portion of patients are still treated with conventional chemotherapy that is only modestly effective. The last two decades have seen exponential developments in the epigenetic understanding of lung cancer. Epigenetic alterations in DNA methylation, non-coding RNA expression, chromatin modeling and post transcriptional regulators are key events in each step of lung cancer pathogenesis. Here, we review the central role epigenetic disruptions play in lung cancer carcinogenesis and the acquisition of cancerous phenotype and aggressive behavior as well as in the resistance to therapy. Epigenetic disruptions could represent reliable biomarkers for lung cancer risk assessment, early diagnosis, prognosis stratification, molecular classification and prediction of treatment efficacy. The therapeutic potential of epigenetics targeted drugs in combination with chemotherapy, targeted therapy and/or immunotherapy is currently being intensively investigated. We suggest that integration of tissue-derived or circulating epigenetic biomarkers and epidrugs in clinical trial design will translate epigenetic knowledge of lung cancer into the clinic and improve lung cancer patient outcomes.
Collapse
Affiliation(s)
- Michaël Duruisseaux
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain and Centro de Investigación Biomédica en Red de Cáncer (CIBERONC); Department of Respiratory Medecine, Hôpital Louis-Pradel, Hospices civils de Lyon, 28 avenue du Doyen Lépine, 69677, Lyon cedex, France.
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), 08908 L'Hospitalet de Llobregat, Barcelona, Catalonia, Spain and Centro de Investigación Biomédica en Red de Cáncer (CIBERONC); Instituciò Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Catalonia, Spain; Department of Physiological Sciences II, School of Medicine, University of Barcelona, 08036, Barcelona, Catalonia, Spain.
| |
Collapse
|
47
|
Belinsky SA, Leng S, Wu G, Thomas CL, Picchi MA, Lee SJ, Aisner S, Ramalingam S, Khuri FR, Karp DD. Gene Methylation Biomarkers in Sputum and Plasma as Predictors for Lung Cancer Recurrence. Cancer Prev Res (Phila) 2017; 10:635-640. [PMID: 28904059 DOI: 10.1158/1940-6207.capr-17-0177] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/20/2017] [Accepted: 08/31/2017] [Indexed: 12/25/2022]
Abstract
Detection of methylated genes in exfoliated cells from the lungs of smokers provides an assessment of the extent of field cancerization, is a validated biomarker for predicting lung cancer, and provides some discrimination when interrogated in blood. The potential utility of this 8-gene methylation panel for predicting tumor recurrence has not been assessed. The Eastern Cooperative Oncology Group initiated a prevention trial (ECOG-ACRIN5597) that enrolled resected stage I non-small cell lung cancer patients who were randomized 2:1 to receive selenized yeast versus placebo for 4 years. We conducted a correlative biomarker study to assess prevalence for methylation of the 8-gene panel in longitudinally collected sputum and blood after tumor resection to determine whether selenium alters their methylation profile and whether this panel predicts local and/or distant recurrence. Patients (N = 1,561) were enrolled into the prevention trial; 565 participated in the biomarker study with 122 recurrences among that group. Assessing the association between recurrence and risk of gene methylation longitudinally for up to 48 months showed a 1.4-fold increase in OR for methylation in sputum in the placebo group independent of location (local or distant). Kaplan-Meier curves evaluating the association between number of methylated genes and time to recurrence showed no increased risk in sputum, while a significant HR of 1.5 was seen in plasma. Methylation detection in sputum and blood is associated with risk for recurrence. Cancer Prev Res; 10(11); 635-40. ©2017 AACR.
Collapse
Affiliation(s)
- Steven A Belinsky
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico.
| | - Shuguang Leng
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Guodong Wu
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Cynthia L Thomas
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Maria A Picchi
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico
| | - Sandra J Lee
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Seena Aisner
- Rutgers New Jersey Medical School, Newark, New Jersey
| | - Suresh Ramalingam
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | - Fadlo R Khuri
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, Georgia
| | | |
Collapse
|
48
|
Nakayama R, Arikawa K, Bhawal UK. The epigenetic regulation of CXCL14 plays a role in the pathobiology of oral cancers. J Cancer 2017; 8:3014-3027. [PMID: 28928893 PMCID: PMC5604453 DOI: 10.7150/jca.21169] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/09/2017] [Indexed: 02/07/2023] Open
Abstract
Background: Chemokines selectively attract and activate leukocytes and play roles in a variety of homeostatic and disease processes. Explore the biological properties of CXCL14 seems complicated due to unknown functional characteristics of CXCL14 in cancer. Methods: To study the multistep process of oral cancer development, we analyzed oral samples spanning normalcy, dysplasia and cancer from multiple perspectives, revealing a cascade of progressive changes. Results: CXCL14 protein was expressed in the cytoplasm adjacent to tumors. T classification (P<0.001), clinical stage (P=0.0013) and nodal metastasis (P=0.0035) were significantly associated with CXCL14 in relationships between CXCL14 expression levels and tumor and patient characteristics. Compared with non-tumor tissue, expression of the epidermal growth factor receptor (EGFR) gene was increased in dysplasia and was further sustained in cancer. Our data show an inverse relationship between CXCL14 and EGFR expression levels in tumor cells indicating that CXCL14 expression is beneficial for tumor suppression. To explore epigenetic regulation and the impact of CXCL14 on oral cancer, analysis of CpG islands methylation in the CXCL14 promoter region indicated that the abnormal hypermethylation of that promoter region in tumor cells and tissues is one of the mechanisms causing the reduced expression. Restoration of CXCL14 expression was induced by treatment with 5-aza-2'-deoxycytidine. Using in vivo mouse models, we demonstrate that the restoration of CXCL14 expression in irradiation-induced oral carcinoma cells induces the expression of Late Cornified Envelope (LCE) genes. Conclusions: Our data suggest that LCE genes are a novel target of CXCL14 and are likely to have a tumor suppressor function through the modulation of CXCL14 expression. In conclusion, CXCL14 might play a pivotal role in the pathobiology of oral cancer, probably by regulating DNA methylation and leukocyte migration. The level of CXCL14 expression may be a valuable adjuvant parameter to predict the prognosis of patients with oral carcinoma and may be a potential therapeutic target.
Collapse
Affiliation(s)
- Ryuji Nakayama
- Department of Preventive and Public Oral Health, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakae-cho Nishi, Matsudo, Chiba 271-8587, Japan
| | - Kazumune Arikawa
- Department of Preventive and Public Oral Health, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakae-cho Nishi, Matsudo, Chiba 271-8587, Japan.,Research Institute of Oral Health, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakae-cho Nishi, Matsudo, Chiba 271-8587, Japan
| | - Ujjal K Bhawal
- Research Institute of Oral Health, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakae-cho Nishi, Matsudo, Chiba 271-8587, Japan.,Department of Oral Health, Graduate School of Dentistry, Kanagawa Dental University, 82 Inaoka-cho, Yokosuka, Kanagawa 238-8580, Japan.,Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, 2-870-1 Sakae-cho Nishi, Matsudo, Chiba 271-8587, Japan
| |
Collapse
|
49
|
Leng S, Wu G, Klinge DM, Thomas CL, Casas E, Picchi MA, Stidley CA, Lee SJ, Aisner S, Siegfried JM, Ramalingam S, Khuri FR, Karp DD, Belinsky SA. Gene methylation biomarkers in sputum as a classifier for lung cancer risk. Oncotarget 2017; 8:63978-63985. [PMID: 28969046 PMCID: PMC5609978 DOI: 10.18632/oncotarget.19255] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 06/05/2017] [Indexed: 01/01/2023] Open
Abstract
CT screening for lung cancer reduces mortality, but will cost Medicare ∼2 billion dollars due in part to high false positive rates. Molecular biomarkers could augment current risk stratification used to select smokers for screening. Gene methylation in sputum reflects lung field cancerization that remains in lung cancer patients post-resection. This population was used in conjunction with cancer-free smokers to evaluate classification accuracy of a validated eight-gene methylation panel in sputum for cancer risk. Sputum from resected lung cancer patients (n=487) and smokers from Lovelace (n=1380) and PLuSS (n=718) cohorts was studied for methylation of an 8-gene panel. Area under a receiver operating characteristic curve was calculated to assess the prediction performance in logistic regressions with different sets of variables. The prevalence for methylation of all genes was significantly increased in the ECOG-ACRIN patients compared to cancer-free smokers as evident by elevated odds ratios that ranged from 1.6 to 8.9. The gene methylation panel showed lung cancer prediction accuracy of 82–86% and with addition of clinical variables improved to 87–90%. With sensitivity at 95%, specificity increased from 25% to 54% comparing clinical variables alone to their inclusion with methylation. The addition of methylation biomarkers to clinical variables would reduce false positive screens by ruling out one-third of smokers eligible for CT screening and could increase cancer detection rates through expanding risk assessment criteria.
Collapse
Affiliation(s)
- Shuguang Leng
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Guodong Wu
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Donna M Klinge
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Cynthia L Thomas
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Elia Casas
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Maria A Picchi
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Christine A Stidley
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Sandra J Lee
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Seena Aisner
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jill M Siegfried
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, USA
| | - Suresh Ramalingam
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Fadlo R Khuri
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | | | - Steven A Belinsky
- Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| |
Collapse
|
50
|
Ali A, Kumar S, Kakaria VK, Mohan A, Luthra K, Upadhyay AD, Guleria R. Detection of Promoter DNA Methylation of APC, DAPK, and GSTP1 Genes in tissue Biopsy and Matched Serum of Advanced-Stage Lung Cancer Patients. Cancer Invest 2017; 35:423-430. [PMID: 28524770 DOI: 10.1080/07357907.2017.1309547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Promoter DNA hypermethylation of APC, DAPK, and GSTP1 genes was evaluated in biopsy and matched serum of 160 lung cancer patients and 70 controls. In biopsy, 83.1, 83.1, and 78.1% of lung cancer patients and 72.9, 70, and 70% of controls, while in serum, 52.5, 30.6, and 65.6% of lung cancer patients and 14.3, 18.6, and 30% of controls were positive for APC, DAPK, and GSTP1 hypermethylation respectively. We couldn't find any significant role of DNA hypermethylation in lung cancer. However, long follow-up of methylation positive controls will be required to confirm its role for the prediction of lung cancer.
Collapse
Affiliation(s)
- Ashraf Ali
- a Department of Pulmonary Medicine and Sleep Disorders , All India Institute of Medical Science , New Delhi , India
| | - Sachin Kumar
- b Department of Medical Oncology, All India Institute of Medical Science , New Delhi , India
| | | | - Anant Mohan
- a Department of Pulmonary Medicine and Sleep Disorders , All India Institute of Medical Science , New Delhi , India
| | - Kalpana Luthra
- d Department of Biochemistry , All India Institute of Medical Science , New Delhi , India
| | - Ashish Dutt Upadhyay
- e Department of Biostatistics , All India Institute of Medical Science , New Delhi , India
| | - Randeep Guleria
- a Department of Pulmonary Medicine and Sleep Disorders , All India Institute of Medical Science , New Delhi , India
| |
Collapse
|