1
|
Khalil A, Dinh T, Parks M, Obeng RC, Gryder B, Kresak A, Wang Y, Maltas J, Bedrock M, Wei X, Faber Z, Rahm M, Scott J, LaFramboise T, Wang Z, McFarland C. In Vivo Multiplexed Modeling Reveals Diverse Roles of the TBX2 Subfamily and Egr1 in Ras -Driven Lung Adenocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.15.642187. [PMID: 40166332 PMCID: PMC11956923 DOI: 10.1101/2025.03.15.642187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The TBX2 subfamily of T-box transcription factors (including Tbx2 , Tbx3 , Tbx4 , Tbx5 ) plays an essential role in lung development. Downregulation of these genes in human Lung adenocarcinoma (LUAD) suggests that these genes may be tumor suppressive, however because downregulation appears to occur primarily via epigenetic change, it remains unclear if these changes causally drive tumor progression or are merely the consequence of upstream events. Herein, we developed the first multiplexed mouse model to study the impact of TBX2 subfamily loss, alongside associated signaling genes Egr1 , Chd2 , Tnfaip3a , and Atf3 , in Ras -driven lung cancer. Using TuBa-seq, a high-throughput tumor-barcoding system, we quantified the growth effects of these knockouts during early and late tumorigenesis. Chd2 loss consistently suppressed tumor progression, while Tbx2 loss exhibited stage-dependent effects. Notably, Egr1 emerged as a potent tumor suppressor, with its knockout increasing tumor size (∼5x) at 20 weeks, surpassing Rb1 loss. Transcriptomic analyses of Egr1 -deficient tumors suggested immune dysregulation, including heightened inflammation and potential markers of T cell exhaustion in the tumor microenvironment. These findings indicate that Egr1 may play a role in suppressing tumor growth through modulating immune dynamics, offering new insights into the interplay between tumor progression and immune regulation in LUAD.
Collapse
|
2
|
Mi R, Wang Q, Liu Q, Jiang F, Ji Y. Expression and prognosis analysis of TBX2 subfamily in human lung carcinoma. Discov Oncol 2024; 15:51. [PMID: 38413457 PMCID: PMC10899548 DOI: 10.1007/s12672-024-00900-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/20/2024] [Indexed: 02/29/2024] Open
Abstract
PURPOSE Lung cancer has a high morbidity and mortality rate of all cancers worldwide. Therefore, there is an urgent need for reliable cancer markers for diagnosis and prognosis of patients with lung cancer. METHODS In this study, we used the bioinformatics database to compare the expression of the TBX2 subfamily at the transcriptional and protein levels in non-small cell lung cancer. Then, to confirm our bioinformatics analysis above, we used western bloting to determine the expression of TBX2, TBX3, TBX4 and TBX5 in human lung squamous carcinoma cell lines. Besides, low expression of TBX2 subfamily predicted a poor prognosis of patients with lung cancer. Finally, The methylation database was used to explore the relationship between the low expression of TBX2 subfamily and methylation of gene promoter region. RESULTS Our data showed a significant decrease of TBX2 subfamily expression in lung cancer tissues of several histological subtypes. Finally, the methylation of TBX2 subfamily members in the promoter region of NSCLC was significantly higher than that in normal tissues. CONCLUSION Our research provided sufficient evidence that TBX2 subfamily might play an inhibitory role in malignancy progression of lung cancer, which is promising to shed light on discovering a novel reliable cancer marker for prognosis of lung cancer patients.
Collapse
Affiliation(s)
- Rui Mi
- Department of Clinical Laboratory, Wuxi 9Th People's Hospital Affiliated to Soochow University, No.999 Liang Xi Road, Binhu District, Wuxi, 214000, Jiangsu, China
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi 9Th People's Hospital Affiliated to Soochow University, No.999 Liang Xi Road, Binhu District, Wuxi, 214000, Jiangsu, China
| | - Qingyang Liu
- Department of Clinical Laboratory, Wuxi 9Th People's Hospital Affiliated to Soochow University, No.999 Liang Xi Road, Binhu District, Wuxi, 214000, Jiangsu, China
| | - Fengying Jiang
- Department of Clinical Laboratory, Wuxi 9Th People's Hospital Affiliated to Soochow University, No.999 Liang Xi Road, Binhu District, Wuxi, 214000, Jiangsu, China
| | - Yuan Ji
- School of Medicine, Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China.
| |
Collapse
|
3
|
Altaf R, Ilyas U, Ma A, Shi M. Identification and validation of differentially expressed genes for targeted therapy in NSCLC using integrated bioinformatics analysis. Front Oncol 2023; 13:1206768. [PMID: 37324026 PMCID: PMC10264625 DOI: 10.3389/fonc.2023.1206768] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Background Despite the high prevalence of lung cancer, with a five-year survival rate of only 23%, the underlying molecular mechanisms of non-small cell lung cancer (NSCLC) remain unknown. There is a great need to identify reliable candidate biomarker genes for early diagnosis and targeted therapeutic strategies to prevent cancer progression. Methods In this study, four datasets obtained from the Gene Expression Omnibus were evaluated for NSCLC- associated differentially expressed genes (DEGs) using bioinformatics analysis. About 10 common significant DEGs were shortlisted based on their p-value and FDR (DOCK4, ID2, SASH1, NPR1, GJA4, TBX2, CD24, HBEGF, GATA3, and DDR1). The expression of significant genes was validated using experimental data obtained from TCGA and the Human Protein Atlas database. The human proteomic data for post- translational modifications was used to interpret the mutations in these genes. Results Validation of DEGs revealed a significant difference in the expression of hub genes in normal and tumor tissues. Mutation analysis revealed 22.69%, 48.95%, and 47.21% sequence predicted disordered regions of DOCK4, GJA4, and HBEGF, respectively. The gene-gene and drug-gene network analysis revealed important interactions between genes and chemicals suggesting they could act as probable drug targets. The system-level network showed important interactions between these genes, and the drug interaction network showed that these genes are affected by several types of chemicals that could serve as potential drug targets. Conclusions The study demonstrates the importance of systemic genetics in identifying potential drug- targeted therapies for NSCLC. The integrative system- level approach should contribute to a better understanding of disease etiology and may accelerate drug discovery for many cancer types.
Collapse
Affiliation(s)
- Reem Altaf
- Department of Pharmacy, Iqra University, Islamabad, Pakistan
| | - Umair Ilyas
- Department of Pharmaceutics, Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Anmei Ma
- Department of Medical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Meiqi Shi
- Department of Medical Oncology, Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Hudlikar RR, Chou PJ, Kuo HCD, Sargsyan D, Wu R, Kong AN. Long term exposure of cigarette smoke condensate (CSC) mediates transcriptomic changes in normal human lung epithelial Beas-2b cells and protection by garlic compounds. Food Chem Toxicol 2023; 174:113656. [PMID: 36758788 DOI: 10.1016/j.fct.2023.113656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023]
Abstract
Chronic cigarette smoke condensate (CSC) exposure is one of the preventable risk factors in the CS-induced lung cancer. However, understanding the mechanism of cellular transformation induced by CS in the lung remains limited. We investigated the effect of long term exposure of CSC in human normal lung epithelial Beas-2b cells, and chemopreventive mechanism of organosulphur garlic compounds, diallyl sulphide (DAS) and diallyl disulphide (DADS) using Next Generation Sequencing (NGS) transcriptomic analysis. CSC regulated 1077 genes and of these 36 genes are modulated by DAS while 101 genes by DADS. DAS modulated genes like IL1RL1 (interleukin-1 receptor like-1), HSPA-6 (heat shock protein family A, member 6) while DADS demonstrating ADTRP (Androgen-Dependent TFPI Regulating Protein), ANGPT4 (Angiopoietin 4), GFI1 (Growth Factor-Independent 1 Transcriptional Repressor), TBX2 (T-Box Transcription Factor 2), with some common genes like NEURL-1 (Neuralized E3-Ubiquitin Protein Ligase 1), suggesting differential effects between these two garlic compounds. They regulate genes by influencing pathways including HIF-1alpha, STAT-3 and matrix metalloproteases, contributing to the chemoprotective ability of organosulfur garlic compounds against CSC-induced cellular transformation. Taken together, we demonstrated CSC induced global gene expression changes pertaining to cellular transformation which potentially can be delayed with dietary chemopreventive phytochemicals like DS and DADS influencing alterations at the transcriptomic level.
Collapse
Affiliation(s)
- Rasika R Hudlikar
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Pochung Jordan Chou
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Hsiao-Chen Dina Kuo
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Davit Sargsyan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Renyi Wu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Ah-Ng Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, 08854, USA.
| |
Collapse
|
5
|
Gholami L, Ivari JR, Nasab NK, Oskuee RK, Sathyapalan T, Sahebkar A. Recent Advances in Lung Cancer Therapy Based on Nanomaterials: A Review. Curr Med Chem 2023; 30:335-355. [PMID: 34375182 DOI: 10.2174/0929867328666210810160901] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/22/2021] [Accepted: 06/26/2021] [Indexed: 11/22/2022]
Abstract
Lung cancer is one of the commonest cancers with a significant mortality rate for both genders, particularly in men. Lung cancer is recognized as one of the leading causes of death worldwide, which threatens the lives of over 1.6 million people every day. Although cancer is the leading cause of death in industrialized countries, conventional anticancer medications are unlikely to increase patients' life expectancy and quality of life significantly. In recent years, there are significant advances in the development and applications of nanotechnology in cancer treatment. The superiority of nanostructured approaches is that they act more selectively than traditional agents. This progress led to the development of a novel field of cancer treatment known as nanomedicine. Various formulations based on nanocarriers, including lipids, polymers, liposomes, nanoparticles and dendrimers have opened new horizons in lung cancer therapy. The application and expansion of nano-agents lead to an exciting and challenging research era in pharmaceutical science, especially for the delivery of emerging anti-cancer agents. The objective of this review is to discuss the recent advances in three types of nanoparticle formulations for lung cancer treatments modalities, including liposomes, polymeric micelles, and dendrimers for efficient drug delivery. Afterward, we have summarized the promising clinical data on nanomaterials based therapeutic approaches in ongoing clinical studies.
Collapse
Affiliation(s)
- Leila Gholami
- Nanotechnology Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Rouhani Ivari
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niloofar Khandan Nasab
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Kazemi Oskuee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, United Kingdom of Great Britain and Northern Ireland
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.,School of Medicine, The University of Western Australia, Perth, Australia.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
6
|
Janke F, Angeles AK, Riediger AL, Bauer S, Reck M, Stenzinger A, Schneider MA, Muley T, Thomas M, Christopoulos P, Sültmann H. Longitudinal monitoring of cell-free DNA methylation in ALK-positive non-small cell lung cancer patients. Clin Epigenetics 2022; 14:163. [PMID: 36461127 PMCID: PMC9719130 DOI: 10.1186/s13148-022-01387-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND DNA methylation (5-mC) signals in cell-free DNA (cfDNA) of cancer patients represent promising biomarkers for minimally invasive tumor detection. The high abundance of cancer-associated 5-mC alterations permits parallel and highly sensitive assessment of multiple 5-mC biomarkers. Here, we performed genome-wide 5-mC profiling in the plasma of metastatic ALK-rearranged non-small cell lung cancer (NSCLC) patients receiving tyrosine kinase inhibitor therapy. We established a strategy to identify ALK-specific 5-mC changes from cfDNA and demonstrated the suitability of the identified markers for cancer detection, prognosis, and therapy monitoring. METHODS Longitudinal plasma samples (n = 79) of 21 ALK-positive NSCLC patients and 13 healthy donors were collected alongside 15 ALK-positive tumor tissue and 10 healthy lung tissue specimens. All plasma and tissue samples were analyzed by cell-free DNA methylation immunoprecipitation sequencing to generate genome-wide 5-mC profiles. Information on genomic alterations (i.e., somatic mutations/fusions and copy number alterations) determined in matched plasma samples was available from previous studies. RESULTS We devised a strategy that identified tumor-specific 5-mC biomarkers by reducing 5-mC background signals derived from hematopoietic cells. This was followed by differential methylation analysis (cases vs. controls) and biomarker validation using 5-mC profiles of ALK-positive tumor tissues. The resulting 245 differentially methylated regions were enriched for lung adenocarcinoma-specific 5-mC patterns in TCGA data and indicated transcriptional repression of several genes described to be silenced in NSCLC (e.g., PCDH10, TBX2, CDO1, and HOXA9). Additionally, 5-mC-based tumor DNA (5-mC score) was highly correlated with other genomic alterations in cell-free DNA (Spearman, ρ > 0.6), while samples with high 5-mC scores showed significantly shorter overall survival (log-rank p = 0.025). Longitudinal 5-mC scores reflected radiologic disease assessments and were significantly elevated at disease progression compared to the therapy start (p = 0.0023). In 7 out of 8 instances, rising 5-mC scores preceded imaging-based evaluation of disease progression. CONCLUSION We demonstrated a strategy to identify 5-mC biomarkers from the plasma of cancer patients and integrated them into a quantitative measure of cancer-associated 5-mC alterations. Using longitudinal plasma samples of ALK-positive NSCLC patients, we highlighted the suitability of cfDNA methylation for prognosis and therapy monitoring.
Collapse
Affiliation(s)
- Florian Janke
- grid.5253.10000 0001 0328 4908Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany ,grid.452624.3German Center for Lung Research (DZL), TLRC Heidelberg, Heidelberg, Germany
| | - Arlou Kristina Angeles
- grid.5253.10000 0001 0328 4908Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany ,grid.452624.3German Center for Lung Research (DZL), TLRC Heidelberg, Heidelberg, Germany
| | - Anja Lisa Riediger
- grid.5253.10000 0001 0328 4908Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany ,grid.7497.d0000 0004 0492 0584Helmholtz Young Investigator Group, Multiparametric Methods for Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.5253.10000 0001 0328 4908Department of Urology, Heidelberg University Hospital, Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Simone Bauer
- grid.5253.10000 0001 0328 4908Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany
| | - Martin Reck
- grid.452624.3Lung Clinic Grosshansdorf, Airway Research Center North, German Center for Lung Research, Grosshansdorf, Germany
| | - Albrecht Stenzinger
- grid.452624.3German Center for Lung Research (DZL), TLRC Heidelberg, Heidelberg, Germany ,grid.5253.10000 0001 0328 4908Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Marc A. Schneider
- grid.452624.3German Center for Lung Research (DZL), TLRC Heidelberg, Heidelberg, Germany ,grid.5253.10000 0001 0328 4908Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Muley
- grid.452624.3German Center for Lung Research (DZL), TLRC Heidelberg, Heidelberg, Germany ,grid.5253.10000 0001 0328 4908Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Thomas
- grid.452624.3German Center for Lung Research (DZL), TLRC Heidelberg, Heidelberg, Germany ,grid.5253.10000 0001 0328 4908Department of Oncology, Thoraxklinik and National Center for Tumor Disease (NCT) at Heidelberg University Hospital, Heidelberg, Germany
| | - Petros Christopoulos
- grid.452624.3German Center for Lung Research (DZL), TLRC Heidelberg, Heidelberg, Germany ,grid.5253.10000 0001 0328 4908Department of Oncology, Thoraxklinik and National Center for Tumor Disease (NCT) at Heidelberg University Hospital, Heidelberg, Germany
| | - Holger Sültmann
- grid.5253.10000 0001 0328 4908Division of Cancer Genome Research, German Cancer Research Center, National Center for Tumor Diseases, Heidelberg, Germany ,grid.452624.3German Center for Lung Research (DZL), TLRC Heidelberg, Heidelberg, Germany ,grid.7497.d0000 0004 0492 0584German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
7
|
Fan M, Arai M, Tawada A, Chiba T, Fukushima R, Uzawa K, Shiiba M, Kato N, Tanzawa H, Takiguchi Y. Contrasting functions of the epithelial‑stromal interaction 1 gene, in human oral and lung squamous cell cancers. Oncol Rep 2022; 47:5. [PMID: 34738627 PMCID: PMC8600417 DOI: 10.3892/or.2021.8216] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022] Open
Abstract
The epithelial‑stromal interaction 1 gene (EPSTI1) is known to play multiple roles in the malignant progression of breast cancer and also in some aspects of the immune responses to the tumor. However, the relevance of the gene in the onset/progression of oral squamous cell carcinoma (OSCC) and lung squamous cell carcinoma (LSCC) is not yet known. The present study was aimed at revealing the roles of EPSTI1 in conferring malignant characteristics to OSCC and LSCC, and the underlying mechanisms. Quantitative real‑time polymerase chain reaction (PCR) and western blot analyses demonstrated significant upregulation of EPSTI1 in all four OSCC cell lines (HSC2, HSC3, HSC3‑M3 and HSC4), and significant downregulation of EPST11 in all three LSCC cell lines (LK‑2, EBC‑1 and H226) used in the present study, as compared to the expression levels in the corresponding control cell lines. Both knockdown of EPST11 in OSCC and overexpression of the gene in LSCC suppressed cell proliferation, and induced cell‑cycle arrest in the G1 phase, with upregulation of p21 and downregulation of CDK2 and cyclin D1. Furthermore, these alterations of EPST11 gene expression in the OSCC and LSCC cell lines suppressed the cell migration ability and reversed the EMT phenotype of the tumor cells. Collectively, while EPSTI1 appears to have oncogenic roles in OSCC, it appears to exert tumor‑suppressive roles in LSCC. PCR array analyses revealed some genes whose expression levels were altered along with the modified EPSTI1 expression in both the OSCC and LSCC cell lines. These findings suggest that EPSTI1 may be a therapeutic target for both OSCC and LSCC.
Collapse
Affiliation(s)
- Mengmeng Fan
- Department of Medical Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Makoto Arai
- Department of Medical Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Akinobu Tawada
- Department of Medical Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Tetsuhiro Chiba
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Reo Fukushima
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Katsuhiro Uzawa
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Masashi Shiiba
- Department of Medical Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Naoya Kato
- Department of Gastroenterology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Hideki Tanzawa
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Yuichi Takiguchi
- Department of Medical Oncology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| |
Collapse
|
8
|
Huang W, Li P, Qiu X. [A Literature Review on the Role of TBX5 in Expression and Progression of Lung Cancer: Current Perspectives]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2020; 23:883-888. [PMID: 32810974 PMCID: PMC7583881 DOI: 10.3779/j.issn.1009-3419.2020.102.27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
T-box转录因子(T-box transcription factor gene, TBX)基因涉及器官的发生,TBX5在人的正常心脏和肺组织中表达水平最高。TBX5的缺乏可能导致胸廓发育畸形和膈肌发育异常,其异位表达和过表达会诱导细胞凋亡和抑制细胞生长。既往研究发现了TBX5在食管腺癌、胃癌、结肠癌和乳腺癌的发生和发展中的潜在作用。我们对TBX2亚家族的基因表达和预后之间的关系进行了综述,同时探究TBX5在调控肺癌发生发展机制中的研究进展。虽然TBX5和肺癌发生之间的关系尚不明确,不过TBX5可以显著抑制人体内肿瘤生长,其表达水平和肺癌的进展呈现负相关。由此,TBX5的基因表达水平和甲基化程度是潜在的表证肺癌增殖和转移的生物标志物,具有作为肺癌治疗靶点的潜力。
Collapse
Affiliation(s)
- Weijia Huang
- West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Peiwei Li
- West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Xiaoming Qiu
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Zhou S, Wang Q, Meng Z, Peng J, Zhou Y, Song W, Wang J, Chen S, Sun K. Mutations in fibroblast growth factor (FGF8) and FGF10 identified in patients with conotruncal defects. J Transl Med 2020; 18:283. [PMID: 32664970 PMCID: PMC7362408 DOI: 10.1186/s12967-020-02445-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 07/02/2020] [Indexed: 12/26/2022] Open
Abstract
Background Conotruncal defects (CTDs) are a type of heterogeneous congenital heart diseases (CHDs), but little is known about their etiology. Increasing evidence has demonstrated that fibroblast growth factor (FGF) 8 and FGF10 may be involved in the pathogenesis of CTDs. Methods The variants of FGF8 and FGF10 in unrelated Chinese Han patients with CHDs (n = 585), and healthy controls (n = 319) were investigated. The expression and function of these patient-identified variants were detected to confirm the potential pathogenicity of the non-synonymous variants. The expression of FGF8 and FGF10 during the differentiation of human embryonic stem cells (hESCs) to cardiomyocytes and in Carnegie stage 13 human embryo was also identified. Results Two probable deleterious variants (p.C10Y, p.R184H) of FGF8 and one deletion mutant (p.23_24del) of FGF10 were identified in three patients with CTD. Immunofluorescence suggested that variants did not affect the intracellular localization, whereas ELISA showed that the p.C10Y and p.23_24del variants reduced the amount of secreted FGF8 and FGF10, respectively. Quantitative RT-PCR and western blotting showed that the expression of FGF8 and FGF10 variants was increased compared with wild-type; however, their functions were reduced. And we found that FGF8 and FGF10 were expressed in the outflow tract (OFT) during human embryonic development, and were dynamically expressed during the differentiation of hESCs into cardiomyocytes. Conclusion Our results provided evidence that damaging variants of FGF8 and FGF10 were likely contribute to the etiology of CTD. This discovery expanded the spectrum of FGF mutations and underscored the pathogenic correlation between FGF mutations and CTD.
Collapse
Affiliation(s)
- Shuang Zhou
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Qingjie Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Zhuo Meng
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Jiayu Peng
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Yue Zhou
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Wenting Song
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China
| | - Jian Wang
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| | - Sun Chen
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| | - Kun Sun
- Department of Pediatric Cardiology, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, No. 1665, Kongjiang Road, Yangpu District, Shanghai, 200092, China.
| |
Collapse
|
10
|
Khalil A, Kamar A, Nemer G. Thalidomide-Revisited: Are COVID-19 Patients Going to Be the Latest Victims of Yet Another Theoretical Drug-Repurposing? Front Immunol 2020; 11:1248. [PMID: 32574274 PMCID: PMC7270289 DOI: 10.3389/fimmu.2020.01248] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/18/2020] [Indexed: 12/22/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is a worldwide threatening health issue. The progression of this viral infection occurs in the airways of the lungs with an exaggerated inflammatory response referred to as the "cytokine storm" that can lead to lethal lung injuries. In the absence of an effective anti-viral molecule and until the formulation of a successful vaccine, anti-inflammatory drugs might offer a complementary tool for controlling the associated complications of COVID-19 and thus decreasing the subsequent fatalities. Drug repurposing for several molecules has emerged as a rapid temporary solution for COVID-19. Among these drugs is Thalidomide; a historically emblematic controversial molecule that harbors an FDA approval for treating erythema nodosum leprosum (ENL) and multiple myeloma (MM). Based on just one-case report that presented positive outcomes in a patient treated amongst others with Thalidomide, two clinical trials on the efficacy and safety of Thalidomide in treating severe respiratory complications in COVID-19 patients were registered. Yet, the absence of substantial evidence on Thalidomide usage in that context along with the discontinued studies on the efficiency of this drug in similar pulmonary diseases, might cause a significant obstacle for carrying out further clinical evaluations. Herein, we will discuss the theoretical effectiveness of Thalidomide in attenuating inflammatory complications that are encountered in COVID-19 patients while pinpointing the lack of the needed evidences to move forward with this drug.
Collapse
Affiliation(s)
- Athar Khalil
- Department or Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Amina Kamar
- Vascular Medicine Program, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Georges Nemer
- Department or Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
- Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
11
|
Zhang H, Guo L, Chen J. Rationale for Lung Adenocarcinoma Prevention and Drug Development Based on Molecular Biology During Carcinogenesis. Onco Targets Ther 2020; 13:3085-3091. [PMID: 32341654 PMCID: PMC7166063 DOI: 10.2147/ott.s248436] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/14/2020] [Indexed: 12/18/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common and aggressive subtype of lung cancer with the greatest heterogeneity and aggression. Inspite of recent years’ achievements in understanding the pathogenesis of this disease, as well as the development of new therapeutic approaches, our knowledge on crucial early molecular events during its development is still rudimentary. Recent classification and grading of LUAD has postulated that LUAD does not arise spontaneously, but through a stepwise process from lung adenomatous premalignancy atypical adenomatous hyperplasia to adenocarcinoma in situ, minimally invasive adenocarcinoma, and eventually frankly invasive predominant adenocarcinoma. In this review, we discuss the molecular processes that drive the evolutionary process that results in the formation of LUAD. We also describe how to handle lung premalignancy in clinical settings based on the most recent advances in genomic biology and our own understanding of lung cancer prevention.
Collapse
Affiliation(s)
- Hongming Zhang
- Department of Respiratory Medicine, Yancheng Third People's Hospital, Affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu Province, People's Republic of China
| | - Liting Guo
- Department of Oncology, Ruijin Hospital,affiliated with Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jibei Chen
- Department of Respiratory Medicine, Yancheng Third People's Hospital, Affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, Jiangsu Province, People's Republic of China
| |
Collapse
|
12
|
Khalil A, Kamar A, Nemer G. Thalidomide-Revisited: Are COVID-19 Patients Going to Be the Latest Victims of Yet Another Theoretical Drug-Repurposing? Front Immunol 2020. [PMID: 32574274 DOI: 10.3389/fimmu.2020.01248/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is a worldwide threatening health issue. The progression of this viral infection occurs in the airways of the lungs with an exaggerated inflammatory response referred to as the "cytokine storm" that can lead to lethal lung injuries. In the absence of an effective anti-viral molecule and until the formulation of a successful vaccine, anti-inflammatory drugs might offer a complementary tool for controlling the associated complications of COVID-19 and thus decreasing the subsequent fatalities. Drug repurposing for several molecules has emerged as a rapid temporary solution for COVID-19. Among these drugs is Thalidomide; a historically emblematic controversial molecule that harbors an FDA approval for treating erythema nodosum leprosum (ENL) and multiple myeloma (MM). Based on just one-case report that presented positive outcomes in a patient treated amongst others with Thalidomide, two clinical trials on the efficacy and safety of Thalidomide in treating severe respiratory complications in COVID-19 patients were registered. Yet, the absence of substantial evidence on Thalidomide usage in that context along with the discontinued studies on the efficiency of this drug in similar pulmonary diseases, might cause a significant obstacle for carrying out further clinical evaluations. Herein, we will discuss the theoretical effectiveness of Thalidomide in attenuating inflammatory complications that are encountered in COVID-19 patients while pinpointing the lack of the needed evidences to move forward with this drug.
Collapse
Affiliation(s)
- Athar Khalil
- Department or Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Amina Kamar
- Vascular Medicine Program, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Georges Nemer
- Department or Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
- Genomics and Translational Biomedicine, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
13
|
Perrard J, Morel A, Meznad K, Paget-Bailly P, Dalstein V, Guenat D, Mourareau C, Clavel C, Fauconnet S, Baguet A, Mougin C, Pretet JL. DNA demethylation agent 5azadC downregulates HPV16 E6 expression in cervical cancer cell lines independently of TBX2 expression. Oncol Lett 2019; 19:1074-1081. [PMID: 31897221 DOI: 10.3892/ol.2019.11158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 09/09/2019] [Indexed: 12/18/2022] Open
Abstract
HPV16 is the most carcinogenic human papillomavirus and causes >50% of cervical cancers, the majority of anal cancers and 30% of oropharyngeal squamous cell carcinomas. HPV carcinogenesis relies on the continuous expression of the two main viral oncoproteins E6 and E7 that target >150 cellular proteins. Among them, epigenetic modifiers, including DNA Methyl Transferases (DNMT), are dysregulated, promoting an aberrant methylation pattern in HPV-positive cancer cells. It has been previously reported that the treatment of HPV-positive cervical cancer cells with DNMT inhibitor 5-aza-2'-deoxycytidine (5azadC) caused the downregulation of E6 expression due to mRNA destabilization that was mediated by miR-375. Recently, the T-box transcription factor 2 (TBX2) has been demonstrated to repress HPV LCR activity. In the current study, the role of TBX2 in E6 repression was investigated in HPV16 cervical cancer cell lines following 5azadC treatment. A decrease of E6 expression was accompanied by p53 and p21 restoration. While TBX2 mRNA was upregulated in 5azadC-treated SiHa and Ca Ski cells, TBX2 protein was not detectable. Furthermore, the overexpression of TBX2 protein in cervical cancer cells did not allow the repression of E6 expression. The TBX2 transcription factor is therefore unlikely to be associated with the repression of E6 following 5azadC treatment of SiHa and Ca Ski cells.
Collapse
Affiliation(s)
- Jerome Perrard
- Équipe d'Accueil 3181, University of Bourgogne Franche-Comté, Laboratoire d'Excellence Lipoprotéines et Santé, Prévention et Traitement des Maladies Inflammatoires et du Cancer, 25000 Besançon, France
| | - Adrien Morel
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 112041, Colombia
| | - Koceila Meznad
- Équipe d'Accueil 3181, University of Bourgogne Franche-Comté, Laboratoire d'Excellence Lipoprotéines et Santé, Prévention et Traitement des Maladies Inflammatoires et du Cancer, 25000 Besançon, France
| | - Philippe Paget-Bailly
- Équipe d'Accueil 3181, University of Bourgogne Franche-Comté, Laboratoire d'Excellence Lipoprotéines et Santé, Prévention et Traitement des Maladies Inflammatoires et du Cancer, 25000 Besançon, France
| | - Veronique Dalstein
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 1250 Pathologies Pulmonaires et Plasticité Cellulaire, Université de Reims Champagne-Ardenne, Faculté de Médecine, 51000 Reims, France.,Centre Hospitalier Universitaire Reims, Laboratoire Biopathologie, 51000 Reims, France
| | - David Guenat
- Équipe d'Accueil 3181, University of Bourgogne Franche-Comté, Laboratoire d'Excellence Lipoprotéines et Santé, Prévention et Traitement des Maladies Inflammatoires et du Cancer, 25000 Besançon, France.,Centre National de Référence Papillomavirus, CHU Besançon, Boulevard Alexandre Fleming, 25000 Besançon, France
| | - Celine Mourareau
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 1250 Pathologies Pulmonaires et Plasticité Cellulaire, Université de Reims Champagne-Ardenne, Faculté de Médecine, 51000 Reims, France.,Centre Hospitalier Universitaire Reims, Laboratoire Biopathologie, 51000 Reims, France
| | - Christine Clavel
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 1250 Pathologies Pulmonaires et Plasticité Cellulaire, Université de Reims Champagne-Ardenne, Faculté de Médecine, 51000 Reims, France.,Centre Hospitalier Universitaire Reims, Laboratoire Biopathologie, 51000 Reims, France
| | - Sylvie Fauconnet
- Équipe d'Accueil 3181, University of Bourgogne Franche-Comté, Laboratoire d'Excellence Lipoprotéines et Santé, Prévention et Traitement des Maladies Inflammatoires et du Cancer, 25000 Besançon, France
| | - Aurelie Baguet
- Équipe d'Accueil 3181, University of Bourgogne Franche-Comté, Laboratoire d'Excellence Lipoprotéines et Santé, Prévention et Traitement des Maladies Inflammatoires et du Cancer, 25000 Besançon, France
| | - Christiane Mougin
- Équipe d'Accueil 3181, University of Bourgogne Franche-Comté, Laboratoire d'Excellence Lipoprotéines et Santé, Prévention et Traitement des Maladies Inflammatoires et du Cancer, 25000 Besançon, France.,Centre National de Référence Papillomavirus, CHU Besançon, Boulevard Alexandre Fleming, 25000 Besançon, France
| | - Jean-Luc Pretet
- Équipe d'Accueil 3181, University of Bourgogne Franche-Comté, Laboratoire d'Excellence Lipoprotéines et Santé, Prévention et Traitement des Maladies Inflammatoires et du Cancer, 25000 Besançon, France.,Centre National de Référence Papillomavirus, CHU Besançon, Boulevard Alexandre Fleming, 25000 Besançon, France
| |
Collapse
|
14
|
Epigenetic Suppression of the T-box Subfamily 2 ( TBX2) in Human Non-Small Cell Lung Cancer. Int J Mol Sci 2019; 20:ijms20051159. [PMID: 30866410 PMCID: PMC6429281 DOI: 10.3390/ijms20051159] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/15/2019] [Accepted: 02/18/2019] [Indexed: 12/25/2022] Open
Abstract
(1) The TBX2 subfamily of transcription factors (TBXs 2, 3, 4 and 5) are markedly down-regulated in human non-small cell lung cancer (NSCLC) and exert tumor suppressor effects in lung malignancy. Yet, mechanisms underlying suppressed expression of the TBX2 subfamily in NSCLC are elusive. Here, we interrogated probable epigenetic mechanisms in suppressed expression of the TBX2 subfamily in human NSCLC. (2) TBX2 subfamily gene expression and methylation levels in NSCLC and normal lung tissues were surveyed using publicly available RNA-sequence and genome-wide methylation datasets. Methylation β-values of the four genes were statistically compared between NSCLCs and normal lung tissues, correlated with gene expression levels, and interrogated with clinicopathological variables. Expression and methylation levels of TBXs were quantified in NSCLC cells using real-time PCR and methylation-specific PCR assays, respectively. Effects of the DNA methyltransferase inhibitor 5-azacytidine (Aza) on TBX2 subfamily expression were assessed in NSCLC cells. Impact of TBX2 subfamily expression on Aza-treated cells was evaluated by RNA interference. (3) All four TBXs were significantly hypermethylated in NSCLCs relative to normal lung tissues (p < 0.05). Methylation β-values of the genes, with exception of TBX2, were significantly inversely correlated with corresponding mRNA expression levels (p < 0.05). We found no statistically significant differences in hypermethylation levels of the TBX2 subfamily by clinicopathological features including stage and tobacco history. Expression levels of the TBX genes were overall suppressed in NSCLC cells relative to normal alveolar cells. Members of the subfamily were significantly hypermethylated in all tested NSCLC cell lines relative to normal alveolar cells. Treatment with Aza induced the expression of the TBX2 subfamily concomitant with NSCLC cell growth inhibition. Further, simultaneous knockdown of the four TBX genes markedly reduced anti-growth effects of Aza in NSCLC cells. (4) Our study sheds light on new epigenetic profiles in the molecular pathogenesis of human NSCLC.
Collapse
|
15
|
Khalil A, Dekmak B, Boulos F, Kantrowitz J, Spira A, Fujimoto J, Kadara H, El-Hachem N, Nemer G. Transcriptomic Alterations in Lung Adenocarcinoma Unveil New Mechanisms Targeted by the TBX2 Subfamily of Tumor Suppressor Genes. Front Oncol 2018; 8:482. [PMID: 30425966 PMCID: PMC6218583 DOI: 10.3389/fonc.2018.00482] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/09/2018] [Indexed: 12/22/2022] Open
Abstract
T-box (TBX) transcription factors are evolutionary conserved genes and master transcriptional regulators. In mammals, TBX2 subfamily (TBX2, TBX3, TBX4, and TBX5) genes are expressed in the developing lung bud and tracheae. Our group previously showed that the expression of TBX2 subfamily was significantly high in human normal lungs, but markedly suppressed in lung adenocarcinoma (LUAD). To further elucidate their role in LUAD pathogenesis, we first confirmed abundant expression of protein products of the four members by immunostaining in adult human normal lung tissues. We also found overall suppressed expression of these genes and their corresponding proteins in a panel of human LUAD cell lines. Transient over-expression of each of the genes in human (NCI-H1299), and mouse (MDA-F471) derived lung cancer cells was found to significantly inhibit growth and proliferation as well as induce apoptosis. Genome-wide transcriptomic analyses on NCI-H1299 cells, overexpressing TBX2 gene subfamily, unraveled novel regulatory pathways. These included, among others, inhibition of cell cycle progression but more importantly activation of the histone demethylase pathway. When using a pattern-matching algorithm, we showed that TBX's overexpression mimic molecular signatures from azacitidine treated NCI-H1299 cells which in turn are inversely correlated to expression profiles of both human and murine lung tumors relative to matched normal lung. In conclusion, we showed that the TBX2 subfamily genes play a critical tumor suppressor role in lung cancer pathogenesis through regulating its methylating pattern, making them putative candidates for epigenetic therapy in LUAD.
Collapse
Affiliation(s)
- Athar Khalil
- Departments of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Batoul Dekmak
- Departments of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Fouad Boulos
- Department of Pathology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jake Kantrowitz
- Section of Computational Biomedicine, Boston University, Boston, MA, United States
| | - Avrum Spira
- Section of Computational Biomedicine, Boston University, Boston, MA, United States
| | - Junya Fujimoto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Humam Kadara
- Departments of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Division of Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nehme El-Hachem
- Faculty of Medicine and Genome Innovation Centre, McGill University, Montreal, QC, Canada
| | - Georges Nemer
- Departments of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
16
|
Ren Y, Zhao S, Jiang D, Feng X, Zhang Y, Wei Z, Wang Z, Zhang W, Zhou QF, Li Y, Hou H, Xu Y, Zhou F. Proteomic biomarkers for lung cancer progression. Biomark Med 2018; 12:205-215. [DOI: 10.2217/bmm-2018-0015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: Lung adenocarcinoma (LUAD) and lung squamous-cell carcinoma (LUSC) are two major subtypes of lung cancer and constitute about 70% of all the lung cancer cases. The patient's lifespan and living quality will be significantly improved if they are diagnosed at an early stage and adequately treated. Methods & results: This study comprehensively screened the proteomic dataset of both LUAD and LUSC, and proposed classification models for the progression stages of LUAD and LUSC with accuracies 86.51 and 89.47%, respectively. Discussion & conclusion: A comparative analysis was also carried out on related transcriptomic datasets, which indicates that the proposed biomarkers provide discerning power for accurate stage prediction, and will be improved when larger-scale proteomic quantitative technologies become available.
Collapse
Affiliation(s)
- Yanjiao Ren
- College of Computer Science & Technology, & Key Laboratory of Symbolic Computation & Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, PR China
| | - Shishun Zhao
- Center for Applied Statistical Research, College of Mathematics, Jilin University, Changchun, Jilin 130012, PR China
| | - Dandan Jiang
- Center for Applied Statistical Research, College of Mathematics, Jilin University, Changchun, Jilin 130012, PR China
| | - Xin Feng
- College of Computer Science & Technology, & Key Laboratory of Symbolic Computation & Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, PR China
| | - Yexian Zhang
- College of Computer Science & Technology, & Key Laboratory of Symbolic Computation & Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, PR China
| | - Zhipeng Wei
- College of Computer Science & Technology, & Key Laboratory of Symbolic Computation & Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, PR China
| | - Zhongyu Wang
- College of Computer Science & Technology, & Key Laboratory of Symbolic Computation & Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, PR China
| | - Wenniu Zhang
- College of Computer Science & Technology, & Key Laboratory of Symbolic Computation & Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, PR China
| | - Qing F Zhou
- School of Electrical Engineering & Intelligentization, Dongguan University of Technology, Dongguan 523000, PR China
| | - Yong Li
- Department of Electronic Engineering, Tsinghua University, Beijing 100084, PR China
| | - Hanxu Hou
- School of Electrical Engineering & Intelligentization, Dongguan University of Technology, Dongguan 523000, PR China
| | - Ying Xu
- Computational Systems Biology Lab, Department of Biochemistry & Molecular Biology, University of Georgia, Athens, GA 30602, USA
- College of Computer Science & Technology, & College of Public Health, Jilin University, Changchun, Jilin 130012, PR China
| | - Fengfeng Zhou
- College of Computer Science & Technology, & Key Laboratory of Symbolic Computation & Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, PR China
| |
Collapse
|