1
|
Meng Y, Meng Y, Zheng H, Huo J, Li P, Shan Y, He J. METTL2B m3C RNA transferase: oncogenic role in ovarian cancer progression via regulation of the mTOR/AKT pathway and its link to the tumor immune microenvironment. BMC Cancer 2024; 24:1455. [PMID: 39592997 PMCID: PMC11600782 DOI: 10.1186/s12885-024-13225-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/21/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Aberrant expression of N3-methylcytidine methyltransferase 2B (METTL2B) has been observed in various human malignancies, including those of the prostate, liver, breasts, and bladder. However, its role in ovarian cancer (OC) remains largely unexplored. This research preliminarily investigated METTL2B expression in OC and elucidated the associated molecular mechanisms. METHODS We utilized three publicly available cancer-related databases (Genotype-Tissue Expression, Gene Expression Omnibus, and The Cancer Genome Atlas) to identify gene signatures in patients with OC and normal individuals with a specific focus on METTL2B. The role of METTL2B in OC was evaluated using patient survival data, and its impact on oncogenic behaviors in both cell and animal models, including growth potential, migration, invasion, and the tumor microenvironment, was examined. This assessment was conducted using bioinformatics tools such as Gene Set Cancer Analysis, GeneMANIA, and Tumor Immune Single-cell Hub 2. Additionally, the association between drug sensitivity and METTL2B expression was analyzed using CellMiner. RESULTS METTL2B expression was significantly elevated in OC, highlighting its potential clinical value in the diagnosis and prognosis of OC. Patients with lower METTL2B expression exhibited favorable survival. Furthermore, METTL2B knockdown significantly disrupted oncogenic behaviors in OC cell lines by suppressing the mTOR/AKT signaling pathway. Additionally, bioinformatics-based Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses suggested a close correlation between METTL2B and immune responses. CONCLUSIONS Our research confirmed the upregulation of METTL2B in OC, suggesting its oncogenic function. However, METTL2B expression was negatively correlated with the infiltration scores of multiple immune cells, including cytotoxic cells and T cells, indicating its complex role in the tumor immune microenvironment. These findings highlight the significant clinical value of METTL2B in the diagnosis and prognosis of OC.
Collapse
Affiliation(s)
- Yizi Meng
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, No. 1 Xinmin Road, Changchun, Jilin, 130000, China
| | - Yimei Meng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, No 246, Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Hui Zheng
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, No. 1 Xinmin Road, Changchun, Jilin, 130000, China
| | - Jinru Huo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, No 246, Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang, China
| | - Peiling Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Harbin Medical University, No 246, Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang, China.
| | - Yanhong Shan
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, No. 1 Xinmin Road, Changchun, Jilin, 130000, China.
| | - Jin He
- Department of Obstetrics, Obstetrics and Gynecology Center, The First Hospital of Jilin University, No. 1 Xinmin Road, Changchun, Jilin, 130000, China.
| |
Collapse
|
2
|
Throll P, G Dolce L, Rico-Lastres P, Arnold K, Tengo L, Basu S, Kaiser S, Schneider R, Kowalinski E. Structural basis of tRNA recognition by the m 3C RNA methyltransferase METTL6 in complex with SerRS seryl-tRNA synthetase. Nat Struct Mol Biol 2024; 31:1614-1624. [PMID: 38918637 PMCID: PMC11479938 DOI: 10.1038/s41594-024-01341-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 05/29/2024] [Indexed: 06/27/2024]
Abstract
Methylation of cytosine 32 in the anticodon loop of tRNAs to 3-methylcytosine (m3C) is crucial for cellular translation fidelity. Misregulation of the RNA methyltransferases setting this modification can cause aggressive cancers and metabolic disturbances. Here, we report the cryo-electron microscopy structure of the human m3C tRNA methyltransferase METTL6 in complex with seryl-tRNA synthetase (SerRS) and their common substrate tRNASer. Through the complex structure, we identify the tRNA-binding domain of METTL6. We show that SerRS acts as the tRNASer substrate selection factor for METTL6. We demonstrate that SerRS augments the methylation activity of METTL6 and that direct contacts between METTL6 and SerRS are necessary for efficient tRNASer methylation. Finally, on the basis of the structure of METTL6 in complex with SerRS and tRNASer, we postulate a universal tRNA-binding mode for m3C RNA methyltransferases, including METTL2 and METTL8, suggesting that these mammalian paralogs use similar ways to engage their respective tRNA substrates and cofactors.
Collapse
Affiliation(s)
| | | | - Palma Rico-Lastres
- Institute of Functional Epigenetics, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Katharina Arnold
- Institute of Functional Epigenetics, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Laura Tengo
- European Molecular Biology Laboratory, Grenoble, France
| | - Shibom Basu
- European Molecular Biology Laboratory, Grenoble, France
| | - Stefanie Kaiser
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Frankfurt, Germany
| | - Robert Schneider
- Institute of Functional Epigenetics, Helmholtz Zentrum Munich, Neuherberg, Germany
| | - Eva Kowalinski
- European Molecular Biology Laboratory, Grenoble, France.
| |
Collapse
|
3
|
Wu Z, Zhou R, Li B, Cao M, Wang W, Li X. Methylation modifications in tRNA and associated disorders: Current research and potential therapeutic targets. Cell Prolif 2024; 57:e13692. [PMID: 38943267 PMCID: PMC11503269 DOI: 10.1111/cpr.13692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/14/2024] [Accepted: 06/03/2024] [Indexed: 07/01/2024] Open
Abstract
High-throughput sequencing has sparked increased research interest in RNA modifications, particularly tRNA methylation, and its connection to various diseases. However, the precise mechanisms underpinning the development of these diseases remain largely elusive. This review sheds light on the roles of several tRNA methylations (m1A, m3C, m5C, m1G, m2G, m7G, m5U, and Nm) in diverse biological functions, including metabolic processing, stability, protein interactions, and mitochondrial activities. It further outlines diseases linked to aberrant tRNA modifications, related enzymes, and potential underlying mechanisms. Moreover, disruptions in tRNA regulation and abnormalities in tRNA-derived small RNAs (tsRNAs) contribute to disease pathogenesis, highlighting their potential as biomarkers for disease diagnosis. The review also delves into the exploration of drugs development targeting tRNA methylation enzymes, emphasizing the therapeutic prospects of modulating these processes. Continued research is imperative for a comprehensive comprehension and integration of these molecular mechanisms in disease diagnosis and treatment.
Collapse
Affiliation(s)
- Zhijing Wu
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Ruixin Zhou
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Baizao Li
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Mingyu Cao
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Wenlong Wang
- Department of Breast Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- Clinical Research Center for Breast Cancer in Hunan ProvinceChangshaHunanChina
| | - Xinying Li
- Department of General Surgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South UniversityChangshaHunanChina
| |
Collapse
|
4
|
He J, Hao F, Song S, Zhang J, Zhou H, Zhang J, Li Y. METTL Family in Healthy and Disease. MOLECULAR BIOMEDICINE 2024; 5:33. [PMID: 39155349 PMCID: PMC11330956 DOI: 10.1186/s43556-024-00194-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/02/2024] [Indexed: 08/20/2024] Open
Abstract
Transcription, RNA splicing, RNA translation, and post-translational protein modification are fundamental processes of gene expression. Epigenetic modifications, such as DNA methylation, RNA modifications, and protein modifications, play a crucial role in regulating gene expression. The methyltransferase-like protein (METTL) family, a constituent of the 7-β-strand (7BS) methyltransferase subfamily, is broadly distributed across the cell nucleus, cytoplasm, and mitochondria. Members of the METTL family, through their S-adenosyl methionine (SAM) binding domain, can transfer methyl groups to DNA, RNA, or proteins, thereby impacting processes such as DNA replication, transcription, and mRNA translation, to participate in the maintenance of normal function or promote disease development. This review primarily examines the involvement of the METTL family in normal cell differentiation, the maintenance of mitochondrial function, and its association with tumor formation, the nervous system, and cardiovascular diseases. Notably, the METTL family is intricately linked to cellular translation, particularly in its regulation of translation factors. Members represent important molecules in disease development processes and are associated with patient immunity and tolerance to radiotherapy and chemotherapy. Moreover, future research directions could include the development of drugs or antibodies targeting its structural domains, and utilizing nanomaterials to carry miRNA corresponding to METTL family mRNA. Additionally, the precise mechanisms underlying the interactions between the METTL family and cellular translation factors remain to be clarified.
Collapse
Affiliation(s)
- Jiejie He
- Department of Gynecologic Oncology, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai Province, China
| | - Fengchen Hao
- Department of Gynecologic Oncology, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai Province, China
| | - Shiqi Song
- Department of Gynecologic Oncology, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai Province, China
| | - Junli Zhang
- Department of Gynecologic Oncology, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai Province, China
| | - Hongyu Zhou
- Department of Radiology, Affiliated Hospital of Qinghai University, Xining, 810000, Qinghai Province, China
| | - Jun Zhang
- Department of Urology Surgery, Affiliated Hospital of Qinghai University, No. 29, Tongren Road, West of the City, Xining, 810000, Qinghai Province, China.
| | - Yan Li
- Department of Gynecologic Oncology, Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, No. 29, Tongren Road, West of the City, Xining, 810000, Qinghai Province, China.
| |
Collapse
|
5
|
Yuan W, Zhang R, Lyu H, Xiao S, Guo D, Zhang Q, Ali DW, Michalak M, Chen XZ, Zhou C, Tang J. Dysregulation of tRNA methylation in cancer: Mechanisms and targeting therapeutic strategies. Cell Death Discov 2024; 10:327. [PMID: 39019857 PMCID: PMC11254935 DOI: 10.1038/s41420-024-02097-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024] Open
Abstract
tRNA is the RNA type that undergoes the most modifications among known RNA, and in recent years, tRNA methylation has emerged as a crucial process in regulating gene translation. Dysregulation of tRNA abundance occurs in cancer cells, along with increased expression and activity of tRNA methyltransferases to raise the level of tRNA modification and stability. This leads to hijacking of translation and synthesis of multiple proteins associated with tumor proliferation, metastasis, invasion, autophagy, chemotherapy resistance, and metabolic reprogramming. In this review, we provide an overview of current research on tRNA methylation in cancer to clarify its involvement in human malignancies and establish a theoretical framework for future therapeutic interventions targeting tRNA methylation processes.
Collapse
Affiliation(s)
- Wenbin Yuan
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| | - Hao Lyu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| | - Shuai Xiao
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| | - Dong Guo
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| | - Qi Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, China
| | - Declan William Ali
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, China.
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Hubei Key Laboratory of Industrial Microbiology, School of Life and Health Sciences, Hubei University of Technology, Wuhan, China.
| |
Collapse
|
6
|
Moghbeli M. PI3K/AKT pathway as a pivotal regulator of epithelial-mesenchymal transition in lung tumor cells. Cancer Cell Int 2024; 24:165. [PMID: 38730433 PMCID: PMC11084110 DOI: 10.1186/s12935-024-03357-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
Lung cancer, as the leading cause of cancer related deaths, is one of the main global health challenges. Despite various progresses in diagnostic and therapeutic methods, there is still a high rate of mortality among lung cancer patients, which can be related to the lack of clinical symptoms to differentiate lung cancer from the other chronic respiratory disorders in the early tumor stages. Most lung cancer patients are identified in advanced and metastatic tumor stages, which is associated with a poor prognosis. Therefore, it is necessary to investigate the molecular mechanisms involved in lung tumor progression and metastasis in order to introduce early diagnostic markers as well as therapeutic targets. Epithelial-mesenchymal transition (EMT) is considered as one of the main cellular mechanisms involved in lung tumor metastasis, during which tumor cells gain the metastatic ability by acquiring mesenchymal characteristics. Since, majority of the oncogenic signaling pathways exert their role in tumor cell invasion by inducing the EMT process, in the present review we discussed the role of PI3K/AKT signaling pathway in regulation of EMT process during lung tumor metastasis. It has been reported that the PI3K/AKT acts as an inducer of EMT process through the activation of EMT-specific transcription factors in lung tumor cells. MicroRNAs also exerted their inhibitory effects during EMT process by inhibition of PI3K/AKT pathway. This review can be an effective step towards introducing the PI3K/AKT pathway as a suitable therapeutic target to inhibit the EMT process and tumor metastasis in lung cancer patients.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Bai X, Chen J, Zhang W, Zhou S, Dong L, Huang J, He X. YTHDF2 promotes gallbladder cancer progression and gemcitabine resistance via m6A-dependent DAPK3 degradation. Cancer Sci 2023; 114:4299-4313. [PMID: 37700438 PMCID: PMC10637062 DOI: 10.1111/cas.15953] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/20/2023] [Accepted: 08/23/2023] [Indexed: 09/14/2023] Open
Abstract
N6-methyladenosine (m6A) is the most abundant internal modification in eukaryotic RNA and involved in the carcinogenesis of various malignancies. However, the functions and mechanisms of m6A in gallbladder cancer (GBC) remain unclear. In this study, we investigated the role and underlying mechanism of the RNA-binding protein YT521-B homology domain-containing family protein 2 (YTHDF2), an m6A reader, in GBC. Herein, we detected that YTHDF2 was remarkably upregulated in GBC tissues compared to normal gallbladder tissues. Functionally, YTHDF2 overexpression promoted the proliferation, tumor growth, migration, and invasion of GBC cells while inhibiting the apoptosis in vitro and in vivo. Conversely, YTHDF2 knockdown induced opposite results. Mechanistically, we further investigated the underlying mechanism by integrating RNA immunoprecipitation sequencing (RIP-seq), m6A-modified RIP-seq, and RNA sequencing, which revealed that death-associated protein kinase 3 (DAPK3) is a direct target of YTHDF2. YTHDF2 binds to the 3'-UTR of DAPK3 mRNA and facilitates its degradation in an m6A-dependent manner. DAPK3 inhibition restores the tumor-suppressive phenotype induced by YTHDF2 deficiency. Moreover, the YTHDF2/DAPK3 axis induces the resistance of GBC cells to gemcitabine. In conclusion, we reveal the oncogenic role of YTHDF2 in GBC, demonstrating that YTHDF2 increases the mRNA degradation of the tumor suppressor DAPK3 in an m6A-dependent way, which promotes GBC progression and desensitizes GBC cells to gemcitabine. Our findings provide novel insights into potential therapeutic strategies for GBC.
Collapse
Affiliation(s)
- Xuesong Bai
- Department of General SurgeryState Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical SciencesBeijingChina
| | - Jiemin Chen
- Department of GastroenterologyState Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical SciencesBeijingChina
| | - Wenqin Zhang
- Department of GastroenterologyState Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical SciencesBeijingChina
| | - Shengnan Zhou
- Department of General SurgeryState Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical SciencesBeijingChina
| | - Liangbo Dong
- Department of General SurgeryState Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical SciencesBeijingChina
| | - Jianhao Huang
- Department of General SurgeryState Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical SciencesBeijingChina
| | - Xiaodong He
- Department of General SurgeryState Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
8
|
Wang S, Li H, Liu J, Zhang Q, Xu W, Xiang J, Fang L, Xu P, Li Z. Integrative analysis of m3C associated genes reveals METTL2A as a potential oncogene in breast Cancer. J Transl Med 2022; 20:476. [PMID: 36266694 PMCID: PMC9583565 DOI: 10.1186/s12967-022-03683-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 10/03/2022] [Indexed: 12/02/2022] Open
Abstract
RNA methylation modifications, especially m6A mRNA modification, are known to be extensively involved in tumor development. However, the relationship between N3-methylcytidine (m3C) related genes and tumorigenesis has rarely been studied. In this research, we found that m3C-related genes were expressed at different levels and affected patients’ prognosis across multiple cancer types from The Cancer Genome Atlas and multi-omics levels. Importantly, methyltransferase-like proteins 2A (METTL2A) had a high amplification frequency (~ 7%) in patients with breast invasive carcinoma (BRCA), and its overexpression was an independent predictor of poor overall survival. Enrichment analysis of associated genes revealed that METTL2A may activate DNA synthesis and cell proliferation pathways in BRCA cells. Through drug sensitivity analysis, Trifluridine, PD407824, and Taselisib were shown to be effective drugs for METTL2A-positive BRCA patients. Overall, our research conducts a holistic view of the expression level and prognostic signature of m3C-related genes with multiple malignancies. Importantly, METTL2A has been intensely explored as a potential oncogene in BRCA, to aid the development of potential drug agents for precision therapy in breast cancer patients.
Collapse
Affiliation(s)
- Shuai Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Huiting Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jiheng Liu
- Department of Hematology and Oncology, First Hospital of Changsha, Changsha, Hunan, China
| | - Qianqian Zhang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wei Xu
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Juanjuan Xiang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Li Fang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ping Xu
- Departments of Respiratory and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Zheng Li
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Bohnsack KE, Kleiber N, Lemus-Diaz N, Bohnsack MT. Roles and dynamics of 3-methylcytidine in cellular RNAs. Trends Biochem Sci 2022; 47:596-608. [DOI: 10.1016/j.tibs.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/14/2022] [Accepted: 03/02/2022] [Indexed: 01/02/2023]
|
10
|
Bolatkan A, Asada K, Kaneko S, Suvarna K, Ikawa N, Machino H, Komatsu M, Shiina S, Hamamoto R. Downregulation of METTL6 mitigates cell progression, migration, invasion and adhesion in hepatocellular carcinoma by inhibiting cell adhesion molecules. Int J Oncol 2022; 60:4. [PMID: 34913069 PMCID: PMC8698744 DOI: 10.3892/ijo.2021.5294] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022] Open
Abstract
RNA modifications have attracted increasing interest in recent years because they have been frequently implicated in various human diseases, including cancer, highlighting the importance of dynamic post‑transcriptional modifications. Methyltransferase‑like 6 (METTL6) is a member of the RNA methyltransferase family that has been identified in many cancers; however, little is known about its specific role or mechanism of action. In the present study, we aimed to study the expression levels and functional role of METTL6 in hepatocellular carcinoma (HCC), and further investigate the relevant pathways. To this end, we systematically conducted bioinformatics analysis of METTL6 in HCC using gene expression data and clinical information from a publicly available dataset. The mRNA expression levels of METTL6 were significantly upregulated in HCC tumor tissues compared to that in adjacent non‑tumor tissues and strongly associated with poorer survival outcomes in patients with HCC. CRISPR/Cas9‑mediated knockout of METTL6 in HCC cell lines remarkably inhibited colony formation, cell proliferation, cell migration, cell invasion and cell attachment ability. RNA sequencing analysis demonstrated that knockout of METTL6 significantly suppressed the expression of cell adhesion‑related genes. However, chromatin immunoprecipitation sequencing results revealed no significant differences in enhancer activities between cells, which suggests that METTL6 may regulate genes of interest post‑transcriptionally. In addition, it was demonstrated for the first time that METTL6 was localized in the cytosol as detected by immunofluorescence analysis, which indicates the plausible location of RNA modification mediated by METTL6. Our findings provide further insight into the function of RNA modifications in cancer and suggest a possible role of METTL6 as a therapeutic target in HCC.
Collapse
Affiliation(s)
- Amina Bolatkan
- Department of Diagnostic Imaging and Interventional Oncology, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Ken Asada
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Syuzo Kaneko
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Kruthi Suvarna
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai 400076, India
| | - Noriko Ikawa
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
| | - Hidenori Machino
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Masaaki Komatsu
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
| | - Shuichiro Shiina
- Department of Diagnostic Imaging and Interventional Oncology, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | - Ryuji Hamamoto
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo 103-0027, Japan
- Department of National Cancer Center Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| |
Collapse
|
11
|
tRNA modifications and their potential roles in pancreatic cancer. Arch Biochem Biophys 2021; 714:109083. [PMID: 34785212 DOI: 10.1016/j.abb.2021.109083] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/05/2021] [Accepted: 11/06/2021] [Indexed: 12/23/2022]
Abstract
Since the breakthrough discovery of N6-methyladenosine (m6A), the field of RNA epitranscriptomics has attracted increasing interest in the biological sciences. Transfer RNAs (tRNAs) are extensively modified, and various modifications play a crucial role in the formation and stability of tRNA, which is universally required for accurate and efficient functioning of tRNA. Abnormal tRNA modification can lead to tRNA degradation or specific cleavage of tRNA into fragmented derivatives, thus affecting the translation process and frequently accompanying a variety of human diseases. Increasing evidence suggests that tRNA modification pathways are also misregulated in human cancers. In this review, we summarize tRNA modifications and their biological functions, describe the type and frequency of tRNA modification alterations in cancer, and highlight variations in tRNA-modifying enzymes and the multiple functions that they regulate in different types of cancers. Furthermore, the current implications and the potential role of tRNA modifications in the progression of pancreatic cancer are discussed. Collectively, this review describes recent advances in tRNA modification in cancers and its potential significance in pancreatic cancer. Further study of the mechanism of tRNA modifications in pancreatic cancer may provide possibilities for therapies targeting enzymes responsible for regulating tRNA modifications in pancreatic cancer.
Collapse
|
12
|
Chen R, Zhou J, Liu L, Mao XL, Zhou X, Xie W. Crystal structure of human METTL6, the m 3C methyltransferase. Commun Biol 2021; 4:1361. [PMID: 34862464 PMCID: PMC8642396 DOI: 10.1038/s42003-021-02890-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 11/10/2021] [Indexed: 01/27/2023] Open
Abstract
In tRNA, the epigenetic m3C modification at position 32 in the anticodon loop is highly conserved in eukaryotes, which maintains the folding and basepairing functions of the anticodon. However, the responsible enzymes METTL2 and METTL6 were identified only in recent years. The loss of human METTL6 (hMETTL6) affects the translational process and proteostasis in cells, while in mESCs cells, it leads to defective pluripotency potential. Despite its important functions, the catalytic mechanism of the C32 methylation by this enzyme is poorly understood. Here we present the 1.9 Å high-resolution crystal structure of hMETTL6 bound by SAH. The key residues interacting with the ligand were identified and their roles were confirmed by ITC. We generated a docking model for the hMETTL6-SAH-CMP ternary complex. Interestingly, the CMP molecule binds into a cavity in a positive patch with the base ring pointing to the inside, suggesting a flipped-base mechanism for methylation. We further generated a model for the quaternary complex with tRNASer as a component, which reasonably explained the biochemical behaviors of hMETTL6. Taken together, our crystallographic and biochemical studies provide important insight into the molecular recognition mechanism by METTL6 and may aid in the METTL-based rational drug design in the future.
Collapse
Affiliation(s)
- Ran Chen
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, The Sun Yat-Sen University, Guangzhou, Guangdong, 510006, People's Republic of China
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, Innovation Academy of South China Sea Ecology and Environmental Engineering, South China Sea Institute of Oceanology, Chinese Academy of Sciences, No. 1119, Haibin Road, Nansha District, Guangzhou, 511458, People's Republic of China
| | - Jie Zhou
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, The Sun Yat-Sen University, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Ling Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, The Sun Yat-Sen University, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Xue-Ling Mao
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, People's Republic of China
| | - Xiaolong Zhou
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, People's Republic of China
| | - Wei Xie
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory for Biocontrol, School of Life Sciences, The Sun Yat-Sen University, Guangzhou, Guangdong, 510006, People's Republic of China.
| |
Collapse
|
13
|
Green AJ, Anchang B, Akhtari FS, Reif DM, Motsinger-Reif A. Extending the lymphoblastoid cell line model for drug combination pharmacogenomics. Pharmacogenomics 2021; 22:543-551. [PMID: 34044623 DOI: 10.2217/pgs-2020-0160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Combination drug therapies have become an integral part of precision oncology, and while evidence of clinical effectiveness continues to grow, the underlying mechanisms supporting synergy are poorly understood. Immortalized human lymphoblastoid cell lines (LCLs) have been proven as a particularly useful, scalable and low-cost model in pharmacogenetics research, and are suitable for elucidating the molecular mechanisms of synergistic combination therapies. In this review, we cover the advantages of LCLs in synergy pharmacogenomics and consider recent studies providing initial evidence of the utility of LCLs in synergy research. We also discuss several opportunities for LCL-based systems to address gaps in the research through the expansion of testing regimens, assessment of new drug classes and higher-order combinations, and utilization of integrated omics technologies.
Collapse
Affiliation(s)
- Adrian J Green
- Department of Biological Sciences & the Bioinformatics Research Center, NC State University, Raleigh, NC, USA
| | - Benedict Anchang
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Farida S Akhtari
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, USA
| | - David M Reif
- Department of Biological Sciences & the Bioinformatics Research Center, NC State University, Raleigh, NC, USA
| | - Alison Motsinger-Reif
- Biostatistics & Computational Biology Branch, National Institute of Environmental Health Sciences, Durham, NC, USA
| |
Collapse
|
14
|
Dai X, Jiang W, Ma L, Sun J, Yan X, Qian J, Wang Y, Shi Y, Ni S, Yao N. A metabolism-related gene signature for predicting the prognosis and therapeutic responses in patients with hepatocellular carcinoma. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:500. [PMID: 33850897 PMCID: PMC8039687 DOI: 10.21037/atm-21-927] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Hepatocellular carcinoma (HCC) often has an insidious onset and rapid progression. Often, when the disease is first diagnosed, the opportune time for surgical intervention has already lapsed. In addition, the effects of systemic treatment is relatively unsatisfactory. Metabolic reprogramming is one of the hallmarks of cancer. This study aimed to identify a set of genes related to metabolism to construct a predictive model for the prognosis of HCC. Methods The transcriptomic and clinical data of 352 HCC patients were obtained from The Cancer Genome Atlas (TCGA) Liver Hepatocellular Carcinoma (LIHC) dataset and divided into a training cohort (n=212) and a testing cohort (n=140) at a ratio of 6:4. Univariate Cox regression analysis and the LASSO Cox regression model were used to identify 5 genes to establish a risk score for predicting the prognosis of HCC patients. Subsequently, the molecular characteristics of the model were assessed and the ability of the model to predict the tumor immune microenvironment and patient response to immunotherapy and chemotherapy was also examined. Results The risk score model was constructed based on the five genes, methyltransferase-like protein 6 (METTL6), RNA polymerase III subunit G (POLR3G), phosphoribosyl pyrophosphate amidotransferase (PPAT), SET Domain Bifurcated 2 (SETDB2), and suppressor of variegation 3-9 homolog 2 (SUV39H2). The Kaplan-Meier survival analysis and time-dependent receiver operating characteristic (ROC) curves demonstrated that high-risk patients had a poorer overall survival (OS) compared to low-risk patients. he nomogram score had a better predictive ability compared to the common factors. Our results finally showed that high-risk cases were associated with cell proliferation and cell cycle related gene sets, high tumor protein P53 (TP53) mutation rate, suppressive immunity and increased sensitivity to cisplatin, gemcitabine and docetaxel. Meanwhile, low-risk cases were associated with cell cycle and immune response related pathways, low TP53 mutation rate, active immunity and more benefit from immunotherapy. Conclusions This study provided novel insights into the role of metabolism-related genes in HCC, and demonstrated that our model could be a promising prognostic biomarker for distinguishing the molecular and immune characteristics and inferring the potential response to chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Xiaoyan Dai
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China.,Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Wei Jiang
- Department of Neurology, the Second People's Hospital of Wuxi, Wuxi, China
| | - Liang Ma
- Department of Chemotherapy, First People's Hospital of Yancheng, Yancheng, China
| | - Jie Sun
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaodi Yan
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jing Qian
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yan Wang
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yu Shi
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Shujie Ni
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| | - Ninghua Yao
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
15
|
Thomas PL, Madubata CJ, Aldrich MC, Lee MM, Owonikoko TK, Minna JD, Rudin CM, Sage J, Lovly CM. A Call to Action: Dismantling Racial Injustices in Preclinical Research and Clinical Care of Black Patients Living with Small Cell Lung Cancer. Cancer Discov 2021; 11:240-244. [PMID: 33318034 PMCID: PMC7858238 DOI: 10.1158/2159-8290.cd-20-1592] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Small cell lung cancer (SCLC) is an aggressive disease with dismal survival rates and limited therapeutic options. SCLC development is strongly associated with exposure to tobacco carcinogens. However, additional genetic and environmental risk factors that contribute to SCLC pathogenesis are beginning to emerge. Here, we specifically assess disparities pertaining to SCLC in Black populations. In contrast to non-small cell lung cancer, preliminary data suggest that Black individuals may actually be at a lower risk of developing SCLC relative to white individuals. This difference remains unexplained but urgently needs to be verified in larger data sets, because it could provide important new insights and approaches to understanding this recalcitrant tumor. Importantly, little biological information exists on SCLC in Black individuals, and few patient-derived preclinical SCLC models from diverse ancestries are available in the laboratory. Unfortunately, we note strikingly low numbers of Black participants in clinical trials testing new treatments for SCLC. Evidence further indicates that care for patients with SCLC may vary between communities with a large fraction of Black patients and those without. Together, these observations underscore the need to better investigate genetic, environmental, and socioeconomic factors associated with SCLC development, preclinical research, clinical care, and outcomes.
Collapse
Affiliation(s)
- Portia L Thomas
- Department of Microbiology, Immunology, and Physiology, School of Medicine, Meharry Medical College, Nashville, Tennessee
- School of Graduate Studies and Research, Meharry Medical College, Nashville, Tennessee
| | | | - Melinda C Aldrich
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Taofeek K Owonikoko
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - John D Minna
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas
- Hamon Center for Therapeutic Oncology Research, Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Julien Sage
- Department of Pediatrics, Stanford University, Stanford, California.
- Department of Genetics, Stanford University, Stanford, California
| | - Christine M Lovly
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt University Medical Center, Nashville, Tennessee.
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
16
|
Zhou L, Zhu J, Chen W, Jiang Y, Hu T, Wang Y, Ye X, Zhan M, Ji C, Xu Z, Wang X, Gu Y, Jia L. Induction of NEDD8-conjugating enzyme E2 UBE2F by platinum protects lung cancer cells from apoptosis and confers to platinum-insensitivity. Cell Death Dis 2020; 11:975. [PMID: 33184273 PMCID: PMC7665193 DOI: 10.1038/s41419-020-03184-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 12/25/2022]
Abstract
Platinum is a widely used first-line chemotherapy in treating non-small cell lung cancer of adenocarcinoma. Unfortunately, platinum resistance leads to relapse and therapeutic failure, enabling the development of platinum-sensitization strategies to be of great clinical significance. Here, we report that the upregulation of the NEDD8-conjugating enzyme UBE2F is an important way for lung cancer cells to escape platinum-induced cell apoptosis, which confers to insensitivity to platinum-based chemotherapy. Mechanistically, platinum treatment impairs the complex formation for proteasome-mediated UBE2F degradation, evidenced by the weaker association between UBE2F and Ring-box protein 1 (RBX1), an essential component of Cullin-Ring E3 ligases (CRLs), thus leading to the accumulation of UBE2F. The accumulated UBE2F promotes the neddylation levels and activity of Cullin5, in accord with the lower expression of pro-apoptotic protein NOXA, a well-known substrate of Cullin-Ring E3 ligase 5 (CRL5). Additionally, knockout of UBE2F significantly sensitizes lung cancer cells to platinum treatment by enhancing the protein levels of NOXA and subsequently promoting cell apoptosis. Our observations uncover a previously unknown regulatory mechanism of UBE2F stability upon platinum chemotherapy and suggest that UBE2F might be a novel therapy target for sensitizing lung cancer cells to platinum-based chemotherapy.
Collapse
Affiliation(s)
- Lisha Zhou
- Department of Basic Medical Science, Medical College, Taizhou University, 318000, Taizhou, Zhejiang, China.
| | - Jin Zhu
- Department of Surgical Oncology, Jiangxi Cancer Hospital, 330029, Nanchang, Jiangxi, China
| | - Wangyang Chen
- Department of Basic Medical Science, Medical College, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Yanyu Jiang
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China
| | - Tao Hu
- Department of Basic Medical Science, Medical College, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Yinxia Wang
- Department of Basic Medical Science, Medical College, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Xiaoling Ye
- Department of Basic Medical Science, Medical College, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Mengxi Zhan
- Department of Basic Medical Science, Medical College, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Chenghao Ji
- Department of Basic Medical Science, Medical College, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Zhuoming Xu
- Department of Basic Medical Science, Medical College, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Xinran Wang
- Department of Basic Medical Science, Medical College, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Yuanlong Gu
- Taizhou Municipal Hospital, Clinical Medical College, Taizhou University, 318000, Taizhou, Zhejiang, China
| | - Lijun Jia
- Cancer Institute of Traditional Chinese Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 200032, Shanghai, China.
| |
Collapse
|
17
|
Ignatova VV, Kaiser S, Ho JSY, Bing X, Stolz P, Tan YX, Lee CL, Gay FPH, Lastres PR, Gerlini R, Rathkolb B, Aguilar-Pimentel A, Sanz-Moreno A, Klein-Rodewald T, Calzada-Wack J, Ibragimov E, Valenta M, Lukauskas S, Pavesi A, Marschall S, Leuchtenberger S, Fuchs H, Gailus-Durner V, de Angelis MH, Bultmann S, Rando OJ, Guccione E, Kellner SM, Schneider R. METTL6 is a tRNA m 3C methyltransferase that regulates pluripotency and tumor cell growth. SCIENCE ADVANCES 2020; 6:eaaz4551. [PMID: 32923617 PMCID: PMC7449687 DOI: 10.1126/sciadv.aaz4551] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 07/14/2020] [Indexed: 06/11/2023]
Abstract
Recently, covalent modifications of RNA, such as methylation, have emerged as key regulators of all aspects of RNA biology and have been implicated in numerous diseases, for instance, cancer. Here, we undertook a combination of in vitro and in vivo screens to test 78 potential methyltransferases for their roles in hepatocellular carcinoma (HCC) cell proliferation. We identified methyltransferase-like protein 6 (METTL6) as a crucial regulator of tumor cell growth. We show that METTL6 is a bona fide transfer RNA (tRNA) methyltransferase, catalyzing the formation of 3-methylcytidine at C32 of specific serine tRNA isoacceptors. Deletion of Mettl6 in mouse stem cells results in changes in ribosome occupancy and RNA levels, as well as impaired pluripotency. In mice, Mettl6 knockout results in reduced energy expenditure. We reveal a previously unknown pathway in the maintenance of translation efficiency with a role in maintaining stem cell self-renewal, as well as impacting tumor cell growth profoundly.
Collapse
Affiliation(s)
- Valentina V. Ignatova
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Steffen Kaiser
- Chemical Faculty, Ludwig-Maximilians Universität München, Munich, Germany
| | - Jessica Sook Yuin Ho
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xinyang Bing
- University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Paul Stolz
- Department of Biology II, Human Biology and BioImaging, Ludwig-Maximilians Universität München, Munich, Germany
| | - Ying Xim Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Chee Leng Lee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Florence Pik Hoon Gay
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Palma Rico Lastres
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Raffaele Gerlini
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-University München, Munich, Germany
| | - Antonio Aguilar-Pimentel
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Adrián Sanz-Moreno
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Tanja Klein-Rodewald
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Julia Calzada-Wack
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Emil Ibragimov
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Magdalena Valenta
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Saulius Lukauskas
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Andrea Pavesi
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Susan Marschall
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Stefanie Leuchtenberger
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Valerie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Chair of Experimental Genetics, School of Life Science Weihenstephan, Technische Universität München, 85354 Freising, Germany
| | - Sebastian Bultmann
- Department of Biology II, Human Biology and BioImaging, Ludwig-Maximilians Universität München, Munich, Germany
| | - Oliver J. Rando
- University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Ernesto Guccione
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Robert Schneider
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Faculty of Biology, Ludwig-Maximilians Universität München, Munich, Germany
| |
Collapse
|
18
|
Zayas J, Qin S, Yu J, Ingle JN, Wang L. Functional genomics based on germline genome-wide association studies of endocrine therapy for breast cancer. Pharmacogenomics 2020; 21:615-625. [PMID: 32539536 DOI: 10.2217/pgs-2019-0191] [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: 11/21/2022] Open
Abstract
Breast cancer is the most common invasive cancer in women worldwide. Functional follow-up of breast cancer genome-wide association studies has led to the discovery of genes that regulate endocrine therapy response in a SNP- and drug-dependent manner. Here, we will present four examples in which functional genomic studies from breast cancer clinical trials led to novel pharmacogenomic insights and molecular mechanisms of selective estrogen receptor modulators and aromatase inhibitors. The approach utilized for studying genetic variability described in this review offers substantial potential for meaningful discoveries that move the field toward precision medicine for patients.
Collapse
Affiliation(s)
- Jacqueline Zayas
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic School of Medicine & Mayo Clinic Medical Scientist Training Program, Rochester, MN 55905, USA
| | - Sisi Qin
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jia Yu
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Liewei Wang
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
19
|
Jia R, Wang C. MiR-29b-3p Reverses Cisplatin Resistance by Targeting COL1A1 in Non-Small-Cell Lung Cancer A549/DDP Cells. Cancer Manag Res 2020; 12:2559-2566. [PMID: 32368137 PMCID: PMC7170551 DOI: 10.2147/cmar.s246625] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 03/16/2020] [Indexed: 12/14/2022] Open
Abstract
Objective To investigate the expression of miR-29b-3p in tissues and cells of non-small-cell lung cancer (NSCLC) and its effect on cisplatin resistance in NSCLC cells and its mechanism. Methods The mRNA expression of miR-29b-3p and COL1A1 in NSCLC tissue, cell line A549 and cisplatin-resistant cell line A549/DDP was detected by RT-qPCR. MiR-29b-3p Mimics was transfected into A549/DDP cells, and the cell viability, proliferation, apoptosis and related protein expression were detected by CCK-8, flow cytometry and Western blot. Also, luciferase reporter gene assay was used to verify the targeting relationship between miR-29b-3p and COL1A1. Moreover, COL1A1 overexpression lentivirus and miR-29b-3p mimics (Mimics+COL1A1) were simultaneously transfected into A549/DDP cells, and then the cell viability and related protein expression were measured. Results In NSCLC tissue and its cell line, miR-29b-3p was downregulated and COL1A1 was upregulated (P<0.05). After A549/DDP cell was transfected by mimics, its cell viability and proliferation rate decreased, apoptosis rate and the expression of tumor suppressor gene PTEN and apoptosis-related protein BAX were increased (P<0.05), which could be reversed by Mimics+COL1A1 co-transfection. Luciferase reporter gene assay indicated that COL1A1 was the target gene of miR-29b-3p. Conclusion All in all, miR-29b-3p can reverse the cisplatin resistance of A549/DDP cells by inhibiting the expression of COL1A1 gene and increasing the expression of PTEN and BAX.
Collapse
Affiliation(s)
- Rui Jia
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300000, People's Republic of China.,Department of Thoracic Surgery, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, People's Republic of China
| | - Changli Wang
- Department of Lung Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300000, People's Republic of China
| |
Collapse
|
20
|
Mao CX, Li M, Zhang W, Zhou HH, Yin JY, Liu ZQ. Pharmacogenomics for the efficacy of platinum-based chemotherapy: Old drugs, new integrated perspective. Biomed Pharmacother 2020; 126:110057. [PMID: 32145590 DOI: 10.1016/j.biopha.2020.110057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/16/2020] [Accepted: 02/25/2020] [Indexed: 01/16/2023] Open
Abstract
Platinum-based chemotherapy remains the cornerstone of treatment for many malignancies. However, although therapeutic efficiency varies greatly among individuals, there is a lack of pharmacogenomic biomarkers that can be used in clinical settings to identify chemosensitive patients and allow stratification. With the development of high-throughput screening techniques and systems biology approaches, a growing body of evidence has shown that platinum resistance is a multifactorial, multi-dimensional, dynamic process incorporating genetic background, tumor evolution and gut microbes. This review critically summarizes potential pharmacogenomic biomarkers for predicting the efficacy of platinum drugs and provides a comprehensive, time-varying perspective that integrates multiple markers.
Collapse
Affiliation(s)
- Chen-Xue Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China
| | - Min Li
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China
| | - Ji-Ye Yin
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China.
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, PR China.
| |
Collapse
|
21
|
Esteve-Puig R, Bueno-Costa A, Esteller M. Writers, readers and erasers of RNA modifications in cancer. Cancer Lett 2020; 474:127-137. [PMID: 31991154 DOI: 10.1016/j.canlet.2020.01.021] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/09/2020] [Accepted: 01/21/2020] [Indexed: 02/08/2023]
Abstract
Although cancer was originally considered a disease driven only by genetic mutations, it has now been proven that it is also an epigenetic disease driven by DNA hypermethylation-associated silencing of tumor suppressor genes and aberrant histone modifications. Very recently, a third component has emerged: the so-called epitranscriptome understood as the chemical modifications of RNA that regulate and alter the activity of RNA molecules. In this regard, the study of genetic and epigenetic disruption of the RNA-modifying proteins is gaining momentum in advancing our understanding of cancer biology. Furthermore, the development of epitranscriptomic anticancer drugs could lead to new promising and unexpected therapeutic strategies for oncology in the coming years.
Collapse
Affiliation(s)
- Rosaura Esteve-Puig
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Alberto Bueno-Costa
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain
| | - Manel Esteller
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain; Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain; Institucio Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Catalonia, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain.
| |
Collapse
|
22
|
Tao X, Yuan C, Zheng D, Ye T, Pan Y, Zhang Y, Xiang J, Hu H, Chen H, Sun Y. Outcomes comparison between neoadjuvant chemotherapy and adjuvant chemotherapy in stage IIIA non-small cell lung cancer patients. J Thorac Dis 2019; 11:1443-1455. [PMID: 31179087 DOI: 10.21037/jtd.2019.03.42] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background A neoadjuvant chemotherapy (NCT) is a feasible second-option other than an adjuvant chemotherapy (ACT); however, no definite conclusions have been drawn about whether or not a NCT is associated with better clinical outcomes for IIIA non-small cell lung cancer (NSCLC) patients. Methods We reviewed 68 clinical IIIA NSCLC patients who received preoperative chemotherapy (NCT group), and 535 pathological IIIA NSCLC patients who received ACT after surgery (ACT group). After a 1:1 propensity score matching (PSM), we compared the relapse-free survival (RFS) and overall survival (OS) rates as the long-term clinical outcomes, and hospital stay, surgery duration, postoperative complications as the short-term clinical outcomes. To evaluate the predictive value of the NCT response, we also assessed the response evaluation criteria in solid tumors (RECIST) response to NCT. Results There was no significant difference in RFS or OS between the NCT group and ACT group (RFS: P=0.1138; OS: P=0.4234). On multivariate analysis, large cell lung carcinoma (P=0.0264), bilobectomy (P=0.0039) and clinical N2 stage (P=0.0309) were independent predictive factors of a worse OS. Short-term clinical outcomes including the hospital stay and postoperative complications had no statistically distinct difference between the ACT and NCT groups. Meanwhile, the OS of the partial response (PR) patients group was better than the stable disease/progressive disease (SD/PD) (P=0.0205) and ACT (P=0.0442) group, but none of the clinical features we tested was found to be a predictive factor for a PR response. Conclusions There was a non-significant difference between the long-term and short-term clinical outcomes of both NCT and ACT. The OS of PR patients was better than SD/PD and ACT, indicating that NCT response acts as a predictor for a higher long-term survival rate.
Collapse
Affiliation(s)
- Xiaoting Tao
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chongze Yuan
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Difan Zheng
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ting Ye
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yunjian Pan
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yawei Zhang
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiaqing Xiang
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Hong Hu
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Haiquan Chen
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yihua Sun
- Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
23
|
de Crécy-Lagard V, Boccaletto P, Mangleburg CG, Sharma P, Lowe TM, Leidel SA, Bujnicki JM. Matching tRNA modifications in humans to their known and predicted enzymes. Nucleic Acids Res 2019; 47:2143-2159. [PMID: 30698754 PMCID: PMC6412123 DOI: 10.1093/nar/gkz011] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/28/2018] [Accepted: 01/10/2019] [Indexed: 12/25/2022] Open
Abstract
tRNA are post-transcriptionally modified by chemical modifications that affect all aspects of tRNA biology. An increasing number of mutations underlying human genetic diseases map to genes encoding for tRNA modification enzymes. However, our knowledge on human tRNA-modification genes remains fragmentary and the most comprehensive RNA modification database currently contains information on approximately 20% of human cytosolic tRNAs, primarily based on biochemical studies. Recent high-throughput methods such as DM-tRNA-seq now allow annotation of a majority of tRNAs for six specific base modifications. Furthermore, we identified large gaps in knowledge when we predicted all cytosolic and mitochondrial human tRNA modification genes. Only 48% of the candidate cytosolic tRNA modification enzymes have been experimentally validated in mammals (either directly or in a heterologous system). Approximately 23% of the modification genes (cytosolic and mitochondrial combined) remain unknown. We discuss these 'unidentified enzymes' cases in detail and propose candidates whenever possible. Finally, tissue-specific expression analysis shows that modification genes are highly expressed in proliferative tissues like testis and transformed cells, but scarcely in differentiated tissues, with the exception of the cerebellum. Our work provides a comprehensive up to date compilation of human tRNA modifications and their enzymes that can be used as a resource for further studies.
Collapse
Affiliation(s)
- Valérie de Crécy-Lagard
- Department of Microbiology and Cell Sciences, University of Florida, Gainesville, FL 32611, USA
- Cancer and Genetic Institute, University of Florida, Gainesville, FL 32611, USA
| | - Pietro Boccaletto
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology, ul. Trojdena 4, 02-109 Warsaw, Poland
| | - Carl G Mangleburg
- Department of Microbiology and Cell Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Puneet Sharma
- Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany
- Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
| | - Todd M Lowe
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Sebastian A Leidel
- Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany
- Cells-in-Motion Cluster of Excellence, University of Muenster, 48149 Muenster, Germany
- Research Group for RNA Biochemistry, Institute of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
| | - Janusz M Bujnicki
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology, ul. Trojdena 4, 02-109 Warsaw, Poland
- Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, ul. Umultowska 89, 61-614 Poznań, Poland
| |
Collapse
|
24
|
CHD1L contributes to cisplatin resistance by upregulating the ABCB1-NF-κB axis in human non-small-cell lung cancer. Cell Death Dis 2019; 10:99. [PMID: 30718500 PMCID: PMC6362241 DOI: 10.1038/s41419-019-1371-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 11/26/2018] [Accepted: 01/09/2019] [Indexed: 12/13/2022]
Abstract
Chromodomain helicase/ATPase DNA binding protein 1-like gene (CHD1L) is a recently identified gene associated with malignant tumor progression and patient chemotherapy resistance in human hepatocellular carcinoma (HCC). Previously, we found an association between CHD1L overexpression and poor patient survival in non-small-cell lung cancer (NSCLC). However, little is known about the relationship between CHD1L expression and chemotherapy resistance of NSCLC. By employing immunohistochemistry, we analyzed the expression of CHD1L in NSCLC samples and elucidated the roles and mechanism of CHD1L in NSCLC chemoresistance. We found that the increased expression of CHD1L is positively correlated with a shorter survival time of patients who had received chemotherapy after surgery. We also found that the expression of CHD1L was increased after cisplatin treatment in A549 cells. Conversely, the depletion of CHD1L in cisplatin-resistance cells increased the cell sensitivity to cisplatin, indicating that CHD1L plays a critical role in cisplatin resistance of NSCLC cells. Importantly, we identified the ATP-Binding Cassette Sub-Family B Member (ABCB1) gene as a potential downstream target of CHD1L in NSCLC cells. Knocking down ABCB1 coupled with ectopic expression of CHD1L enhanced the effect of cisplatin on NSCLC cells apoptosis. In addition, overexpressed CHD1L increase the transcription of c-Jun which targeted directly to the promoter of ABCB1. Our data demonstrate that CHD1L could induce cisplatin resistance in NSCLC via c-Jun-ABCB1-NF-κB axis, and may serve as a novel predictive marker and the potential therapeutic target for cisplatin resistance in NSCLC.
Collapse
|
25
|
Song X, Kong F, Zong Z, Ren M, Meng Q, Li Y, Sun Z. miR-124 and miR-142 enhance cisplatin sensitivity of non-small cell lung cancer cells through repressing autophagy via directly targeting SIRT1. RSC Adv 2019; 9:5234-5243. [PMID: 35514612 PMCID: PMC9060797 DOI: 10.1039/c8ra09914f] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 02/04/2019] [Indexed: 12/18/2022] Open
Abstract
Background: Drug resistance is a major obstacle in the treatment of non-small cell lung cancer (NSCLC). Recently, miRNAs are reported to be involved in the drug resistance of NSCLC. The roles of miR-124 and miR-142 in the multidrug resistance of NSCLC cells have been reported. However, the underlying mechanism by which miR-124 and miR-142 regulate resistance to cisplatin (CDDP) remains unknown. Methods: The expressions of miR-124, miR-142 and sirtuin 1 (SIRT1) in CDDP-sensitive and CDDP-resistant NSCLC tissues and cells were detected by qRT-PCR and western blot. IC50 value and cell proliferation were determined by MTT assay. Apoptosis was assessed by flow cytometry analysis. Autophagy was evaluated by western blot analysis of the protein levels of LC3-I, LC3-II and p62, and FITC-LC3 punctate formation assay. The interaction between miR-124 or miR-142 and SIRT1 was determined by luciferase reporter, RNA immunoprecipitation (RIP) and western blot assays. A tumor xenograft was performed to further validate the role of miR-124 and miR-142 in the sensitivity of CDDP-resistant NSCLC to cisplatin. Results: miR-124 and miR-142 were downregulated, while SIRT1 was upregulated in CDDP-resistant NSCLC tissues and cells compared to CDDP-sensitive groups. Functionally, overexpression of miR-124 and miR-142 or SIRT1 silencing enhanced the CDDP sensitivity of H1299/CDDP cells via suppressing autophagy, as evidenced by the reduced LC3-II/LC3-I radio, elevated p62 protein, and suppressed FITC-LC3 punctate formation in H1299/CDDP cells. miR-124 and miR-142 were demonstrated to co-target SIRT1. Re-expression of SIRT1 overturned miR-124 and miR-142-mediated chemosensitivity in H1299/CDDP cells via triggering autophagy. Conclusion: miR-124 and miR-142 enhance the cytotoxic effect of CDDP through repressing autophagy via targeting SIRT1 in CDDP-resistant NSCLC cells. Drug resistance is a major obstacle in the treatment of non-small cell lung cancer (NSCLC).![]()
Collapse
Affiliation(s)
- Xiang Song
- Department of Thoracic Surgery
- Cangzhou Central Hospital
- Cangzhou 061000
- China
| | - Fanyi Kong
- Department of Thoracic Surgery
- Cangzhou Central Hospital
- Cangzhou 061000
- China
| | - Zhenfeng Zong
- Department of Thoracic Surgery
- Cangzhou Central Hospital
- Cangzhou 061000
- China
| | - Mingming Ren
- Department of Thoracic Surgery
- Cangzhou Central Hospital
- Cangzhou 061000
- China
| | - Qingjun Meng
- Department of Thoracic Surgery
- Cangzhou Central Hospital
- Cangzhou 061000
- China
| | - Yanguang Li
- Department of Thoracic Surgery
- Cangzhou Central Hospital
- Cangzhou 061000
- China
| | - Zhen Sun
- Department of Thoracic Surgery
- Cangzhou Central Hospital
- Cangzhou 061000
- China
| |
Collapse
|
26
|
Valdés MG, Galván-Femenía I, Ripoll VR, Duran X, Yokota J, Gavaldà R, Rafael-Palou X, de Cid R. Pipeline design to identify key features and classify the chemotherapy response on lung cancer patients using large-scale genetic data. BMC SYSTEMS BIOLOGY 2018; 12:97. [PMID: 30458782 PMCID: PMC6245589 DOI: 10.1186/s12918-018-0615-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
BACKGROUND During the last decade, the interest to apply machine learning algorithms to genomic data has increased in many bioinformatics applications. Analyzing this type of data entails difficulties for managing high-dimensional data, class imbalance for knowledge extraction, identifying important features and classifying individuals. In this study, we propose a general framework to tackle these challenges with different machine learning algorithms and techniques. We apply the configuration of this framework on lung cancer patients, identifying genetic signatures for classifying response to drug treatment response. We intersect these relevant SNPs with the GWAS Catalog of the National Human Genome Research Institute and explore the Regulomedb, GTEx databases for functional analysis purposes. RESULTS The machine learning based solution proposed in this study is a scalable and flexible alternative to the classical uni-variate regression approach to analyze large-scale data. From 36 experiments executed using the machine learning framework design, we obtain good classification performance from the top 5 models with the highest cross-validation score and the smallest standard deviation. One thousand two hundred twenty four SNPs corresponding to the key features from the top 20 models (cross validation F1 mean >= 0.65) were compared with the GWAS Catalog finding no intersection with genome-wide significant reported hits. From these, new genetic signatures in MAE, CEP104, PRKCZ and ADRB2 show relevant biological regulatory functionality related to lung physiology. CONCLUSIONS We have defined a machine learning framework using data with an unbalanced large data-set of SNP-arrays and imputed genotyping data from a pharmacogenomics study in lung cancer patients subjected to first-line platinum-based treatment. This approach found genome signals with no genome-wide significance in the uni-variate regression approach (GWAS Catalog) that are valuable for classifying patients, only few of them with related biological function. The effect results of these variants can be explained by the recently proposed omnigenic model hypothesis, which states that complex traits can be influenced mostly by genes outside not only by the "core genes", mainly found by the genome-wide significant SNPs, but also by the rest of genes outside of the "core pathways" with apparent unrelated biological functionality.
Collapse
Affiliation(s)
- María Gabriela Valdés
- Eurecat. Technology Centre of Catalonia, Av. Diagonal 177, 9th floor, Barcelona, 08018 Spain
| | - Iván Galván-Femenía
- PMPPC-IGTP. Programa de Medicina Predictiva i Personalitzada del Càncer - Institut Germans Trias i Pujol (IGTP). Genomes for Life - GCAT lab Group, Badalona, Spain
| | - Vicent Ribas Ripoll
- Eurecat. Technology Centre of Catalonia, Av. Diagonal 177, 9th floor, Barcelona, 08018 Spain
| | - Xavier Duran
- PMPPC-IGTP. Programa de Medicina Predictiva i Personalitzada del Càncer - Institut Germans Trias i Pujol (IGTP). Genomes for Life - GCAT lab Group, Badalona, Spain
| | - Jun Yokota
- PMPPC-IGTP. Programa de Medicina Predictiva i Personalitzada del Càncer - Institut Germans Trias i Pujol (IGTP). CancerGenome Biology, Badalona, Spain
| | - Ricard Gavaldà
- Universitat Politècnica de Catalunya, Barcelona, Spain
- Barcelona Graduate School of Mathematics, BGSMath, Barcelona, Spain
| | - Xavier Rafael-Palou
- Eurecat. Technology Centre of Catalonia, Av. Diagonal 177, 9th floor, Barcelona, 08018 Spain
| | - Rafael de Cid
- PMPPC-IGTP. Programa de Medicina Predictiva i Personalitzada del Càncer - Institut Germans Trias i Pujol (IGTP). Genomes for Life - GCAT lab Group, Badalona, Spain
| |
Collapse
|
27
|
Wang C, Wang S, Ma F, Zhang W. miRNA‑328 overexpression confers cisplatin resistance in non‑small cell lung cancer via targeting of PTEN. Mol Med Rep 2018; 18:4563-4570. [PMID: 30221716 DOI: 10.3892/mmr.2018.9478] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 04/12/2018] [Indexed: 11/06/2022] Open
Abstract
Chemotherapy resistance, the molecular mechanism of which is complex and has not been fully understood, poses a major challenge in the treatment of patients with non‑small cell lung cancer (NSCLC). The dysregulation of microRNAs (miRs) has been reported to serve a pivotal role in the development of cancer and drug resistance. In the present study, reverse transcription‑quantitative polymerase chain reaction analysis revealed a significant increase in miR‑328 and a significant decrease in phosphatase and tensin homolog (PTEN) mRNA expression levels within tumor tissues from patients with cisplatin‑resistant NSCLC compared with those of cisplatin‑sensitive NSCLC patients. In addition, there was a negative correlation between PTEN mRNA and the miR‑328 expression levels. In addition, higher miR‑328 expression levels, and lower PTEN mRNA and protein expression levels, were detected in cisplatin‑resistant A549 (A549rCDDP) cells when compared with in their parental cells. A549rCDDP cells demonstrated significantly higher cell viability compared with A549 cells following treatment with all concentrations of cisplatin tested (2, 4, 6 and 8 µM). Additionally, transfection of miR‑328 inhibitor significantly increased PTEN mRNA and protein expression levels. Furthermore, the present study predicted and confirmed PTEN, a well‑known tumor suppressor, as a direct target of miR‑328 in NSCLC cells via the online tool MiRanda and a dual luciferase assay, respectively. Cell viability assay and flow cytometry analysis demonstrated that inhibition of miR‑328 also induced cellular apoptosis and decreased cell proliferation in A549rCDDP cells treated with cisplatin. In conclusion, these results suggested that abnormal expression of miR‑328 may contribute to cisplatin resistance in NSCLC, and may be considered to be a novel therapeutic target and indicator for the treatment and prognosis of patients with NSCLC treated with cisplatin‑based chemotherapy.
Collapse
Affiliation(s)
- Chunmei Wang
- Department of Respiration, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| | - Shijun Wang
- Department of Emergency, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| | - Feng'e Ma
- Department of Outpatients, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| | - Weidan Zhang
- Department of Intensive Medicine, Dezhou People's Hospital, Dezhou, Shandong 253014, P.R. China
| |
Collapse
|
28
|
Chen FF, Lv X, Zhao QF, Xu YZ, Song SS, Yu W, Li XJ. Inhibitor of DNA binding 3 reverses cisplatin resistance in human lung adenocarcinoma cells by regulating the PI3K/Akt pathway. Oncol Lett 2018; 16:1634-1640. [PMID: 30008847 PMCID: PMC6036442 DOI: 10.3892/ol.2018.8849] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 01/26/2018] [Indexed: 11/06/2022] Open
Abstract
Inhibitor of DNA-binding 3 (ID3) is a helix-loop-helix transcription factor that is associated with cell proliferation, differentiation and drug resistance in human cancer, and with anticancer effects in certain types of cancer cells. The present study investigated whether and how ID3 was involved in multidrug resistance (MDR) in human cisplatin (DDP)-resistant A549/DDP lung adenocarcinoma cells. The underlying mechanism of action was investigated in vitro. Cell Counting Kit-8 (CCK-8) and flow cytometry assays demonstrated that overexpression of ID3 enhanced chemosensitivity and decreased drug efflux in A549/DDP cells. Reverse transcription-quantitative polymerase chain reaction revealed that the expression of anti-apoptotic gene B-cell lymphoma-2 was significantly downregulated in cells expressing exogenous ID3 (P<0.05). These results indicated that ID3 may synergize with DDP to increase apoptosis in A549/DDP cells. ID3 overexpression modulated the activity of phosphoinositide 3-kinase/RAC serine/threonine-protein kinase signaling and downregulated the expression of multi-drug resistance protein-1, indicating that ID3 expression can reverse multi-drug resistance in A549/DDP cells. Collectively, these results indicate that ID3 is a potential effective chemotherapeutic target for the treatment of human DDP-resistant A549 lung adenocarcinoma therapy.
Collapse
Affiliation(s)
- Fang-Fang Chen
- Center of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Xing Lv
- Center of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qin-Fei Zhao
- Center of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Yu-Zhong Xu
- Center of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Shu-Sheng Song
- Center of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Wei Yu
- Center of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Xiao-Jun Li
- Center of Clinical Laboratory Science, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China.,State Key Laboratory of Analytical Chemistry for Life Science, Department of Chemistry, Nanjing University, Nanjing, Jiangsu 210093, P.R. China
| |
Collapse
|
29
|
Lin C, Xie L, Lu Y, Hu Z, Chang J. miR-133b reverses cisplatin resistance by targeting GSTP1 in cisplatin-resistant lung cancer cells. Int J Mol Med 2018; 41:2050-2058. [PMID: 29328427 PMCID: PMC5810210 DOI: 10.3892/ijmm.2018.3382] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs play a critical role in chemoresistance and are implicated in various biological and pathological processes of cells. The objective of the present study was to explore the role of miR‑133b and its mechanism in the regulation of cisplatin resistance and tumor progression in cisplatin‑resistant non‑small cell lung cancer (NSCLC) cells. Reverse transcription‑quantitative polymerase chain reaction and western blot assays of the cisplatin‑resistant cell lines A549/DPP and H1299/DDP displayed the reduced expression of miR‑133b and increased expression of glutathione-S-transferase P1 (GSTP1) in the resistant cells compared with the respective parental cell lines A549 and H1299. Cell Counting kit‑8, flow cytometry, colony formation and Transwell migration assays indicated that the overexpression of miR‑133b increased the chemosensitivity to cisplatin and attenuated the proliferation and migration capacities of the cisplatin‑resistant NSCLC cell lines in vitro. A dual‑luciferase reporter assay demonstrated that miR‑133b negatively regulated the expression of GSTP1 by targeting its 3'‑untranslated region. In addition, the knockdown of GSTP1 by transfection with small interfering RNA exerted similar effects on cell chemosensitivity, proliferation and migration as did ectopic miR‑133b expression, in addition to the upregulation of Bax and downregulation of Bcl‑2, survivin and matrix metalloproteinase expression. In conclusion, the present study findings provide the insights that miR‑133b reduces cisplatin resistance and its overexpression contributes to the suppression of the malignant growth and aggressiveness of cisplatin‑resistant NSCLC cells by targeting GSTP1. This could potentially be exploited as a novel therapeutic strategy for the reversal of cisplatin resistance.
Collapse
Affiliation(s)
- Chen Lin
- Department of Oncology, Shanghai Medical College, Fudan University
- Department of Medical Oncology
| | - Liyi Xie
- Department of Oncology, Shanghai Medical College, Fudan University
- Department of Radiation Oncology
| | - Yan Lu
- Department of Oncology, Shanghai Medical College, Fudan University
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Zhihuang Hu
- Department of Oncology, Shanghai Medical College, Fudan University
- Department of Medical Oncology
| | - Jianhua Chang
- Department of Oncology, Shanghai Medical College, Fudan University
- Department of Medical Oncology
| |
Collapse
|
30
|
Galván-Femenía I, Guindo M, Duran X, Calabuig-Fariñas S, Mercader JM, Ramirez JL, Rosell R, Torrents D, Carreras A, Kohno T, Jantus-Lewintre E, Camps C, Perucho M, Sumoy L, Yokota J, de Cid R. Genomic profiling in advanced stage non-small-cell lung cancer patients with platinum-based chemotherapy identifies germline variants with prognostic value in SMYD2. Cancer Treat Res Commun 2018; 15:21-31. [PMID: 30207284 DOI: 10.1016/j.ctarc.2018.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 01/26/2018] [Accepted: 02/19/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE The aim of the study was to investigate the relationship between germline variations as a prognosis biomarker in patients with advanced Non-Small-Cell-Lung-Cancer (NSCLC) subjected to first-line platinum-based treatment. MATERIALS AND METHODS We carried out a two-stage genome-wide-association study in non-small-cell lung cancer patients with platinum-based chemotherapy in an exploratory sample of 181 NSCLC patients from Caucasian origin, followed by a validation on 356 NSCLC patients from the same ancestry (Valencia, Spain). RESULTS We identified germline variants in SMYD2 as a prognostic factor for survival in patients with advanced NSCLC receiving chemotherapy. SMYD2 alleles are associated to a decreased overall survival and with a reduced Time to Progression. In addition, enrichment pathway analysis identified 361 variants in 40 genes to be involved in poorer outcome in advanced-stage NSCLC patients. CONCLUSION Germline SMYD2 alleles are associated with bad clinical outcome of first-line platinum-based treatment in advanced NSCLC patients. This result supports the role of SMYD2 in the carcinogenic process, and might be used as prognostic signature directing patient stratification and the choice of therapy. MICROABSTRACT A two-Stage Genome wide association study in Caucasian population reveals germline genetic variation in SMYD2 associated to progression disease in first-line platinum-based treatment in advanced NSCLC patients. SMYD2 profiling might have prognostic / predictive value directing choice of therapy and enlighten current knowledge on pathways involved in human carcinogenesis as well in resistance to chemotherapy.
Collapse
Affiliation(s)
- Iván Galván-Femenía
- Genomes For life-GCAT Lab. Program of Predictive and Personalized Medicine of Cancer (PMPPC), Institute for Health Science Research Germans Trias i Pujol (IGTP), Can Ruti Biomedical Campus, Crta de Can Ruti, Camí de les Escoles S/N, 08916 Badalona, Barcelona, Spain.
| | - Marta Guindo
- Barcelona Supercomputing Center (BSC-CNS), Joint BSC-CRG-IRB Research Program in Computational Biology, Carrer de Jordi Girona, 29-31, 08034 Barcelona, Spain.
| | - Xavier Duran
- Genomes For life-GCAT Lab. Program of Predictive and Personalized Medicine of Cancer (PMPPC), Institute for Health Science Research Germans Trias i Pujol (IGTP), Can Ruti Biomedical Campus, Crta de Can Ruti, Camí de les Escoles S/N, 08916 Badalona, Barcelona, Spain.
| | - Sílvia Calabuig-Fariñas
- Department of Medical Oncology, Hospital General Universitario de Valencia, Avenida Tres Cruces, 2, 46014, València, Spain; Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Avda. Tres Cruces s/n 46014 València, Spain; Department of Pathology, Universitat de València, Av. de Blasco Ibáñez, 13, 46010 València, Spain.
| | - Josep Maria Mercader
- Barcelona Supercomputing Center (BSC-CNS), Joint BSC-CRG-IRB Research Program in Computational Biology, Carrer de Jordi Girona, 29-31, 08034 Barcelona, Spain.
| | - Jose Luis Ramirez
- Program of Predictive and Personalized Medicine of Cancer (PMPPC), Institute for Health Science Research Germans Trias i Pujol (IGTP), Can Ruti Biomedical Campus, Crta de Can Ruti, Camí de les Escoles S/N, 08916 Badalona, Barcelona, Spain.
| | - Rafael Rosell
- Program of Predictive and Personalized Medicine of Cancer (PMPPC), Institute for Health Science Research Germans Trias i Pujol (IGTP), Can Ruti Biomedical Campus, Crta de Can Ruti, Camí de les Escoles S/N, 08916 Badalona, Barcelona, Spain.
| | - David Torrents
- Barcelona Supercomputing Center (BSC-CNS), Joint BSC-CRG-IRB Research Program in Computational Biology, Carrer de Jordi Girona, 29-31, 08034 Barcelona, Spain; ICREA, Catalan Institution for Research and Advanced Studies, Spain.
| | - Anna Carreras
- Genomes For life-GCAT Lab. Program of Predictive and Personalized Medicine of Cancer (PMPPC), Institute for Health Science Research Germans Trias i Pujol (IGTP), Can Ruti Biomedical Campus, Crta de Can Ruti, Camí de les Escoles S/N, 08916 Badalona, Barcelona, Spain.
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Eloisa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Avda. Tres Cruces s/n 46014 València, Spain; Department of Pathology, Universitat de València, Av. de Blasco Ibáñez, 13, 46010 València, Spain; Molecular Oncology Laboratory, Fundación Hospital General Universitario de València, Avda. Tres Cruces s/n, 46014 València.
| | - Carlos Camps
- Department of Medical Oncology, Hospital General Universitario de Valencia, Avenida Tres Cruces, 2, 46014, València, Spain; Molecular Oncology Laboratory, Fundación Hospital General Universitario de Valencia, Avda. Tres Cruces s/n 46014 València, Spain; Department of Biotechnology, Universitat Politècnica de València, Camí de Vera, s/n, 46022 València, Spain; Department of Medicine, Universitat de València, Av. de Blasco Ibáñez, 13, 46010 València, Spain.
| | - Manuel Perucho
- Program of Predictive and Personalized Medicine of Cancer (PMPPC), Institute for Health Science Research Germans Trias i Pujol (IGTP), Can Ruti Biomedical Campus, Crta de Can Ruti, Camí de les Escoles S/N, 08916 Badalona, Barcelona, Spain.
| | - Lauro Sumoy
- Genomics and Bioinformatics. Program of Predictive and Personalized Medicine of Cancer (PMPPC), Institute for Health Science Research Germans Trias i Pujol (IGTP), Can Ruti Biomedical Campus, Crta de Can Ruti, Camí de les Escoles S/N, 08916 Badalona, Barcelona, Spain.
| | - Jun Yokota
- Program of Predictive and Personalized Medicine of Cancer (PMPPC), Institute for Health Science Research Germans Trias i Pujol (IGTP), Can Ruti Biomedical Campus, Crta de Can Ruti, Camí de les Escoles S/N, 08916 Badalona, Barcelona, Spain.
| | - Rafael de Cid
- Genomes For life-GCAT Lab. Program of Predictive and Personalized Medicine of Cancer (PMPPC), Institute for Health Science Research Germans Trias i Pujol (IGTP), Can Ruti Biomedical Campus, Crta de Can Ruti, Camí de les Escoles S/N, 08916 Badalona, Barcelona, Spain.
| |
Collapse
|
31
|
Chen QY, Jiao DM, Wang J, Hu H, Tang X, Chen J, Mou H, Lu W. miR-206 regulates cisplatin resistance and EMT in human lung adenocarcinoma cells partly by targeting MET. Oncotarget 2017; 7:24510-26. [PMID: 27014910 PMCID: PMC5029718 DOI: 10.18632/oncotarget.8229] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 03/04/2016] [Indexed: 01/15/2023] Open
Abstract
MicroRNAs (miRNAs) play a critical role in drug resistance and epithelial-mesenchymal transition (EMT). The aims of this study were to explore the potential role of miR-206 in governing cisplatin resistance and EMT in lung cancer cells. We found that both lung adenocarcinoma A549 cisplatin-resistant cells (A549/DDP) and H1299 cisplatin-resistant cells (H1299/DDP) acquired mesenchymal features and were along with low expression of miR-206 and high migration and invasion abilities. Ectopic expression of miR-206 mimics inhibited cisplatin resistance, reversed the EMT phenotype, decreased the migration and invasion in these DDP-resistant cells. In contrast, miR-206 inhibitors increased cisplatin resistance, EMT, cell migration and invasion in non-DDP-resistant cells. Furthermore, we found that MET is the direct target of miR-206 in lung cancer cells. Knockdown of MET exhibited an EMT and DDP resistant inhibitory effect on DDP-resistant cells. Conversely, overexpression of MET in non-DDP- resistant cells produced a promoting effect on cell EMT and DDP resistance. In lung adenocarcinoma tissues, we demonstrated that low expression of miR-206 were also correlated with increased cisplatin resistance and MET expression. In addition, we revealed that miR-206 overexpression reduced cisplatin resistance and EMT in DDP-resistant cells, partly due to inactivation of MET/PI3K/AKT/mTOR signaling pathway, and subsequent downregulation of MDR1, ZEB1 and Snail expression. Finally, we found that miR-206 could also sensitize A549/DDP cells to cisplatin in mice model. Taken together, our study implied that activation of miR-206 or inactivation of its target gene pathway could serve as a novel approach to reverse cisplatin resistance in lung adenocarcinomas cells.
Collapse
Affiliation(s)
- Qing-Yong Chen
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - De-Min Jiao
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Jian Wang
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Huizhen Hu
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Xiali Tang
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Jun Chen
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Hao Mou
- Department of Respiratory Disease, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| | - Wei Lu
- Department of Oncology, The 117th Hospital of PLA, Hangzhou, Zhejiang, 310013, P.R. China
| |
Collapse
|
32
|
Zhang T, Chen L, Zhang S, Xu Y, Fan Y, Zhang L. Effects of high-intensity focused ultrasound on cisplatin-resistant human lung adenocarcinoma in vitro and in vivo. Acta Biochim Biophys Sin (Shanghai) 2017; 49:1092-1098. [PMID: 29077784 DOI: 10.1093/abbs/gmx107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Indexed: 01/26/2023] Open
Abstract
It is widely accepted that high-intensity focused ultrasound (HIFU) is a minimally invasive treatment option for different tumors, but its roles and the corresponding mechanism in cisplatin (DDP) chemoresistance in lung adenocarcinoma (LA) remain unclear. In this study, we investigated the response of DDP-resistant LA cells to HIFU and its underlying molecular mechanisms using molecular biology techniques. It was found that HIFU exposure inhibited the proliferation of DDP-resistant A549 (A549/DDP) cells through arresting cell cycle at the G1/G0 phase via the Cyclin-dependent pathway and promoting apoptosis in a Bcl-2-dependent manner. Furthermore, the results also showed that HIFU exposure could down-regulate the expressions of MDR1, MRP1, and LRP mRNAs, as well as P-gp, MRP1, and LRP proteins related to drug resistance in A549/DDP cells. In vivo experiments also demonstrated that HIFU could reduce the size and mass of subcutaneously transplanted tumors produced by A549/DDP cells through mediating Cyclin-dependent and Bcl-2-dependent pathways. These results suggested that HIFU treatment could inhibit the proliferation of DDP-resistant lung cancer cells and might be a novel therapeutic method for patients with DDP resistance.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Ultrasound, Ningbo No. 9 Hospital, Ningbo, China
| | - Libin Chen
- Department of Ultrasound, Ningbo First Hospital, Ningbo, China
| | - Shengmin Zhang
- Department of Ultrasound, Ningbo First Hospital, Ningbo, China
| | - Youfeng Xu
- Department of Ultrasound, Ningbo First Hospital, Ningbo, China
| | - Yabo Fan
- Department of Ultrasound, Ningbo No. 9 Hospital, Ningbo, China
| | - Lizhong Zhang
- Department of Ultrasound, Ningbo No. 9 Hospital, Ningbo, China
| |
Collapse
|
33
|
Pinto R, Assis J, Nogueira A, Pereira C, Pereira D, Medeiros R. Rethinking ovarian cancer genomics: where genome-wide association studies stand? Pharmacogenomics 2017; 18:1611-1625. [DOI: 10.2217/pgs-2017-0108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Genome-wide association studies (GWAS) allow the finding of genetic variants associated with several traits. Regarding ovarian cancer (OC), 15 GWAS have been conducted since 2009, with the discovery of 49 SNPs associated with disease susceptibility and 46 with impact in the clinical outcome of patients (p < 5.00 × 10-2). Among them, 14 variants reached the genome-wide significance (p < 5.00 × 10−8). Despite the results obtained, they should be validated in independent sets. So far, five validation studies have been conducted which could confirm the association of 12 OC susceptibility SNPs. Consequently, post-GWAS studies are crucial unravel the biological plausibility of GWAS’ findings and the allelic spectrum of OC.
Collapse
Affiliation(s)
- Ricardo Pinto
- Molecular Oncology & Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr. António Bernardino de Almeida, 4200–4072, Porto, Portugal
- ICBAS, Abel Salazar Institute for the Biomedical Sciences, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Joana Assis
- Molecular Oncology & Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr. António Bernardino de Almeida, 4200–4072, Porto, Portugal
- FMUP, Faculty of Medicine, Porto University, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Augusto Nogueira
- Molecular Oncology & Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr. António Bernardino de Almeida, 4200–4072, Porto, Portugal
- FMUP, Faculty of Medicine, Porto University, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Carina Pereira
- Molecular Oncology & Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr. António Bernardino de Almeida, 4200–4072, Porto, Portugal
- CINTESIS, Center for Health technology and Services Research, Faculty of Medicine, Porto University, Rua Dr. Plácido da Costa, 4200-450, Porto, Portugal
| | - Deolinda Pereira
- Oncology Department, Portuguese Institute of Oncology, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology & Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr. António Bernardino de Almeida, 4200–4072, Porto, Portugal
- Research Department, Portuguese League AgainstCancer (NRNorte), Estrada Interior da Circunvalação, 6657, 4200-172, Porto, Portugal
- CEBIMED, Faculty of Health Sciences, FernandoPessoa University, Praça 9 de Abril, 349, 4249-004, Porto, Portugal
| |
Collapse
|
34
|
Pan JY, Sun CC, Bi ZY, Chen ZL, Li SJ, Li QQ, Wang YX, Bi YY, Li DJ. miR-206/133b Cluster: A Weapon against Lung Cancer? MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:442-449. [PMID: 28918043 PMCID: PMC5542379 DOI: 10.1016/j.omtn.2017.06.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/25/2017] [Accepted: 06/02/2017] [Indexed: 12/29/2022]
Abstract
Lung cancer is a deadly disease that ends numerous lives around the world. MicroRNAs (miRNAs) are a group of non-coding RNAs involved in a variety of biological processes, such as cell growth, organ development, and tumorigenesis. The miR-206/133b cluster is located on the human chromosome 6p12.2, which is essential for growth and rebuilding of skeletal muscle. The miR-206/133b cluster has been verified to be dysregulated and plays a crucial role in lung cancer. miR-206 and miR-133b participate in lung tumor cell apoptosis, proliferation, migration, invasion, angiogenesis, drug resistance, and cancer treatment. The mechanisms are sophisticated, involving various target genes and molecular pathways, such as MET, EGFR, and the STAT3/HIF-1α/VEGF signal pathway. Hence, in this review, we summarize the role and potential mechanisms of the miR-206/133b cluster in lung cancer.
Collapse
Affiliation(s)
- Jing-Yu Pan
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071 Hubei, P.R. China
| | - Cheng-Cao Sun
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071 Hubei, P.R. China.
| | - Zhuo-Yue Bi
- Hubei Provincial Key Laboratory for Applied Toxicology (Hubei Provincial Academy for Preventive Medicine), Wuhan 430079 Hubei, P.R. China
| | - Zhen-Long Chen
- Wuhan Hospital for the Prevention and Treatment of Occupational Diseases, Wuhan 430022 Hubei, P.R. China
| | - Shu-Jun Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071 Hubei, P.R. China; Wuhan Hospital for the Prevention and Treatment of Occupational Diseases, Wuhan 430022 Hubei, P.R. China
| | - Qing-Qun Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071 Hubei, P.R. China
| | - Yu-Xuan Wang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071 Hubei, P.R. China
| | - Yong-Yi Bi
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071 Hubei, P.R. China
| | - De-Jia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071 Hubei, P.R. China.
| |
Collapse
|
35
|
Lopes-Ramos CM, Paulson JN, Chen CY, Kuijjer ML, Fagny M, Platig J, Sonawane AR, DeMeo DL, Quackenbush J, Glass K. Regulatory network changes between cell lines and their tissues of origin. BMC Genomics 2017; 18:723. [PMID: 28899340 PMCID: PMC5596945 DOI: 10.1186/s12864-017-4111-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 09/01/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell lines are an indispensable tool in biomedical research and often used as surrogates for tissues. Although there are recognized important cellular and transcriptomic differences between cell lines and tissues, a systematic overview of the differences between the regulatory processes of a cell line and those of its tissue of origin has not been conducted. The RNA-Seq data generated by the GTEx project is the first available data resource in which it is possible to perform a large-scale transcriptional and regulatory network analysis comparing cell lines with their tissues of origin. RESULTS We compared 127 paired Epstein-Barr virus transformed lymphoblastoid cell lines (LCLs) and whole blood samples, and 244 paired primary fibroblast cell lines and skin samples. While gene expression analysis confirms that these cell lines carry the expression signatures of their primary tissues, albeit at reduced levels, network analysis indicates that expression changes are the cumulative result of many previously unreported alterations in transcription factor (TF) regulation. More specifically, cell cycle genes are over-expressed in cell lines compared to primary tissues, and this alteration in expression is a result of less repressive TF targeting. We confirmed these regulatory changes for four TFs, including SMAD5, using independent ChIP-seq data from ENCODE. CONCLUSIONS Our results provide novel insights into the regulatory mechanisms controlling the expression differences between cell lines and tissues. The strong changes in TF regulation that we observe suggest that network changes, in addition to transcriptional levels, should be considered when using cell lines as models for tissues.
Collapse
Affiliation(s)
- Camila M. Lopes-Ramos
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Joseph N. Paulson
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Cho-Yi Chen
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Marieke L. Kuijjer
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Maud Fagny
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - John Platig
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Abhijeet R. Sonawane
- Channing Division of Network Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA USA
| | - Dawn L. DeMeo
- Channing Division of Network Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women’s Hospital, Boston, MA USA
| | - John Quackenbush
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215 USA
| | - Kimberly Glass
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA USA
| |
Collapse
|
36
|
Gong X, Liu J, Zhang D, Yang D, Min Z, Wen X, Wang G, Li H, Song Y, Bai C, Li J, Zhou J. GLIPR1 modulates the response of cisplatin-resistant human lung cancer cells to cisplatin. PLoS One 2017; 12:e0182410. [PMID: 28771580 PMCID: PMC5542429 DOI: 10.1371/journal.pone.0182410] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 07/18/2017] [Indexed: 01/01/2023] Open
Abstract
Background and objective Chemotherapy drugs, such as cisplatin (DDP), improve the survival of patients with lung cancer by inducing apoptosis in cancer cells, which quickly develop resistance to DDP through uncharacterized mechanisms. Glioma Pathogenesis-Related Protein 1 (GLIPR1) plays an important role in cell proliferation, migration and apoptosis. However, the expression and function of GLIPR1 in mediating DDP resistance in human lung adenocarcinoma A549/DDP and human large cell lung cancer H460/DDP cells has not yet been reported. Methods In this study, real-time PCR (RT-PCR) and western blot were used to examine the mRNA and protein expression of GLIPR1, respectively. Bright-field microscopy, the cell counting kit-8 (CCK-8) assay, flow cytometry analysis and JC-1 dye were used to measure the cellular morphology, proliferation, apoptosis and mitochondrial membrane potential, respectively. Results Compared to human lung adenocarcinoma A549 cells, the mRNA and protein expression of GLIPR1 were significantly increased in DDP-resistant A549/DDP cells (p < 0.05). Similarly, the mRNA level of GLIPR1 in DDP-resistant H460/DDP cells was also significantly higher than that in DDP-sensitive H460 cells (p < 0.05). Silencing of GLIPR1 in A549/DDP and H460/DDP cells led to increased apoptosis via a mitochondrial signaling pathway following incubation with various concentrations of DDP. Furthermore, GLIPR1 downregulation markedly reduced the protein expression of Bcl-2, and increased the cleaved Poly (ADP-Ribose) Polymerase (PARP) and cleaved caspase-3 in DDP-resistant A549/DDP cells. Conclusion In this study, we demonstrated for the first time that GLIPR1 could modulate the response of DDP-resistant A549/DDP and H460/DDP cells to cisplatin. Therefore, GLIPR1 deserves further investigation in the context of none-small lung cancer (NSCLC).
Collapse
Affiliation(s)
- Xin Gong
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Liu
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Dan Zhang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dawei Yang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhihui Min
- Biomedical Research Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoxing Wen
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guifang Wang
- Department of Pulmonary Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Huayin Li
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuanlin Song
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunxue Bai
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Jing Li
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- * E-mail: (JZ); (JL)
| | - Jian Zhou
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- * E-mail: (JZ); (JL)
| |
Collapse
|
37
|
Jones CC, Bush WS, Crawford DC, Wenzlaff AS, Schwartz AG, Wiencke JK, Wrensch MR, Blot WJ, Chanock SJ, Grogan EL, Aldrich MC. Germline Genetic Variants and Lung Cancer Survival in African Americans. Cancer Epidemiol Biomarkers Prev 2017; 26:1288-1295. [PMID: 28619829 DOI: 10.1158/1055-9965.epi-16-0998] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/14/2017] [Accepted: 05/17/2017] [Indexed: 11/16/2022] Open
Abstract
Background: African Americans have the highest lung cancer mortality in the United States. Genome-wide association studies (GWASs) of germline variants influencing lung cancer survival have not yet been conducted with African Americans. We examined five previously reported GWAS catalog variants and explored additional genome-wide associations among African American lung cancer cases.Methods: Incident non-small cell lung cancer cases (N = 286) in the Southern Community Cohort Study were genotyped on the Illumina HumanExome BeadChip. We used Cox proportional hazards models to estimate HRs and 95% confidence intervals (CIs) for overall mortality. Two independent African American studies (N = 316 and 298) were used for replication.Results: One previously reported variant, rs1878022 on 12q23.3, was significantly associated with mortality (HR = 0.70; 95% CI: 0.54-0.92). Replication findings were in the same direction, although attenuated (HR = 0.87 and 0.94). Meta-analysis had a HR of 0.83 (95% CI, 0.71-0.97). Analysis of common variants identified an association between chromosome 6q21.33 and mortality (HR = 0.46; 95% CI, 0.33-0.66).Conclusions: We identified an association between rs1878022 in CMKLR1 and lung cancer survival. However, our results in African Americans have a different direction of effect compared with a prior study in European Americans, suggesting a different genetic architecture or presence of gene-environment interactions. We also identified variants on chromosome 6 within the gene-rich HLA region, which has been previously implicated in lung cancer risk and survival.Impact: We found evidence that inherited genetic risk factors influence lung cancer survival in African Americans. Replication in additional populations is necessary to confirm potential genetic differences in lung cancer survival across populations. Cancer Epidemiol Biomarkers Prev; 26(8); 1288-95. ©2017 AACR.
Collapse
Affiliation(s)
- Carissa C Jones
- Department of Thoracic Surgery, Vanderbilt University Medical School, Nashville, Tennessee.,Vanderbilt Genetics Institute, Vanderbilt University Medical School, Nashville, Tennessee
| | - William S Bush
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dana C Crawford
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Angela S Wenzlaff
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - Ann G Schwartz
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan
| | - John K Wiencke
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Margaret R Wrensch
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California.,Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California.,Institute of Human Genetics, University of California San Francisco, San Francisco, California
| | - William J Blot
- Division of Epidemiology, Vanderbilt University Medical School, Nashville, Tennessee.,International Epidemiology Institute, Rockville, Maryland
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| | - Eric L Grogan
- Department of Thoracic Surgery, Vanderbilt University Medical School, Nashville, Tennessee.,Tennessee Valley Health System Veterans Affairs, Nashville, Tennessee
| | - Melinda C Aldrich
- Department of Thoracic Surgery, Vanderbilt University Medical School, Nashville, Tennessee. .,Vanderbilt Genetics Institute, Vanderbilt University Medical School, Nashville, Tennessee.,Division of Epidemiology, Vanderbilt University Medical School, Nashville, Tennessee
| |
Collapse
|
38
|
Genetic Variants Contributing to Colistin Cytotoxicity: Identification of TGIF1 and HOXD10 Using a Population Genomics Approach. Int J Mol Sci 2017; 18:ijms18030661. [PMID: 28335481 PMCID: PMC5372673 DOI: 10.3390/ijms18030661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 03/15/2017] [Accepted: 03/16/2017] [Indexed: 12/27/2022] Open
Abstract
Colistin sulfate (polymixin E) is an antibiotic prescribed with increasing frequency for severe Gram-negative bacterial infections. As nephrotoxicity is a common side effect, the discovery of pharmacogenomic markers associated with toxicity would benefit the utility of this drug. Our objective was to identify genetic markers of colistin cytotoxicity that were also associated with expression of key proteins using an unbiased, whole genome approach and further evaluate the functional significance in renal cell lines. To this end, we employed International HapMap lymphoblastoid cell lines (LCLs) of Yoruban ancestry with known genetic information to perform a genome-wide association study (GWAS) with cellular sensitivity to colistin. Further association studies revealed that single nucleotide polymorphisms (SNPs) associated with gene expression and protein expression were significantly enriched in SNPs associated with cytotoxicity (p ≤ 0.001 for gene and p = 0.015 for protein expression). The most highly associated SNP, chr18:3417240 (p = 6.49 × 10−8), was nominally a cis-expression quantitative trait locus (eQTL) of the gene TGIF1 (transforming growth factor β (TGFβ)-induced factor-1; p = 0.021) and was associated with expression of the protein HOXD10 (homeobox protein D10; p = 7.17 × 10−5). To demonstrate functional relevance in a murine colistin nephrotoxicity model, HOXD10 immunohistochemistry revealed upregulated protein expression independent of mRNA expression in response to colistin administration. Knockdown of TGIF1 resulted in decreased protein expression of HOXD10 and increased resistance to colistin cytotoxicity. Furthermore, knockdown of HOXD10 in renal cells also resulted in increased resistance to colistin cytotoxicity, supporting the physiological relevance of the initial genomic associations.
Collapse
|
39
|
Petros Z, Makonnen E, Aklillu E. Genome-Wide Association Studies for Idiosyncratic Drug-Induced Hepatotoxicity: Looking Back-Looking Forward to Next-Generation Innovation. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2017; 21:123-131. [PMID: 28253087 DOI: 10.1089/omi.2017.0006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Idiosyncratic drug-induced hepatotoxicity is a formidable challenge for rational drug discovery and development, as well as the science of personalized medicine. There is evidence that hereditary factors, in part, contribute to drug toxicity. This expert analysis and review offer the insights gained, and the challenges ahead, for genome-wide association studies (GWASs) of idiosyncratic drug-induced hepatotoxicity. Published articles on genome-wide and subsequent replication studies were systematically searched in the PubMed electronic database. We found that the genetic risk variants that were identified genome-wide, and replication confirmed, are mainly related to polymorphisms in the human leukocyte antigen (HLA) region that include HLA-DQB1*06:02 for amoxicillin-clavulanate, HLA-B*57:01 for flucloxacillin, HLA-DRB1*15:01 for lumiracoxib, and HLA-DRB1*07:01 for lapatinib and ximelagatran-induced hepatotoxicity. Additionally, polymorphisms in ST6 β-galactosamide α-2, 6-sialyltranferase-1 (ST6GAL1), which plays a role in systemic inflammatory response, and variants in intron of family with sequence similarity-65 member-B (FAM65B) that play roles in liver inflammation displayed association with flucloxacillin and antituberculosis drug-induced hepatotoxicity, respectively. Taken together, these GWAS findings offer molecular leads on the central role that the immune system plays in idiosyncratic drug-induced hepatotoxicity. We conclude the expert review with a brief discussion of the salient challenges ahead. These include, for example, the need for discursive discovery paradigms that incorporate alternating GWASs and candidate gene studies, as well as the study of the environtome, the entire complement of environmental factors, including science and innovation policies that enact on global society and the human host, and by extension, on susceptibility for idiosyncratic drug-induced hepatotoxicity.
Collapse
Affiliation(s)
- Zelalem Petros
- 1 Department of Pharmacology, School of Medicine, College of Health Sciences, Addis Ababa University , Addis Ababa, Ethiopia
| | - Eyasu Makonnen
- 1 Department of Pharmacology, School of Medicine, College of Health Sciences, Addis Ababa University , Addis Ababa, Ethiopia
| | - Eleni Aklillu
- 2 Division of Clinical Pharmacology, Department of Laboratory Medicine, Karolinska University Hospital , Huddinge C1:68, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Gallo A, Vella S, Miele M, Timoneri F, Di Bella M, Bosi S, Sciveres M, Conaldi PG. Global profiling of viral and cellular non-coding RNAs in Epstein-Barr virus-induced lymphoblastoid cell lines and released exosome cargos. Cancer Lett 2016; 388:334-343. [PMID: 27956246 DOI: 10.1016/j.canlet.2016.12.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 12/01/2016] [Accepted: 12/02/2016] [Indexed: 01/01/2023]
Abstract
The human EBV-transformed lymphoblastoid cell line (LCL), obtained by infecting peripheral blood monocular cells with Epstein-Barr Virus, has been extensively used for human genetic, pharmacogenomic, and immunologic studies. Recently, the role of exosomes has also been indicated as crucial in the crosstalk between EBV and the host microenvironment. Because the role that the LCL and LCL exosomal cargo might play in maintaining persistent infection, and since little is known regarding the non-coding RNAs of LCL, the aim of our work was the comprehensive characterization of this class of RNA, cellular and viral miRNAs, and cellular lncRNAs, in LCL compared with PBMC derived from the same donors. In this study, we have demonstrated, for the first time, that all the viral miRNAs expressed by LCL are also packaged in the exosomes, and we found that two miRNAs, ebv-miR-BART3 and ebv-miR-BHRF1-1, are more abundant in the exosomes, suggesting a microvescicular viral microRNA transfer. In addition, lncRNA profiling revealed that LCLs were enriched in lncRNA H19 and H19 antisense, and released these through exosomes, suggesting a leading role in the regulation of the tumor microenvironment.
Collapse
Affiliation(s)
- Alessia Gallo
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Italy.
| | - Serena Vella
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Italy
| | | | | | | | | | - Marco Sciveres
- Pediatric Hepatology and Liver Transplantation, IRCCS ISMETT, University of Pittsburgh Medical Center, Palermo, Italy
| | - Pier Giulio Conaldi
- Department of Laboratory Medicine and Advanced Biotechnologies, IRCCS-ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta specializzazione), Italy; Fondazione Ri.MED, Italy
| |
Collapse
|
41
|
Jia M, Xu Y, Zhu M, Wang M, Sun M, Qian J, Chang J, Wei Q. The P38α rs3804451 Variant Predicts Chemotherapy Response and Survival of Patients with Non-Small Cell Lung Cancer Treated with Platinum-Based Chemotherapy. Transl Oncol 2016; 9:531-539. [PMID: 27835790 PMCID: PMC5109261 DOI: 10.1016/j.tranon.2016.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 09/16/2016] [Accepted: 09/19/2016] [Indexed: 01/01/2023] Open
Abstract
The JNK and P38α pathways play an important role in the sensitivity and outcomes of chemotherapy. We hypothesize that functional single nucleotide polymorphisms (SNPs) of genes of these pathways modulate outcomes of patients with advanced non-small cell lung cancer (NSCLC) treated with first-line platinum-based chemotherapy (PBC). We selectively genotyped 11 independent, potentially functional SNPs of 9 genes in the JNK and P38α pathways first in a discovery group of 355 patients with advanced NSCLC treated with PBC, and we evaluated their associations with progression-free survival (PFS) and overall survival (OS) by Cox proportional hazards regression analysis. Then, resultant significant SNPs were further validated in a replication group of 355 patients. In both discovery and validation groups as well as their combined analysis, the MAPK14 rs3804451GA/AA genotypes showed a strong association with a reduced PFS (adjusted hazards ratio [HR]=1.39; 95% confidence interval [CI]=1.16-1.66; P=.0003) and OS (adjusted HR=1.41; 95% CI=1.11-1.80; P=.005) compared with the wild-type GG genotype. In contrast, patients with or without the MAPK14 rs3804451A allele had no significant difference in OS in response to tyrosine-kinase inhibitor treatment (adjusted HR=0.86; 95% CI=0.56-1.33; P=.505). The present study provides evidence that the MAPK14 rs3804451 G>A variant may modulate survival outcomes in patients with advanced NSCLC treated with PBC. Larger studies of additional patient populations are needed to validate our findings.
Collapse
Affiliation(s)
- Ming Jia
- Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Fudan University Shanghai Cancer Center, 270 Dongan Road, Xuhui, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Xuhui, Shanghai, 200032, China.
| | - Yuan Xu
- Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Fudan University Shanghai Cancer Center, 270 Dongan Road, Xuhui, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Xuhui, Shanghai, 200032, China.
| | - Meiling Zhu
- Department of Oncology, Xinhua Hospital affiliated to Shanghai Jiaotong University, School of Medicine, No. 1665, Kong Jiang Road, Shanghai, 200092, China.
| | - Mengyun Wang
- Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Fudan University Shanghai Cancer Center, 270 Dongan Road, Xuhui, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Xuhui, Shanghai, 200032, China.
| | - Menghong Sun
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Xuhui, Shanghai, 200032, China; Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Xuhui, Shanghai, 200032, China.
| | - Ji Qian
- State Key Laboratory of Genetic Engineering and MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences and Fudan Taizhou Institute of Health Sciences, Fudan University, Shanghai, 200032, China.
| | - Jianhua Chang
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Xuhui, Shanghai, 200032, China; Department of Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Xuhui, Shanghai, 200032, China.
| | - Qingyi Wei
- Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Fudan University Shanghai Cancer Center, 270 Dongan Road, Xuhui, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 270 Dongan Road, Xuhui, Shanghai, 200032, China; Duke Cancer Institute, Duke University Medical Center, and Department of Medicine, Duke University School of Medicine, Durham, NC, 27710, USA.
| |
Collapse
|
42
|
Hanson C, Cairns J, Wang L, Sinha S. Computational discovery of transcription factors associated with drug response. THE PHARMACOGENOMICS JOURNAL 2016; 16:573-582. [PMID: 26503816 PMCID: PMC4848185 DOI: 10.1038/tpj.2015.74] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 08/04/2015] [Accepted: 08/07/2015] [Indexed: 02/01/2023]
Abstract
This study integrates gene expression, genotype and drug response data in lymphoblastoid cell lines with transcription factor (TF)-binding sites from ENCODE (Encyclopedia of Genomic Elements) in a novel methodology that elucidates regulatory contexts associated with cytotoxicity. The method, GENMi (Gene Expression iN the Middle), postulates that single-nucleotide polymorphisms within TF-binding sites putatively modulate its regulatory activity, and the resulting variation in gene expression leads to variation in drug response. Analysis of 161 TFs and 24 treatments revealed 334 significantly associated TF-treatment pairs. Investigation of 20 selected pairs yielded literature support for 13 of these associations, often from studies where perturbation of the TF expression changes drug response. Experimental validation of significant GENMi associations in taxanes and anthracyclines across two triple-negative breast cancer cell lines corroborates our findings. The method is shown to be more sensitive than an alternative, genome-wide association study-based approach that does not use gene expression. These results demonstrate the utility of GENMi in identifying TFs that influence drug response and provide a number of candidates for further testing.
Collapse
Affiliation(s)
- C Hanson
- Department of Computer Science, University of Illinois at Urbana–Champaign, Urbana, IL, USA
| | - J Cairns
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - L Wang
- Division of Clinical Pharmacology, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - S Sinha
- Department of Computer Science and Institute of Genomic Biology, University of Illinois at Urbana–Champaign, Urbana, IL, USA
| |
Collapse
|
43
|
Duan S, Tsai Y, Keng P, Chen Y, Lee SO, Chen Y. IL-6 signaling contributes to cisplatin resistance in non-small cell lung cancer via the up-regulation of anti-apoptotic and DNA repair associated molecules. Oncotarget 2016; 6:27651-60. [PMID: 26313152 PMCID: PMC4695015 DOI: 10.18632/oncotarget.4753] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/17/2015] [Indexed: 12/27/2022] Open
Abstract
Cisplatin-based chemotherapy is currently the most effective treatment regimen for non-small cell lung cancer (NSCLC), but eventually tumor resistance develops which limits its success. The potential implication of IL-6 signaling in the cisplatin resistance of NSCLC was explored by testing whether NSCLC cells with different levels of intracellular IL-6 show different responses to the cytotoxic treatment of cisplatin. When the cisplatin cytotoxicity of the IL-6 knocked down human NSCLC cells (A549IL-6si and H157IL-6si) were compared with their corresponding scramble control cells (A549sc and H157sc), higher cisplatin cytotoxicity was found in IL-6 si cells than sc cells. Subcutaneous xenograft mouse models were developed using a pair of A549sc and A549IL-6si cells. When the tumor grew to about 400 mm2, mice were treated with cisplatin and tumor regression was monitored. Higher tumor regression was detected in the A549IL-6si xenografts compared to A549sc xenografts following cisplatin treatment. Immunostaining study results from tumor tissues also supported this finding. Expression of anti-apoptotic proteins Bcl-2 and Mcl-1 and DNA repair associated molecules ATM, CHK1, TP73, p53, and ERCC1 were significantly up regulated in cisplatin-treated A549sc and H157sc cells, but no increase was detected in A549IL-6si and H157IL-6si cells. Further inhibitor studies revealed that up regulation of these molecules by IL-6 may be through activation of IL-6 downstream signaling pathways like Akt, MAPK, Stat3, and Erk. These results provide potential for combining cisplatin and inhibitors of IL-6 signaling or its downstream signaling pathway as a future therapeutic approach in preventing development of cisplatin resistant NSCLC tumors.
Collapse
Affiliation(s)
- Shanzhou Duan
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA.,Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Ying Tsai
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Peter Keng
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yongbing Chen
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| | - Soo Ok Lee
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| |
Collapse
|
44
|
HU CHUNFANG, HUANG YIYING, WANG YONGJIE, GAO FENGGUANG. Upregulation of ABCG2 via the PI3K-Akt pathway contributes to acidic microenvironment-induced cisplatin resistance in A549 and LTEP-a-2 lung cancer cells. Oncol Rep 2016; 36:455-61. [DOI: 10.3892/or.2016.4827] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/17/2016] [Indexed: 11/06/2022] Open
|
45
|
French JD, Johnatty SE, Lu Y, Beesley J, Gao B, Kalimutho M, Henderson MJ, Russell AJ, Kar S, Chen X, Hillman KM, Kaufmann S, Sivakumaran H, O'Reilly M, Wang C, Korbie DJ, Lambrechts D, Despierre E, Van Nieuwenhuysen E, Lambrechts S, Vergote I, Karlan B, Lester J, Orsulic S, Walsh C, Fasching PA, Beckmann MW, Ekici AB, Hein A, Matsuo K, Hosono S, Pisterer J, Hillemanns P, Nakanishi T, Yatabe Y, Goodman MT, Lurie G, Matsuno RK, Thompson PJ, Pejovic T, Bean Y, Heitz F, Harter P, du Bois A, Schwaab I, Hogdall E, Kjaer SK, Jensen A, Hogdall C, Lundvall L, Engelholm SA, Brown B, Flanagan JM, Metcalf MD, Siddiqui N, Sellers T, Fridley B, Cunningham J, Schildkraut JM, Iversen E, Weber RP, Brennan D, Berchuck A, Pharoah P, Harnett P, Norris MD, Haber M, Goode EL, Lee JS, Khanna KK, Meyer KB, Chenevix-Trench G, deFazio A, Edwards SL, MacGregor S. Germline polymorphisms in an enhancer of PSIP1 are associated with progression-free survival in epithelial ovarian cancer. Oncotarget 2016; 7:6353-68. [PMID: 26840454 PMCID: PMC4872719 DOI: 10.18632/oncotarget.7047] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 01/21/2016] [Indexed: 11/25/2022] Open
Abstract
Women with epithelial ovarian cancer (EOC) are usually treated with platinum/taxane therapy after cytoreductive surgery but there is considerable inter-individual variation in response. To identify germline single-nucleotide polymorphisms (SNPs) that contribute to variations in individual responses to chemotherapy, we carried out a multi-phase genome-wide association study (GWAS) in 1,244 women diagnosed with serous EOC who were treated with the same first-line chemotherapy, carboplatin and paclitaxel. We identified two SNPs (rs7874043 and rs72700653) in TTC39B (best P=7x10-5, HR=1.90, for rs7874043) associated with progression-free survival (PFS). Functional analyses show that both SNPs lie in a putative regulatory element (PRE) that physically interacts with the promoters of PSIP1, CCDC171 and an alternative promoter of TTC39B. The C allele of rs7874043 is associated with poor PFS and showed increased binding of the Sp1 transcription factor, which is critical for chromatin interactions with PSIP1. Silencing of PSIP1 significantly impaired DNA damage-induced Rad51 nuclear foci and reduced cell viability in ovarian cancer lines. PSIP1 (PC4 and SFRS1 Interacting Protein 1) is known to protect cells from stress-induced apoptosis, and high expression is associated with poor PFS in EOC patients. We therefore suggest that the minor allele of rs7874043 confers poor PFS by increasing PSIP1 expression.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/genetics
- Apoptosis
- Biomarkers, Tumor/genetics
- Cell Proliferation
- Chromatin Immunoprecipitation
- Cohort Studies
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/mortality
- Cystadenocarcinoma, Serous/pathology
- Electrophoretic Mobility Shift Assay
- Enhancer Elements, Genetic/genetics
- Fallopian Tube Neoplasms/drug therapy
- Fallopian Tube Neoplasms/genetics
- Fallopian Tube Neoplasms/mortality
- Fallopian Tube Neoplasms/pathology
- Female
- Follow-Up Studies
- Germ-Line Mutation/genetics
- Humans
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/pathology
- Peritoneal Neoplasms/drug therapy
- Peritoneal Neoplasms/genetics
- Peritoneal Neoplasms/mortality
- Peritoneal Neoplasms/pathology
- Polymorphism, Single Nucleotide/genetics
- Prognosis
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Survival Rate
- Transcription Factors/genetics
- Tumor Cells, Cultured
Collapse
Affiliation(s)
| | | | - Yi Lu
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Bo Gao
- Department of Gynaecological Oncology and Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead Hospital, Sydney, Australia
| | | | | | | | - Siddhartha Kar
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| | - Xiaoqing Chen
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | | | | | - Martin O'Reilly
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
| | - Chen Wang
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Darren J. Korbie
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Australia
| | - Australian Ovarian Cancer Study Group
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Department of Gynaecological Oncology and Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead Hospital, Sydney, Australia
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | - Diether Lambrechts
- Vesalius Research Center, VIB, Leuven, Belgium and Laboratory for Translational Genetics, Department of Oncology, University of Leuven, Leuven, Belgium
- Gynecologic Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Evelyn Despierre
- Gynecologic Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Els Van Nieuwenhuysen
- Gynecologic Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Sandrina Lambrechts
- Gynecologic Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Ignace Vergote
- Gynecologic Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Beth Karlan
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jenny Lester
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sandra Orsulic
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Christine Walsh
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Peter A. Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen- Nuremberg, Comprehensive Cancer Center Erlangen-Nuremberg, Erlangen, Germany
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Matthias W. Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen- Nuremberg, Comprehensive Cancer Center Erlangen-Nuremberg, Erlangen, Germany
| | - Arif B. Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Alexander Hein
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen- Nuremberg, Comprehensive Cancer Center Erlangen-Nuremberg, Erlangen, Germany
| | - Keitaro Matsuo
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan
| | - Satoyo Hosono
- Division of Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Aichi, Japan
| | | | - Peter Hillemanns
- Departments of Obstetrics and Gynaecology, Hannover Medical School, Hannover, Germany
| | - Toru Nakanishi
- Department of Gynecology, Aichi Cancer Center Central Hospital, Nagoya, Aichi, Japan
| | - Yasushi Yatabe
- Department of Pathology and Molecular Diagnostics, Aichi Cancer Center Central Hospital, Nagoya, Aichi, Japan
| | - Marc T. Goodman
- Cancer Prevention and Control Program, Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Galina Lurie
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Hawaii, USA
| | - Rayna K. Matsuno
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Hawaii, USA
| | - Pamela J. Thompson
- Cancer Prevention and Control Program, Samuel Oschin Comprehensive Cancer Institute, Cedars Sinai Medical Center, Los Angeles, CA, USA
| | - Tanja Pejovic
- Department of Obstetrics and Gynecology, Oregon Health and Science University and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Yukie Bean
- Department of Obstetrics and Gynecology, Oregon Health and Science University and Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Florian Heitz
- Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany
| | - Philipp Harter
- Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany
| | - Andreas du Bois
- Department of Gynecology and Gynecologic Oncology, Dr. Horst Schmidt Kliniken Wiesbaden, Wiesbaden, Germany
- Department of Gynecology and Gynecologic Oncology, Kliniken Essen-Mitte, Essen, Germany
| | - Ira Schwaab
- Institut für Humangenetik Wiesbaden, Germany
| | - Estrid Hogdall
- Danish Cancer Society Research Center, Unit of Virus, Lifestyle and Genes, Copenhagen, Denmark
- Molecular Unit, Department of Pathology, Herlev Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Susanne K. Kjaer
- Danish Cancer Society Research Center, Unit of Virus, Lifestyle and Genes, Copenhagen, Denmark
- Department of Gynecology, Rigshospitalet, University of Copenhagen, Denmark
| | - Allan Jensen
- Danish Cancer Society Research Center, Unit of Virus, Lifestyle and Genes, Copenhagen, Denmark
| | - Claus Hogdall
- Department of Gynecology, Rigshospitalet, University of Copenhagen, Denmark
| | - Lene Lundvall
- Department of Gynecology, Rigshospitalet, University of Copenhagen, Denmark
| | | | - Bob Brown
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - James M. Flanagan
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Nadeem Siddiqui
- North Glasgow University Hospitals NHS Trust, Stobhill Hospital, Glasgow, UK
| | - Thomas Sellers
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Brooke Fridley
- Department of Biostatistics, University of Kansas Medical Center, Kansas City, KS, USA
| | - Julie Cunningham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Joellen M. Schildkraut
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
- Cancer Control and Population Sciences, Duke Cancer Institute, Durham, NC, USA
| | - Ed Iversen
- Department of Statistical Science, Duke University, Durham, NC, USA
| | - Rachel Palmieri Weber
- Department of Community and Family Medicine, Duke University Medical Center, Durham, NC, USA
| | - Donal Brennan
- Queensland Centre for Gynaecological Cancer, Brisbane, Australia
| | - Andrew Berchuck
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, USA
| | - Paul Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK
| | - Paul Harnett
- Crown Princess Mary Cancer Centre and Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead Hospital, Sydney, Australia
| | | | - Michelle Haber
- Children's Cancer Institute Australia, Randwick, Australia
| | - Ellen L. Goode
- Department of Health Science Research, Division of Epidemiology, Mayo Clinic, Rochester, MN, USA
| | - Jason S. Lee
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kum Kum Khanna
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kerstin B. Meyer
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
| | | | - Anna deFazio
- Department of Gynaecological Oncology and Centre for Cancer Research, The Westmead Institute for Medical Research, The University of Sydney, Westmead Hospital, Sydney, Australia
| | | | | |
Collapse
|
46
|
Niu N, Wang L. In vitro human cell line models to predict clinical response to anticancer drugs. Pharmacogenomics 2015; 16:273-85. [PMID: 25712190 DOI: 10.2217/pgs.14.170] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
In vitro human cell line models have been widely used for cancer pharmacogenomic studies to predict clinical response, to help generate pharmacogenomic hypothesis for further testing, and to help identify novel mechanisms associated with variation in drug response. Among cell line model systems, immortalized cell lines such as Epstein-Barr virus (EBV)-transformed lymphoblastoid cell lines (LCLs) have been used most often to test the effect of germline genetic variation on drug efficacy and toxicity. Another model, especially in cancer research, uses cancer cell lines such as the NCI-60 panel. These models have been used mainly to determine the effect of somatic alterations on response to anticancer therapy. Even though these cell line model systems are very useful for initial screening, results from integrated analyses of multiple omics data and drug response phenotypes using cell line model systems still need to be confirmed by functional validation and mechanistic studies, as well as validation studies using clinical samples. Future models might include the use of patient-specific inducible pluripotent stem cells and the incorporation of 3D culture which could further optimize in vitro cell line models to improve their predictive validity.
Collapse
Affiliation(s)
- Nifang Niu
- Division of Clinical Pharmacology, Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | |
Collapse
|
47
|
Jack J, Havener TM, McLeod HL, Motsinger-Reif AA, Foster M. Evaluating the role of admixture in cancer therapy via in vitro drug response and multivariate genome-wide associations. Pharmacogenomics 2015; 16:1451-63. [PMID: 26314407 DOI: 10.2217/pgs.15.85] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM We investigate the role of ethnicity and admixture in drug response across a broad group of chemotherapeutic drugs. Also, we generate hypotheses on the genetic variants driving differential drug response through multivariate genome-wide association studies. METHODS Immortalized lymphoblastoid cell lines from 589 individuals (Hispanic or non-Hispanic/Caucasian) were used to investigate dose-response for 28 chemotherapeutic compounds. Univariate and multivariate statistical models were used to elucidate associations between genetic variants and differential drug response as well as the role of ethnicity in drug potency and efficacy. RESULTS & CONCLUSION For many drugs, the variability in drug response appears to correlate with self-reported race and estimates of genetic ancestry. Additionally, multivariate genome-wide association analyses offered interesting hypotheses governing these differential responses.
Collapse
Affiliation(s)
- John Jack
- Department of Statistics, North Carolina State University, 2601 Stinson Drive, Raleigh, NC 27695, USA.,Bioinformatics Research Center, North Carolina State University, 2601 Stinson Drive, Raleigh, NC 27695, USA
| | - Tammy M Havener
- Center for Pharmacogenomics & Individualized Therapy, University of North Carolina, 120 Mason Farm Road, Chapel Hill, NC 27514, USA
| | - Howard L McLeod
- DeBartolo Family Personalized Medicine Institute, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.,Pharmacogenetics for Every Nation Initiative, 1119 Oxbridge Drive, Tampa, FL 33549, USA
| | - Alison A Motsinger-Reif
- Department of Statistics, North Carolina State University, 2601 Stinson Drive, Raleigh, NC 27695, USA.,Bioinformatics Research Center, North Carolina State University, 2601 Stinson Drive, Raleigh, NC 27695, USA
| | - Matthew Foster
- Lineberger Comprehensive Cancer Center, University of North Carolina, 101 Manning Drive, Chapel Hill, NC 27514, USA
| |
Collapse
|
48
|
Meng L, Tian Z, Wang Y, Liu Y, Liu J. Predictive and prognostic molecular markers for cholangiocarcinoma in Han Chinese population. Int J Clin Exp Med 2015; 8:13680-9. [PMID: 26550313 PMCID: PMC4612998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/03/2015] [Indexed: 06/05/2023]
Abstract
Cholangiocarcinoma (CCA) is the most common malignant heterogeneous polygenetic carcinoma with a high incidence in Asia. Most patients would die within 1 year after diagnosis and the 5 year survival rate is less than 10-20% worldwide. Single nucleotide polymorphisms (SNPs) in genes regulate telomere maintenance, mitosis, and inflammation, and may help predict individual susceptibility to certain drugs, environmental factor, and risks to particular diseases. The gene-gene interaction and the regulation of SNPs have not been assessed extensively in CCA. According to our previous study, the GRB2-associated-binding protein (Gab1) gene rs3805246 (X(2) =5.015, P=0.025, OR=0.531, 95% CI 0.304-0.928) and epidermal growth factor receptor (EGFR) gene rs2007000 (X(2) =7.934, P=0.005, OR=2.148, 95% CI 1.255-3.675) presented significant difference between CCA patients and controls. This study conducted a population-based analysis using 225 CCA cases (153 biliary tract cancer patients and 72 gall bladder cancer patients) to assess the association between SNPs and progression of CCA patients, including the overall survival and the prognosis analysis. Results showed that an increased susceptibility of BTC was significantly associated with SNP loci distribution frequency in EGFR rs2107000 (X(2) =7.934, P=0.005, OR=2.148, 95% CI 1.255-3.675). Furthermore, multivariate factor regression analysis represented cholelithiasis medical history of BTC patients can be an effective evaluation criteria of BTC susceptibility in early stage. This study also assessed the relationship between these genotypic polymorphisms and clinicopathologic data, including tumor differentiation stage and overall survival. This is the first study identifying that EGFR polymorphisms are associated with BTC and EGFR rs2017000 polymorphisms may be an important survival predictor in BTC patients.
Collapse
Affiliation(s)
- Lingqin Meng
- Department of General Surgery, Shengjing Hospital of China Medical University Shenyang 110004, Liaoning, China
| | - Zhong Tian
- Department of General Surgery, Shengjing Hospital of China Medical University Shenyang 110004, Liaoning, China
| | - Yong Wang
- Department of General Surgery, Shengjing Hospital of China Medical University Shenyang 110004, Liaoning, China
| | - Yuan Liu
- Department of General Surgery, Shengjing Hospital of China Medical University Shenyang 110004, Liaoning, China
| | - Jingang Liu
- Department of General Surgery, Shengjing Hospital of China Medical University Shenyang 110004, Liaoning, China
| |
Collapse
|
49
|
Genome-wide Association Study on Platinum-induced Hepatotoxicity in Non-Small Cell Lung Cancer Patients. Sci Rep 2015; 5:11556. [PMID: 26100964 PMCID: PMC4477405 DOI: 10.1038/srep11556] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/22/2015] [Indexed: 01/09/2023] Open
Abstract
Platinum-based chemotherapy has been shown to improve the survival of advanced non-small cell lung cancer (NSCLC) patients; the platinum-induced toxicity severely impedes the success of chemotherapy. Genetic variations, such as single nucleotide polymorphisms (SNPs), may contribute to patients’ responses to the platinum-based chemotherapy. To identify SNPs that modify the risk of hepatotoxicity in NSCLC patients receiving platinum-based chemotherapy, we performed a genome-wide association scan in 334 subjects followed by a replication study among 375 subjects. Consistent associations with platinum-induced hepatotoxicity risk was identified for SNP rs2838566 located at 21q22.3, as the minor A allele could significantly increase the risk of liver injury (OR = 3.78, 95%CI = 1.99–7.19, P = 4.90 × 10−5 for GWAS scan, OR = 1.89, 95%CI = 1.03–3.46, P = 0.039 for replication, and OR = 2.56, 95%CI = 1.65–3.95, P = 2.55 × 10−5 for pooled population). These results suggested that genetic variants at 21q22.3 may contribute to the susceptibility of platinum-induced hepatotoxicity in NSCLC patients.
Collapse
|
50
|
Zhou Y, Wang M, Wu J, Jie Z, Chang S, Shuang T. The clinicopathological significance of miR-1307 in chemotherapy resistant epithelial ovarian cancer. J Ovarian Res 2015; 8:23. [PMID: 25887170 PMCID: PMC4449560 DOI: 10.1186/s13048-015-0143-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 03/12/2015] [Indexed: 01/29/2023] Open
Abstract
Background We aimed to examine the expression of miR-1307 in chemosensitive and chemoresistant epithelial ovarian cancer tissues and cell lines and to analyze the clinicopathological significance of miR-1307 in ovarian cancer. Methods MicroRNA microarray was used to screen differentially expressed microRNAs between the chemosensitive and chemoresistant epithelial ovarian cancer tissues. RT-PCR was used to validate the candidate microRNA. The potential target genes and their enriched biological pathways of microRNA were also analyzed. Dual Luciferase Reporter Gene Assay was conducted to validate the regulation of miRNA-1307 on the 3’-UTR of DAPK3. Results miRNA-1307 was up-regulated in the chemoresistant epithelial ovarian cancer tissues compared to the chemosensitive counterparts. The up-regulation of miRNA-1307 was not associated with menopause, tumor differentiation state, clinical stage, and lymph node metastasis of ovarian cancer. Gene ontology analysis of miR-1307 candidate target genes indicated that miR-1307 candidate target genes were enriched in the processes of cell proliferation and differentiation, nucleotide synthesis and metabolism, and lymphocytes activation. Conclusion Our results suggest that miRNA-1307 may play a role in the development of chemoresistance in ovarian cancer. Electronic supplementary material The online version of this article (doi:10.1186/s13048-015-0143-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yingying Zhou
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, P.R. China.
| | - Min Wang
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, P.R. China.
| | - Jianlei Wu
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, P.R. China.
| | - Zhihui Jie
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, P.R. China.
| | - Shuang Chang
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, P.R. China.
| | - Ting Shuang
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, 36 Sanhao Street, Shenyang, Liaoning, 110004, P.R. China.
| |
Collapse
|