1
|
Lin G, Tian F, Yu Q, Weng X, Yu N, Zhang F, Yi C, Ye J, Ye D. IL-17RA/CTSK axis mediates H. pylori-induced castration-resistant prostate cancer growth. Oncogene 2024:10.1038/s41388-024-03169-z. [PMID: 39424989 DOI: 10.1038/s41388-024-03169-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
In this investigation, we explored the molecular dynamics guiding the progression of castration-resistant prostate cancer (CRPC) influenced by Helicobacter pylori (H. pylori)-mediated M2 polarization of macrophages through the IL-17RA/CTSK/EMT axis. An 830-patient clinical trial categorized subjects into hormone-sensitive prostate cancer (HSPC) and CRPC groups. H. pylori infection, evaluated by ELISA, exhibited a higher incidence in CRPC patients, impacting overall survival (OS) and progression-free survival. In-depth in vitro and in vivo experiments, including 16S rDNA sequencing, immunohistochemical tests, and transcriptome analysis, unveiled that H. pylori promotes CRPC growth and metastasis by upregulating IL-17RA and CTSK, leading to enhanced EMT. Notably, M2 macrophages emerged as pivotal immune cells influencing CRPC progression. This study uncovers a novel pathway wherein H. pylori enrichment exacerbates CRPC by inducing macrophage M2 polarization, IL-17RA/CTSK expression, and EMT activation, shedding light on a previously unrecognized mechanism contributing to the growth and metastasis of CRPC.
Collapse
Affiliation(s)
- Guowen Lin
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Feng Tian
- Department Of Urology, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Qiwei Yu
- Department of Urology, Kunshan Hospital of Traditional Chinese Medicine, Suzhou, 215399, China
| | - Xiaoling Weng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200001, China
| | - Nanhui Yu
- Department of Gastrointestinal Surgery, the Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Feng Zhang
- Department Of Urology, Shanghai Eighth People's Hospital, Shanghai, 200235, China
| | - Chen Yi
- Department of Urology, Changsha Central Hospital Affiliated to University of South China, Changsha, 410000, China
| | - Jian Ye
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA, USA
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Suman M, Löfgren M, Fransson S, Yousuf JI, Svensson J, Djos A, Martinsson T, Kogner P, Kling T, Carén H. Altered methylation of imprinted genes in neuroblastoma: implications for prognostic refinement. J Transl Med 2024; 22:808. [PMID: 39217334 PMCID: PMC11366169 DOI: 10.1186/s12967-024-05634-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Neuroblastoma (NB) is a complex disease, and the current understanding of NB biology is limited. Deregulation in genomic imprinting is a common event in malignancy. Since imprinted genes play crucial roles in early fetal growth and development, their role in NB pathogenesis could be suggested. METHODS We examined alterations in DNA methylation patterns of 369 NB tumours at 49 imprinted differentially methylated regions (DMRs) and assessed its association with overall survival probabilities and selected clinical and genomic features of the tumours. In addition, an integrated analysis of DNA methylation and allele-specific copy number alterations (CNAs) was performed, to understand the correlation between the two molecular events. RESULTS Several imprinted regions with aberrant methylation patterns in NB were identified. Regions that underwent loss of methylation in > 30% of NB samples were DMRs annotated to the genes NDN, SNRPN, IGF2, MAGEL2 and HTR5A and regions with gain of methylation were NNAT, RB1 and GPR1. Methylation alterations at six of the 49 imprinted DMRs were statistically significantly associated with reduced overall survival: MIR886, RB1, NNAT/BLCAP, MAGEL2, MKRN3 and INPP5F. RB1, NNAT/BLCAP and MKRN3 were further able to stratify low-risk NB tumours i.e. tumours that lacked MYCN amplification and 11q deletion into risk groups. Methylation alterations at NNAT/BLCAP, MAGEL2 and MIR886 predicted risk independently of MYCN amplification or 11q deletion and age at diagnosis. Investigation of the allele-specific CNAs demonstrated that the imprinted regions that displayed most alterations in NB tumours harbor true epigenetic changes and are not result of the underlying CNAs. CONCLUSIONS Aberrant methylation in imprinted regions is frequently occurring in NB tumours and several of these regions have independent prognostic value. Thus, these could serve as potentially important clinical epigenetic markers to identify individuals with adverse prognosis. Incorporation of methylation status of these regions together with the established risk predictors may further refine the prognostication of NB patients.
Collapse
Affiliation(s)
- Medha Suman
- Sahlgrenska Center for Cancer Research, Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1F, 405 30, Gothenburg, Sweden
| | - Maja Löfgren
- Sahlgrenska Center for Cancer Research, Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1F, 405 30, Gothenburg, Sweden
| | - Susanne Fransson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jewahri Idris Yousuf
- Sahlgrenska Center for Cancer Research, Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1F, 405 30, Gothenburg, Sweden
| | - Johanna Svensson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Anna Djos
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tommy Martinsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Per Kogner
- Childhood Cancer Research Unit, Women's, and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Teresia Kling
- Sahlgrenska Center for Cancer Research, Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1F, 405 30, Gothenburg, Sweden
| | - Helena Carén
- Sahlgrenska Center for Cancer Research, Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 1F, 405 30, Gothenburg, Sweden.
| |
Collapse
|
3
|
朱 权, 黄 柏, 位 磊, 罗 奇. [Overexpression of LncRNA MEG3 promotes ferroptosis and enhances chemotherapy sensitivity of hepatocellular carcinoma cells to cisplatin]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:17-24. [PMID: 38293972 PMCID: PMC10878888 DOI: 10.12122/j.issn.1673-4254.2024.01.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Indexed: 02/01/2024]
Abstract
OBJECTIVE To investigate the effect of overexpression of LncRNA MEG3 on proliferation, migration and cisplatin sensitivity of hepatoma cells HepG2 and LM3 and explore the underlying and mechanism. METHODS The expression of MEG3 in healthy individuals and patients with hepatocellular carcinoma (HCC) was analyzed by online bioinformatics analysis, and Real-time fluorescence quantitative PCR (qRT-PCR) was used to detect MEG3 expression in different HCC cell lines. A MEG3-overexpresing plasmid was transfected in HepG2 and LM3 cells, and the changes in cell proliferation and migration were examined using CCK8 assay and scratch assay. CCK8 assay was used to determine the inhibitory rate of cisplatin on the transfected cells. A reactive oxygen species (ROS) fluorescence probe (DCFH-DA) and malondialdehyde (MDA) kit were used to assess the changes in ROS production and MDA level in the cells. Western blotting was performed to detect the expression levels of ferroptosis-related proteins glutathione peroxidase 4 (GPX4) and ferritin heavy chain 1 (FTH1). RESULTS The expression of MEG3 was significantly lower in HCC cells than in LO2 cells, which was consistent with the results of bioinformatic analysis (P < 0.05). Overexpression of MEG3 in the HCC cell lines significantly suppressed cell proliferation and migration (P < 0.05), increased the cell inhibition rate of cisplatin (P < 0.05), enhanced cellular ROS production and increased MDA levels in the cells (P < 0.05). MEG3 overexpression significantly decreased the expressions of GPX4 and FTH1 in the HCC cell lines. CONCLUSION The expression of MEG3 is decreased in HCC cells, and its overexpression inhibits proliferation and migration and enhances cisplatin sensitivity of HCC cells by promoting ferroptosis of the cells.
Collapse
Affiliation(s)
- 权 朱
- 中南大学基础医学院免疫学系,湖南 长沙 410008Department of Immunology, School of Basic Medical Sciences, Central South University, Changsha 410008, China
| | - 柏胜 黄
- 中南大学基础医学院生理学系,湖南 长沙 410008Department of Physiology, School of Basic Medical Sciences, Central South University, Changsha 410008, China
| | - 磊艳 位
- 中南大学基础医学院免疫学系,湖南 长沙 410008Department of Immunology, School of Basic Medical Sciences, Central South University, Changsha 410008, China
| | - 奇志 罗
- 中南大学基础医学院免疫学系,湖南 长沙 410008Department of Immunology, School of Basic Medical Sciences, Central South University, Changsha 410008, China
| |
Collapse
|
4
|
Tao W, Wang BY, Luo L, Li Q, Meng ZA, Xia TL, Deng WM, Yang M, Zhou J, Zhang X, Gao X, Li LY, He YD. A urine extracellular vesicle lncRNA classifier for high-grade prostate cancer and increased risk of progression: A multi-center study. Cell Rep Med 2023; 4:101240. [PMID: 37852185 PMCID: PMC10591064 DOI: 10.1016/j.xcrm.2023.101240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 07/03/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023]
Abstract
To construct a urine extracellular vesicle long non-coding RNA (lncRNA) classifier that can detect high-grade prostate cancer (PCa) of grade group 2 or greater and estimate the risk of progression during active surveillance, we identify high-grade PCa-specific lncRNAs by combined analyses of cohorts from TAHSY, TCGA, and the GEO database. We develop and validate a 3-lncRNA diagnostic model (Clnc, being made of AC015987.1, CTD-2589M5.4, RP11-363E6.3) that can detect high-grade PCa. Clnc shows higher accuracy than prostate cancer antigen 3 (PCA3), multiparametric magnetic resonance imaging (mpMRI), and two risk calculators (Prostate Cancer Prevention Trial [PCPT]-RC 2.0 and European Randomized Study of Screening for Prostate Cancer [ERSPC]-RC) in the training cohort (n = 350), two independent cohorts (n = 232; n = 251), and TCGA cohort (n = 499). In the prospective active surveillance cohort (n = 182), Clnc at diagnosis remains a powerful independent predictor for overall active surveillance progression. Thus, Clnc is a potential biomarker for high-grade PCa and can also serve as a biomarker for improved selection of candidates for active surveillance.
Collapse
Affiliation(s)
- Wen Tao
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Bang-Yu Wang
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200080, China
| | - Liang Luo
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Qing Li
- Food and Nutritional Sciences Programme, School of Life Sciences, The Chinese University of Hong Kong, Shatin 999077, Hong Kong
| | - Zhan-Ao Meng
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Tao-Lin Xia
- Department of Urology, Foshan First Municipal People's Hospital, Sun Yat-sen University, Foshan 528000, China
| | - Wei-Ming Deng
- Department of Urology, The First Affiliated Hospital, University of South China, Hengyang 421000, China
| | - Ming Yang
- Department of Urology, Foshan Municipal Chinese Medicine Hospital, Foshan 528000, China
| | - Jing Zhou
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xin Zhang
- Department of Pathology, Foshan First Municipal People's Hospital, Sun Yat-sen University, Foshan 528000, China
| | - Xin Gao
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Liao-Yuan Li
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Ya-Di He
- Health Management Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
5
|
Bozgeyik E. Variations in genomic regions encoding long non-coding RNA genes associated with increased prostate cancer risk. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2023; 791:108456. [PMID: 36948485 DOI: 10.1016/j.mrrev.2023.108456] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/24/2023]
Abstract
From a single restriction fragment length polymorphism analysis to next generation sequencing analysis that screens the entire human genome, testing for genomic variations provides a great and robust approach to cancer testing. Non-coding RNAs have been shown to have a major impact on the development and progression of human cancers, including prostate cancer. However, the low stability of these molecules under laboratory conditions has made their clinical utility challenging, as in the case of PCA3 long non-coding RNA. Since testing for variations in genomic regions encoding non-coding RNAs offers a promising approach for cancer testing, identification and interpretation of single nucleotide polymorphisms associated with prostate cancer susceptibility is of great interest. Accordingly, here, for the first time, we review and discuss current available knowledge about genomic variation of long non-coding RNA molecules in prostate cancer.
Collapse
Affiliation(s)
- Esra Bozgeyik
- Department of Medical Services and Techniques, Vocational School of Health Services, Adiyaman University, Adiyaman, Turkey.
| |
Collapse
|
6
|
Liang X, Fu Z, Tang L, Zheng M, Chen D, Liu A, Shi L, Yang L, Shao C, Dong X. PLAGL1 is associated with prognosis and cell proliferation in pancreatic adenocarcinoma. BMC Gastroenterol 2023; 23:2. [PMID: 36600208 PMCID: PMC9811725 DOI: 10.1186/s12876-022-02609-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 12/08/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Emerging evidence has shown the crucial roles of pleomorphic adenoma gene (PLAG) family genes in multiple cancers. However, their functions and mechanisms in pancreatic adenocarcinoma (PAAD) remain poorly understood. METHODS We analyzed the expression levels of PLAG family genes in both The Cancer Genome Atlas (TCGA) database and a Gene Expression Omnibus (GEO) database, and confirmed the results in our three independent cohorts of 382 PAAD tissues and 362 adjacent nontumor pancreatic tissues. Integrated analyses were carried out to explore the function, mechanism and prognostic value of the selected PLAG family gene in PAAD patients. RESULTS By analyzing the TCGA and GEO databases, PLAGL1 was identified to be downregulated in PAAD tissues, and its decreasing levels of both mRNA and protein were verified in our three independent PAAD cohorts. PLAGL1 expression was inversely correlated with clinicopathological factors including the Ki67+ cell rate and pathologic stage. Further GSEA of the TCGA-PAAD cohort demonstrated that multiple signaling pathways implicated in cell proliferation were enriched in the lower PLAGL1 expressing PAAD group. Moreover, we demonstrated that PLAGL1 expression was obviously negatively associated with patients' overall survival outcome in both the TCGA-PAAD cohort and our verification cohorts. Additionally, through MTS and BrdU assays, we further demonstrated in vitro that PLAGL1 had the impact of preventing the proliferation of pancreatic cancer cells. CONCLUSIONS Our present study suggested that downregulated PLAGL1 might act as a biomarker in predicts poor prognosis and one of important factors in increasing cell proliferation in PAAD. This study provides us with a novel prognostic marker and therapeutic strategy for PAAD, which deserves further study.
Collapse
Affiliation(s)
- Xing Liang
- grid.429222.d0000 0004 1798 0228Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu Province China ,grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Fengyang Road 415, Shanghai, 200003 China
| | - Zhiping Fu
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Fengyang Road 415, Shanghai, 200003 China
| | - Liang Tang
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Fengyang Road 415, Shanghai, 200003 China
| | - Minghui Zheng
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Fengyang Road 415, Shanghai, 200003 China
| | - Danlei Chen
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Fengyang Road 415, Shanghai, 200003 China
| | - Anan Liu
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Fengyang Road 415, Shanghai, 200003 China
| | - Ligang Shi
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Fengyang Road 415, Shanghai, 200003 China
| | - Linhua Yang
- grid.16821.3c0000 0004 0368 8293Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127 China
| | - Chenghao Shao
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Fengyang Road 415, Shanghai, 200003 China
| | - Xiaoqiang Dong
- grid.429222.d0000 0004 1798 0228Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006 Jiangsu Province China
| |
Collapse
|
7
|
Molecular Basis of Beckwith–Wiedemann Syndrome Spectrum with Associated Tumors and Consequences for Clinical Practice. Cancers (Basel) 2022; 14:cancers14133083. [PMID: 35804856 PMCID: PMC9265096 DOI: 10.3390/cancers14133083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/19/2022] [Accepted: 06/20/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Beckwith–Wiedemann syndrome (BWS, OMIM 130650) is an inborn overgrowth disorder caused by molecular alterations in chromosome 11p15.5. These molecular changes affect so-called imprinted genes, i.e., genes which underlie a complex regulation which is linked to the parental origin of the gene copy. Thus, either the maternal gene copy is expressed or the paternal, but this balanced regulation is prone to disturbances. In fact, different types of molecular variants have been identified in BWS, resulting in a variable phenotype; thus, it was consented that the syndromic entity was extended to the Beckwith–Wiedemann spectrum (BWSp). Some molecular subgroups of BWSp are associated with an increased embryonic tumor risk and have different likelihoods for specific tumors. Therefore, the precise determination of the molecular subgroup is needed for precise monitoring and treatment, but the molecular diagnostic procedure has several limitations and challenges which have to be considered. Abstract Beckwith–Wiedemann syndrome (BWS, OMIM 130650) is a congenital imprinting condition with a heterogenous clinical presentation of overgrowth and an increased childhood cancer risk (mainly nephroblastoma, hepatoblastoma or neuroblastoma). Due to the varying clinical presentation encompassing classical, clinical BWS without a molecular diagnosis and BWS-related phenotypes with an 11p15.5 molecular anomaly, the syndromic entity was extended to the Beckwith–Wiedemann spectrum (BWSp). The tumor risk of up to 30% depends on the molecular subtype of BWSp with causative genetic or epigenetic alterations in the chromosomal region 11p15.5. The molecular diagnosis of BWSp can be challenging for several reasons, including the range of causative molecular mechanisms which are frequently mosaic. The molecular basis of tumor formation appears to relate to stalled cellular differentiation in certain organs that predisposes persisting embryonic cells to accumulate additional molecular defects, which then results in a range of embryonal tumors. The molecular subtype of BWSp not only influences the overall risk of neoplasia, but also the likelihood of specific embryonal tumors.
Collapse
|
8
|
Chirom K, Malik MZ, Mangangcha IR, Somvanshi P, Singh RKB. Network medicine in ovarian cancer: topological properties to drug discovery. Brief Bioinform 2022; 23:6555408. [PMID: 35352113 DOI: 10.1093/bib/bbac085] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 02/11/2022] [Accepted: 02/20/2022] [Indexed: 12/21/2022] Open
Abstract
Network medicine provides network theoretical tools, methods and properties to study underlying laws governing human interactome to identify disease states and disease complexity leading to drug discovery. Within this framework, we investigated the topological properties of ovarian cancer network (OCN) and the roles of hubs to understand OCN organization to address disease states and complexity. The OCN constructed from the experimentally verified genes exhibits fractal nature in the topological properties with deeply rooted functional communities indicating self-organizing behavior. The network properties at all levels of organization obey one parameter scaling law which lacks centrality lethality rule. We showed that $\langle k\rangle $ can be taken as a scaling parameter, where, power law exponent can be estimated from the ratio of network diameters. The betweenness centrality $C_B$ shows two distinct behaviors one shown by high degree hubs and the other by segregated low degree nodes. The $C_B$ power law exponent is found to connect the exponents of distributions of high and low degree nodes. OCN showed the absence of rich-club formation which leads to the missing of a number of attractors in the network causing formation of weakly tied diverse functional modules to keep optimal network efficiency. In OCN, provincial and connector hubs, which includes identified key regulators, take major responsibility to keep the OCN integrity and organization. Further, most of the key regulators are found to be over expressed and positively correlated with immune infiltrates. Finally, few potential drugs are identified related to the key regulators.
Collapse
Affiliation(s)
- Keilash Chirom
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India.,Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, 110019, India
| | - Md Zubbair Malik
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India
| | | | - Pallavi Somvanshi
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India
| | - R K Brojen Singh
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi-110067, India
| |
Collapse
|
9
|
Javier-DesLoges J, McKay RR, Swafford AD, Sepich-Poore GD, Knight R, Parsons JK. The microbiome and prostate cancer. Prostate Cancer Prostatic Dis 2022; 25:159-164. [PMID: 34267333 PMCID: PMC8767983 DOI: 10.1038/s41391-021-00413-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/24/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022]
Abstract
There is growing evidence that the microbiome is involved in development and treatment of many human diseases, including prostate cancer. There are several potential pathways for microbiome-based mechanisms for the development of prostate cancer: direct impacts of microbes or microbial products in the prostate or the urine, and indirect impacts from microbes or microbial products in the gastrointestinal tract. Unique microbial signatures have been identified within the stool, oral cavity, tissue, urine, and blood of prostate cancer patients, but studies vary in their findings. Recent studies describe potential diagnostic and therapeutic applications of the microbiome, but further clinical investigation is needed. In this review, we explore the existing literature on the discovery of the human microbiome and its relationship to prostate cancer.
Collapse
Affiliation(s)
| | - Rana R McKay
- Department of Urology, UC San Diego Health, La Jolla, CA, USA
- Department of Medicine, Division of Hematology/Oncology, UC San Diego Health, La Jolla, CA, USA
| | | | | | - Rob Knight
- Department of Bioengineering, UC San Diego, La Jolla, CA, USA
| | | |
Collapse
|
10
|
Zhou C, Cao H, Meng X, Zhang Q. Lnc-MEG3 inhibits invasion, migration, and epithelial- mesenchymal transition of nasopharyngeal carcinoma cells by regulating sequestosome 1. Head Neck 2021; 44:201-211. [PMID: 34704649 DOI: 10.1002/hed.26917] [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] [Received: 04/13/2021] [Revised: 09/24/2021] [Accepted: 10/18/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Long non-coding RNAs regulate malignant behaviors of nasopharyngeal carcinoma (NPC). We aim to investigate the roles and mechanisms of long non-coding RNA maternally expressed gene 3 (lnc-MEG3) in NPC. METHODS The expression levels of lnc-MEG3 and sequestosome 1 (SQSTM1) in NPC tissues and cell lines were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Cell invasion and migration abilities were evaluated using transwell and wound healing assays, respectively. RESULTS Downregulated lnc-MEG3 expression and upregulated SQSTM1 expression were found in NPC tissues and cells. Overexpression of lnc-MEG3 inhibited invasion, migration, and epithelial-mesenchymal transition in NPC cells. Overexpression of lnc-MEG3 reduced the expression level of SQSTM1, and SQSTM1 expression was inversely correlated with lnc-MEG3 level in NPC tissues. Besides, overexpression of SQSTM1 reversed the effects of lnc-MEG3 overexpression. Moreover, knockdown of lnc-MEG3 enhanced NPC progression while its effects were eased by SQSTM1 silence. CONCLUSION Lnc-MEG3 inhibits malignant behaviors by regulating SQSTM1 expression. It may serve as a therapeutic target to treat NPC.
Collapse
Affiliation(s)
- Caifeng Zhou
- Department of Oncology, Shouguang People's Hospital, Weifang, China
| | - Huiling Cao
- Departments of Head and Neck Surgery, Chengwu Hospital Affiliated to Shandong First Medical University, Heze, China
| | - Xinyu Meng
- Department of Otolaryngology, Jinan People's Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiyao Zhang
- Department of Pharmacy, Jinan Municipal Hospital of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
11
|
The Role of Microbial Factors in Prostate Cancer Development-An Up-to-Date Review. J Clin Med 2021; 10:jcm10204772. [PMID: 34682893 PMCID: PMC8538262 DOI: 10.3390/jcm10204772] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/03/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Up-to-date studies emphasize the role of human urinary and intestinal microbiome in maintaining urogenital health. Both microbial flora and sexually transmitted pathogens may affect metabolic or immune mechanisms and consequently promote or inhibit prostate carcinogenesis. Hereby, we review the most current evidence regarding the microbial factors and their link to prostate cancer. We conducted a literature search up to December 2020. The microbial impact on prostate cancer initiation and progression is complex. The proposed mechanisms of action include induction of chronic inflammatory microenvironment (Propionibacterium spp., sexually-transmitted pathogens) and direct dysregulation of cell cycle (Helicobacter pylori, Kaposi’s sarcoma-associated herpesvirus- KSHV, human papilloma virus 18- HPV18). Suppression of immune cell expression and downregulating immune-associated genes are also observed (Gardnerella vaginalis). Additionally, the impact of the gut microbiome proved relevant in promoting tumorigenesis (Bacteroides massiliensis). Nevertheless, certain microbes appear to possess anti-tumor properties (Listeria monocytogenes, Pseudomonas spp.), such as triggering a robust immune response and apoptotic cancer cell death. The role of microbial factors in prostate cancer development is an emerging field that merits further studies. In the future, translating microbial research into clinical action may prove helpful in predicting diagnosis and potential outcomes of the disease.
Collapse
|
12
|
Mendiola AJP, LaSalle JM. Epigenetics in Prader-Willi Syndrome. Front Genet 2021; 12:624581. [PMID: 33659026 PMCID: PMC7917289 DOI: 10.3389/fgene.2021.624581] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/18/2021] [Indexed: 11/16/2022] Open
Abstract
Prader-Willi Syndrome (PWS) is a rare neurodevelopmental disorder that affects approximately 1 in 20,000 individuals worldwide. Symptom progression in PWS is classically characterized by two nutritional stages. Stage 1 is hypotonia characterized by poor muscle tone that leads to poor feeding behavior causing failure to thrive in early neonatal life. Stage 2 is followed by the development of extreme hyperphagia, also known as insatiable eating and fixation on food that often leads to obesity in early childhood. Other major features of PWS include obsessive-compulsive and hoarding behaviors, intellectual disability, and sleep abnormalities. PWS is genetic disorder mapping to imprinted 15q11.2-q13.3 locus, specifically at the paternally expressed SNORD116 locus of small nucleolar RNAs and noncoding host gene transcripts. SNORD116 is processed into several noncoding components and is hypothesized to orchestrate diurnal changes in metabolism through epigenetics, according to functional studies. Here, we review the current status of epigenetic mechanisms in PWS, with an emphasis on an emerging role for SNORD116 in circadian and sleep phenotypes. We also summarize current ongoing therapeutic strategies, as well as potential implications for more common human metabolic and psychiatric disorders.
Collapse
Affiliation(s)
| | - Janine M. LaSalle
- Department of Medical Microbiology and Immunology, Genome Center, MIND Institute, University of California, Davis, Davis, CA, United States
| |
Collapse
|
13
|
Uddin MN, Wang X. The landscape of long non-coding RNAs in tumor stroma. Life Sci 2020; 264:118725. [PMID: 33166593 DOI: 10.1016/j.lfs.2020.118725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 02/06/2023]
Abstract
AIMS Long non-coding RNAs (lncRNAs) are associated with cancer development, while their relationship with the cancer-associated stromal components remains poorly understood. In this review, we performed a broad description of the functional landscape of stroma-associated lncRNAs in various cancers and their roles in regulating the tumor-stroma crosstalk. MATERIALS AND METHODS We carried out a systematic literature review of PubMed, Scopus, Medline, Bentham, and EMBASE (Elsevier) databases by using the keywords "LncRNAs in cancer," "LncRNAs in tumor stroma," "stroma," "cancer-associated stroma," "stroma in the tumor microenvironment," "tumor-stroma crosstalk," "drug resistance of stroma," and "stroma in immunosuppression" till July 2020. We collected the latest articles addressing the biological functions of stroma-associated lncRNAs in cancer. KEY FINDINGS These articles reported that dysregulated stroma-associated lncRNAs play significant roles in modulating the tumor microenvironment (TME) by the regulation of tumor-stroma crosstalk, epithelial to mesenchymal transition (EMT), endothelial to mesenchymal transition (EndMT), extracellular matrix (ECM) turnover, and tumor immunity. SIGNIFICANCE The tumor stroma is a substantial portion of the TME, and the dysregulation of tumor stroma-associated lncRNAs significantly contributes to cancer initiation, progression, angiogenesis, immune evasion, metastasis, and drug resistance. Thus, stroma-associated lncRNAs could be potentially useful targets for cancer therapy.
Collapse
Affiliation(s)
- Md Nazim Uddin
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China; Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka 1205, Bangladesh
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
14
|
Holly JMP, Biernacka K, Perks CM. The role of insulin-like growth factors in the development of prostate cancer. Expert Rev Endocrinol Metab 2020; 15:237-250. [PMID: 32441162 DOI: 10.1080/17446651.2020.1764844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 05/01/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Preclinical, clinical, and population studies have provided robust evidence for an important role for the insulin-like growth factor (IGF) system in the development of prostate cancer. AREAS COVERED An overview of the IGF system is provided. The evidence implicating the IGF system in the development of prostate cancer is summarized. The compelling evidence culminated in a number of clinical trials of agents targeting the system; the reasons for the failure of these trials are discussed. EXPERT OPINION Clinical trials of agents targeting the IGF system in prostate cancer were terminated due to limited objective clinical responses and are unlikely to be resumed unless a convincing predictive biomarker is identified that would enable the selection of likely responders. The aging population and increased screening will lead to greater diagnosis of prostate cancer. Although the vast majority will be indolent disease, the epidemics of obesity and diabetes will increase the proportion that progress to clinical disease. The increased population of worried men will result in more trials aimed to reduce the risk of disease progression; actual clinical endpoints will be challenging and the IGFs remain the best intermediate biomarkers to indicate a response that could alter the course of disease.
Collapse
Affiliation(s)
- Jeff M P Holly
- IGFs & Metabolic Endocrinology Group, Faculty of Health Sciences, School of Translational Health Science, University of Bristol, Southmead Hospital , Bristol, UK
| | - Kalina Biernacka
- IGFs & Metabolic Endocrinology Group, Faculty of Health Sciences, School of Translational Health Science, University of Bristol, Southmead Hospital , Bristol, UK
| | - Claire M Perks
- IGFs & Metabolic Endocrinology Group, Faculty of Health Sciences, School of Translational Health Science, University of Bristol, Southmead Hospital , Bristol, UK
| |
Collapse
|
15
|
Peng X, Zhang K, Ma L, Xu J, Chang W. The Role of Long Non-Coding RNAs in Thyroid Cancer. Front Oncol 2020; 10:941. [PMID: 32596158 PMCID: PMC7300266 DOI: 10.3389/fonc.2020.00941] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
Thyroid cancer, the most common endocrine malignancy, has become the most commonly diagnosed malignant solid tumor. Moreover, some cases have poor prognosis, and the survival period is only 3-5 months. Long noncoding RNAs (lncRNAs) are a group of functional RNA molecules more than 200 nucleotides in length that lack the ability to encode protein but participate in all aspects of gene regulation. Functionally, many lncRNAs play essential roles in epigenetic regulation at transcriptional and post-transcriptional levels via various molecular mechanisms. Recent studies have discovered important roles for lncRNAs during the complex process of carcinogenesis in thyroid cancer. In this review, we focus on lncRNAs dysregulated in thyroid cancer and summarize recently reported associations between lncRNAs and thyroid cancer in order to demonstrate the significant value of lncRNAs in diagnosis and treatment.
Collapse
Affiliation(s)
- Xuejiao Peng
- Department of Thyroid Surgery, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Kun Zhang
- Medical Research Center, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Li Ma
- Department of Thyroid Surgery, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Junfeng Xu
- Department of Thyroid Surgery, Second Affiliated Hospital of Jilin University, Changchun, China
| | - Weiqin Chang
- Department of Thyroid Surgery, Second Affiliated Hospital of Jilin University, Changchun, China
| |
Collapse
|
16
|
Argyraki M, Damdimopoulou P, Chatzimeletiou K, Grimbizis GF, Tarlatzis BC, Syrrou M, Lambropoulos A. In-utero stress and mode of conception: impact on regulation of imprinted genes, fetal development and future health. Hum Reprod Update 2020; 25:777-801. [PMID: 31633761 DOI: 10.1093/humupd/dmz025] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/04/2019] [Accepted: 07/12/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Genomic imprinting is an epigenetic gene regulatory mechanism; disruption of this process during early embryonic development can have major consequences on both fetal and placental development. The periconceptional period and intrauterine life are crucial for determining long-term susceptibility to diseases. Treatments and procedures in assisted reproductive technologies (ART) and adverse in-utero environments may modify the methylation levels of genomic imprinting regions, including insulin-like growth factor 2 (IGF2)/H19, mesoderm-specific transcript (MEST), and paternally expressed gene 10 (PEG10), affecting the development of the fetus. ART, maternal psychological stress, and gestational exposures to chemicals are common stressors suspected to alter global epigenetic patterns including imprinted genes. OBJECTIVE AND RATIONALE Our objective is to highlight the effect of conception mode and maternal psychological stress on fetal development. Specifically, we monitor fetal programming, regulation of imprinted genes, fetal growth, and long-term disease risk, using the imprinted genes IGF2/H19, MEST, and PEG10 as examples. The possible role of environmental chemicals in genomic imprinting is also discussed. SEARCH METHODS A PubMed search of articles published mostly from 2005 to 2019 was conducted using search terms IGF2/H19, MEST, PEG10, imprinted genes, DNA methylation, gene expression, and imprinting disorders (IDs). Studies focusing on maternal prenatal stress, psychological well-being, environmental chemicals, ART, and placental/fetal development were evaluated and included in this review. OUTCOMES IGF2/H19, MEST, and PEG10 imprinted genes have a broad developmental effect on fetal growth and birth weight variation. Their disruption is linked to pregnancy complications, metabolic disorders, cognitive impairment, and cancer. Adverse early environment has a major impact on the developing fetus, affecting mostly growth, the structure, and subsequent function of the hypothalamic-pituitary-adrenal axis and neurodevelopment. Extensive evidence suggests that the gestational environment has an impact on epigenetic patterns including imprinting, which can lead to adverse long-term outcomes in the offspring. Environmental stressors such as maternal prenatal psychological stress have been found to associate with altered DNA methylation patterns in placenta and to affect fetal development. Studies conducted during the past decades have suggested that ART pregnancies are at a higher risk for a number of complications such as birth defects and IDs. ART procedures involve multiple steps that are conducted during critical windows for imprinting establishment and maintenance, necessitating long-term evaluation of children conceived through ART. Exposure to environmental chemicals can affect placental imprinting and fetal growth both in humans and in experimental animals. Therefore, their role in imprinting should be better elucidated, considering the ubiquitous exposure to these chemicals. WIDER IMPLICATIONS Dysregulation of imprinted genes is a plausible mechanism linking stressors such as maternal psychological stress, conception using ART, and chemical exposures with fetal growth. It is expected that a greater understanding of the role of imprinted genes and their regulation in fetal development will provide insights for clinical prevention and management of growth and IDs. In a broader context, evidence connecting impaired imprinted gene function to common diseases such as cancer is increasing. This implies early regulation of imprinting may enable control of long-term human health, reducing the burden of disease in the population in years to come.
Collapse
Affiliation(s)
- Maria Argyraki
- First Department of Obstetrics and Gynecology, Laboratory of Genetics, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece
| | - Pauliina Damdimopoulou
- Karolinska Institutet, Department of Clinical Sciences, Intervention and Technology, Unit of Obstetrics and Gynecology, K57 Karolinska University Hospital Huddinge, SE-14186 Stockholm, Sweden
| | - Katerina Chatzimeletiou
- First Department of Obstetrics and Gynecology, Unit for Human Reproduction, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece
| | - Grigoris F Grimbizis
- First Department of Obstetrics and Gynecology, Unit for Human Reproduction, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece
| | - Basil C Tarlatzis
- First Department of Obstetrics and Gynecology, Unit for Human Reproduction, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece
| | - Maria Syrrou
- Department of Biology, Laboratory of Biology, School of Health Sciences, University of Ioannina, Dourouti University Campus, 45110, Ioannina, Greece
| | - Alexandros Lambropoulos
- First Department of Obstetrics and Gynecology, Laboratory of Genetics, School of Medicine, Aristotle University of Thessaloniki, Papageorgiou General Hospital, Ring Road, Nea Efkarpia, 56403 Thessaloniki, Greece
| |
Collapse
|
17
|
Shen R, Cheng T, Xu C, Yung RC, Bao J, Li X, Yu H, Lu S, Xu H, Wu H, Zhou J, Bu W, Wang X, Si H, Shi P, Zhao P, Liu Y, Deng Y, Zhu Y, Zeng S, Pineda JP, Lin C, Zhou N, Bai C. Novel visualized quantitative epigenetic imprinted gene biomarkers diagnose the malignancy of ten cancer types. Clin Epigenetics 2020; 12:71. [PMID: 32448196 PMCID: PMC7245932 DOI: 10.1186/s13148-020-00861-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/12/2020] [Indexed: 12/12/2022] Open
Abstract
Background Epigenetic alterations are involved in most cancers, but its application in cancer diagnosis is still limited. More practical and intuitive methods to detect the aberrant expressions from clinical samples using highly sensitive biomarkers are needed. In this study, we developed a novel approach in identifying, visualizing, and quantifying the biallelic and multiallelic expressions of an imprinted gene panel associated with cancer status. We evaluated the normal and aberrant expressions measured using the imprinted gene panel to formulate diagnostic models which could accurately distinguish the imprinting differences of normal and benign cases from cancerous tissues for each of the ten cancer types. Results The Quantitative Chromogenic Imprinted Gene In Situ Hybridization (QCIGISH) method developed from a 1013-case study which provides a visual and quantitative analysis of non-coding RNA allelic expressions identified the guanine nucleotide-binding protein, alpha-stimulating complex locus (GNAS), growth factor receptor-bound protein (GRB10), and small nuclear ribonucleoprotein polypeptide N (SNRPN) out of five tested imprinted genes as efficient epigenetic biomarkers for the early-stage detection of ten cancer types. A binary algorithm developed for cancer diagnosis showed that elevated biallelic expression (BAE), multiallelic expression (MAE), and total expression (TE) measurements for the imprinted gene panel were associated with cell carcinogenesis, with the formulated diagnostic models achieving consistently high sensitivities (91–98%) and specificities (86–98%) across the different cancer types. Conclusions The QCIGISH method provides an innovative way to visually assess and quantitatively analyze individual cells for cancer potential extending from hyperplasia and dysplasia until carcinoma in situ and invasion, which effectively supplements standard clinical cytologic and histopathologic diagnosis for early cancer detection. In addition, the diagnostic models developed from the BAE, MAE, and TE measurements of the imprinted gene panel GNAS, GRB10, and SNRPN could provide important predictive information which are useful in early-stage cancer detection and personalized cancer management.
Collapse
Affiliation(s)
- Rulong Shen
- Department of Pathology, Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Tong Cheng
- Epigenetics Lab, Chinese Alliance Against Lung Cancer, 6th Floor, Building 5, No.66, Jinghuidongdao Road, Wuxi, 214135, Jiangsu, China
| | - Chuanliang Xu
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - Rex C Yung
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21207, USA
| | - Jiandong Bao
- Departments of Endocrinology, Ultrasound and Pathology, JiangYuan Hospital Affiliated to Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu, China
| | - Xing Li
- Epigenetics Lab, Chinese Alliance Against Lung Cancer, 6th Floor, Building 5, No.66, Jinghuidongdao Road, Wuxi, 214135, Jiangsu, China
| | - Hongyu Yu
- Department of Pathology, Changzheng Hospital, Navy Medical University, Shanghai, 200003, China
| | - Shaohua Lu
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, 200072, China
| | - Huixiong Xu
- Department of Medical Ultrasound, Shanghai Tenth People's Hospital, Ultrasound Research and Education Institute, Tongji University School of Medicine, Shanghai, 200072, China
| | - Hongxun Wu
- Departments of Endocrinology, Ultrasound and Pathology, JiangYuan Hospital Affiliated to Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu, China
| | - Jian Zhou
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Wenbo Bu
- Hospital for Skin Disease, Institute of Dermatology, Chinese Academy of Medical Science, Peking Union Medical College, Nanjing, 210042, China
| | - Xiaonan Wang
- Epigenetics Lab, Chinese Alliance Against Lung Cancer, 6th Floor, Building 5, No.66, Jinghuidongdao Road, Wuxi, 214135, Jiangsu, China
| | - Han Si
- Epigenetics Lab, Chinese Alliance Against Lung Cancer, 6th Floor, Building 5, No.66, Jinghuidongdao Road, Wuxi, 214135, Jiangsu, China
| | - Panying Shi
- Epigenetics Lab, Chinese Alliance Against Lung Cancer, 6th Floor, Building 5, No.66, Jinghuidongdao Road, Wuxi, 214135, Jiangsu, China
| | - Pengcheng Zhao
- Epigenetics Lab, Chinese Alliance Against Lung Cancer, 6th Floor, Building 5, No.66, Jinghuidongdao Road, Wuxi, 214135, Jiangsu, China
| | - Yun Liu
- Epigenetics Lab, Chinese Alliance Against Lung Cancer, 6th Floor, Building 5, No.66, Jinghuidongdao Road, Wuxi, 214135, Jiangsu, China
| | - Yongjie Deng
- Epigenetics Lab, Chinese Alliance Against Lung Cancer, 6th Floor, Building 5, No.66, Jinghuidongdao Road, Wuxi, 214135, Jiangsu, China
| | - Yun Zhu
- Departments of Endocrinology, Ultrasound and Pathology, JiangYuan Hospital Affiliated to Jiangsu Institute of Nuclear Medicine, Wuxi, 214063, Jiangsu, China
| | - Shuxiong Zeng
- Department of Urology, Changhai Hospital, Navy Medical University, Shanghai, 200433, China
| | - John P Pineda
- Epigenetics Lab, Chinese Alliance Against Lung Cancer, 6th Floor, Building 5, No.66, Jinghuidongdao Road, Wuxi, 214135, Jiangsu, China
| | - Chunlin Lin
- Department of Molecular Medicine, The University of Texas Health Science Center at San Antonio, San Antonio, TX, 78245, USA
| | - Ning Zhou
- Epigenetics Lab, Chinese Alliance Against Lung Cancer, 6th Floor, Building 5, No.66, Jinghuidongdao Road, Wuxi, 214135, Jiangsu, China.
| | - Chunxue Bai
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
18
|
Banerjee S, Alwine JC, Wei Z, Tian T, Shih N, Sperling C, Guzzo T, Feldman MD, Robertson ES. Microbiome signatures in prostate cancer. Carcinogenesis 2020; 40:749-764. [PMID: 30794288 DOI: 10.1093/carcin/bgz008] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 11/21/2018] [Accepted: 02/01/2019] [Indexed: 12/20/2022] Open
Abstract
We have established a microbiome signature for prostate cancer using an array-based metagenomic and capture-sequencing approach. A diverse microbiome signature (viral, bacterial, fungal and parasitic) was observed in the prostate cancer samples compared with benign prostate hyperplasia controls. Hierarchical clustering analysis identified three distinct prostate cancer-specific microbiome signatures. The three signatures correlated with different grades, stages and scores of the cancer. Thus, microbiome signature analysis potentially provides clinical diagnosis and outcome predictions. The array data were validated by PCR and targeted next-generation sequencing (NGS). Specific NGS data suggested that certain viral genomic sequences were inserted into the host somatic chromosomes of the prostate cancer samples. A randomly selected group of these was validated by direct PCR and sequencing. In addition, PCR validation of Helicobacter showed that Helicobacter cagA sequences integrated within specific chromosomes of prostate tumor cells. The viral and Helicobacter integrations are predicted to affect the expression of several cellular genes associated with oncogenic processes.
Collapse
Affiliation(s)
- Sagarika Banerjee
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - James C Alwine
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, USA
| | - Tian Tian
- Department of Computer Science, New Jersey Institute of Technology, Newark, NJ, USA
| | - Natalie Shih
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Colin Sperling
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas Guzzo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Erle S Robertson
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
19
|
Gong R, Jiang Y. Non-coding RNAs in Pancreatic Ductal Adenocarcinoma. Front Oncol 2020; 10:309. [PMID: 32257946 PMCID: PMC7089935 DOI: 10.3389/fonc.2020.00309] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/20/2020] [Indexed: 12/15/2022] Open
Abstract
Non-coding RNAs (ncRNAs) are reported to be expressed in human cancers, including pancreatic ductal adenocarcinoma (PDAC). These ncRNAs affect the growth, migration and invasion of tumor cells by regulating cell cycle and apoptosis, as well as playing important roles in epigenetic processes, transcription and post-transcriptional regulation. It is still unclear whether alterations in ncRNAs influence PDAC development and progression. Because of this, analysis based on existing data on ncRNAs, which are crucial for modulating pancreatic tumorigenesis, will be important for future research on PDAC. Here, we summarize ncRNAs with tumor-promoting functions: HOTAIR, HOTTIP, MALAT1, lncRNA H19, lncRNA PVT1, circ-RNA ciRS-7, circ-0030235, circ-RNA_100782, circ-LDLRAD3, circ-0007534, circRHOT1, circZMYM2, circ-IARS, circ-RNA PDE8A, miR-21, miR-155, miR-221/222, miR-196b, miR-10a. While others including GAS5, MEG3, and lncRNA ENST00000480739, has_circ_0001649, miR-34a, miR-100, miR-217, miR-143 inhibit the proliferation and invasion of PDAC. Hence, we summarize the functions of ncRNAs in the occurrence, development and metastasis of PDAC, with the goal to provide guidance in the clinical diagnosis and treatment of PDAC.
Collapse
Affiliation(s)
- Ruining Gong
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yueping Jiang
- Department of Gastroenterology, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
20
|
Das R, Feng FY, Selth LA. Long non-coding RNAs in prostate cancer: Biological and clinical implications. Mol Cell Endocrinol 2019; 480:142-152. [PMID: 30391670 DOI: 10.1016/j.mce.2018.10.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/12/2018] [Accepted: 10/31/2018] [Indexed: 12/31/2022]
Abstract
Prostate cancer (PCa) is a major health issue in the Western world. Current clinical imperatives for this disease include better stratification of indolent versus aggressive disease to enable improved patient management, as well as the identification of more effective therapies for the prevention and treatment of metastatic and therapy-resistant PCa. The advent of next-generation transcriptomics led to the identification of an important class of molecules, long non-coding RNAs (lncRNAs). LncRNAs have critical functions in normal physiology, but their dysregulation has also been implicated in the development and progression of a variety of cancers, including PCa. Importantly, a subset of lncRNAs are highly prostate-specific, suggesting potential for utility as both biomarkers and therapeutic targets. In this review, we summarise the biology of lncRNAs and their mechanisms of action in the development and progression of prostate cancer. Additionally, we cast a critical eye over the potential for this class of molecules to impact on clinical practice.
Collapse
Affiliation(s)
- Rajdeep Das
- Department of Radiation Oncology, University of California San Francisco, USA; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, USA.
| | - Felix Y Feng
- Department of Radiation Oncology, University of California San Francisco, USA; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, USA; Department of Urology, University of California San Francisco, USA
| | - Luke A Selth
- Dame Roma Mitchell Cancer Research Laboratories, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia; Freemasons Foundation Centre for Men's Health, Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
21
|
Li J, Shen H, Xie H, Ying Y, Jin K, Yan H, Wang S, Xu M, Wang X, Xu X, Xie L. Dysregulation of ncRNAs located at the DLK1‑DIO3 imprinted domain: involvement in urological cancers. Cancer Manag Res 2019; 11:777-787. [PMID: 30697070 PMCID: PMC6339654 DOI: 10.2147/cmar.s190764] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Genomic imprinting has been found to be involved in human physical development and several diseases. The DLK1-DIO3 imprinted domain is located on human chromosome 14 and contains paternally expressed protein-coding genes (DLK1, RTL1, DIO3) and numerous maternally expressed ncRNA genes (MEG3, MEG8, antisense RTL1, miRNAs, piRNAs, and snoRNAs). Emerging evidence has implicated that dysregulation of the DLK1-DIO3 imprinted domain especially the imprinted ncRNAs is critical for tumor progressions. Multiple miRNAs and lncRNAs have been investigated in urological cancers, of which several are transcribed from this domain. In this review, we present current data about the associated miRNAs, lncRNAs, and piRNAs and the regulation of differentially methylated regions methylation status in the progression of urological cancers and preliminarily propose certain concepts about the potential regulatory networks involved in DLK1-DIO3 imprinted domain.
Collapse
Affiliation(s)
- Jiangfeng Li
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| | - Haixiang Shen
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| | - Haiyun Xie
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| | - Yufan Ying
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| | - Ke Jin
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| | - Huaqing Yan
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| | - Song Wang
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| | - Mingjie Xu
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| | - Xiao Wang
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| | - Xin Xu
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| | - Liping Xie
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China, ;
| |
Collapse
|
22
|
Goovaerts T, Steyaert S, Vandenbussche CA, Galle J, Thas O, Van Criekinge W, De Meyer T. A comprehensive overview of genomic imprinting in breast and its deregulation in cancer. Nat Commun 2018; 9:4120. [PMID: 30297886 PMCID: PMC6175939 DOI: 10.1038/s41467-018-06566-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 09/13/2018] [Indexed: 12/21/2022] Open
Abstract
Genomic imprinting plays an important role in growth and development. Loss of imprinting (LOI) has been found in cancer, yet systematic studies are impeded by data-analytical challenges. We developed a methodology to detect monoallelically expressed loci without requiring genotyping data, and applied it on The Cancer Genome Atlas (TCGA, discovery) and Genotype-Tissue expression project (GTEx, validation) breast tissue RNA-seq data. Here, we report the identification of 30 putatively imprinted genes in breast. In breast cancer (TCGA), HM13 is featured by LOI and expression upregulation, which is linked to DNA demethylation. Other imprinted genes typically demonstrate lower expression in cancer, often associated with copy number variation and aberrant DNA methylation. Downregulation in cancer frequently leads to higher relative expression of the (imperfectly) silenced allele, yet this is not considered canonical LOI given the lack of (absolute) re-expression. In summary, our novel methodology highlights the massive deregulation of imprinting in breast cancer.
Collapse
Affiliation(s)
- Tine Goovaerts
- Department Data Analysis and Mathematical Modelling, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Sandra Steyaert
- Department Data Analysis and Mathematical Modelling, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Chari A Vandenbussche
- Department Data Analysis and Mathematical Modelling, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Jeroen Galle
- Department Data Analysis and Mathematical Modelling, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Olivier Thas
- Department Data Analysis and Mathematical Modelling, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
- Bioinformatics Institute Ghent - from Nucleotides to Networks (BIG N2N), Ghent University, Technologiepark 927, 9052, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Wim Van Criekinge
- Department Data Analysis and Mathematical Modelling, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
- Bioinformatics Institute Ghent - from Nucleotides to Networks (BIG N2N), Ghent University, Technologiepark 927, 9052, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium
| | - Tim De Meyer
- Department Data Analysis and Mathematical Modelling, Ghent University, Coupure Links 653, 9000, Ghent, Belgium.
- Bioinformatics Institute Ghent - from Nucleotides to Networks (BIG N2N), Ghent University, Technologiepark 927, 9052, Ghent, Belgium.
- Cancer Research Institute Ghent (CRIG), Ghent University, Corneel Heymanslaan 10, 9000, Ghent, Belgium.
| |
Collapse
|
23
|
Sui F, Ji M, Hou P. Long non-coding RNAs in thyroid cancer: Biological functions and clinical significance. Mol Cell Endocrinol 2018; 469:11-22. [PMID: 28751134 DOI: 10.1016/j.mce.2017.07.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 01/07/2023]
Abstract
Thyroid cancer is the most common endocrine malignant tumor with rapidly increasing incidence in recent decades. Although the majority of thyroid cancers are relatively indolent, some cases still have a risk of developing into more aggressive and lethal forms of thyroid cancers. Similar to other malignancies, thyroid tumorigenesis is a multistep process involving the accumulation of a large number of genetic and epigenetic alterations. Thus, determination of the mechanisms of tumorigenesis is an urgent need for thyroid cancer treatment. Long noncoding RNAs (LncRNAs) have recently been demonstrated to participate in cancer progression. However, their role and molecular mechanism in thyroid cancer remain largely unclear. In this review, we focus on the dysregulation of lncRNAs in thyroid cancer, summarize the latest findings regarding the functions and mechanism of lncRNAs in thyroid cancer, and discuss their potential clinical significance in diagnosis and prognosis of thyroid cancer.
Collapse
Affiliation(s)
- Fang Sui
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Meiju Ji
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China
| | - Peng Hou
- Department of Endocrinology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China; Key Laboratory for Tumor Precision Medicine of Shanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, PR China.
| |
Collapse
|
24
|
Hu YH, Chen Q, Lu YX, Zhang JM, Lin C, Zhang F, Zhang WJ, Li XM, Zhang W, Li XN. Hypermethylation of NDN promotes cell proliferation by activating the Wnt signaling pathway in colorectal cancer. Oncotarget 2018; 8:46191-46203. [PMID: 28521288 PMCID: PMC5542259 DOI: 10.18632/oncotarget.17580] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 04/07/2017] [Indexed: 12/18/2022] Open
Abstract
The progression of CRC is a multistep process involving several genetic changes or epigenetic modifications. NDN is a member of the MAGE family, encoding a protein that generally suppresses cell proliferation and acting as a transcriptional repressor. Immunohistochemical staining revealed that the expression of NDN was significantly down-regulated in CRC tissues compared with normal tissues and the down-regulation of NDN in CRC could reflect the hypermethylation of the NDN promoter. Treatment of the CRC cell line SW480 with the demethylating agent 5-Aza-CdR restored the NDN expression level. The down-regulation of NDN was closely related to poor differentiation, advanced TNM stage and poor prognosis of CRC. The inhibition of NDN promoted CRC cell proliferation by enriching cells in the S phase. Furthermore, we observed that NDN binds to the GN box in the promoter of LRP6 to attenuate LRP6 transcription and inhibit the Wnt signaling pathway in CRC. In conclusion, our study revealed that the hypermethylation of NDN promotes cell proliferation by activating the Wnt signaling pathway through directly increasing the transcription of LRP6 in CRC. These findings might provide a new theoretical basis for the pathogenesis of CRC and facilitate the development of new therapeutic strategies against CRC.
Collapse
Affiliation(s)
- Yu-Han Hu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Qing Chen
- Department of Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yan-Xia Lu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jian-Ming Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chun Lin
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Fan Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wen-Juan Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiao-Min Li
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wei Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xue-Nong Li
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
25
|
Nasrollahzadeh-Khakiani M, Emadi-Baygi M, Schulz WA, Nikpour P. Long noncoding RNAs in gastric cancer carcinogenesis and metastasis. Brief Funct Genomics 2018; 16:129-145. [PMID: 27122631 DOI: 10.1093/bfgp/elw011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Recent studies of the human transcriptome, most prominently by the ENCyclopedia Of DNA Elements project, have revealed an unexpected number of noncoding RNAs (ncRNAs). Long noncoding RNAs (lncRNAs) are typically referred to a heterogeneous group of polyadenylated long ncRNAs, with a length of > 200 nt. LncRNAs constitute an integral part of tumor biology, with many lncRNAs discovered to be aberrantly expressed in various cancer types. They are involved in many aspects of cancer pathogenesis from its initiation to progression, metastasis and treatment response. Gastric cancer (GC) is the third leading cause of cancer death worldwide. Despite the current improvements of life expectancy and survival rate, most of the patients are diagnosed when their cancer has been progressed to advanced stages. Therefore, unraveling the molecular mechanisms of GC to find early-stage biomarkers is urgent. As the list of lncRNAs with deregulated expression in GC is steadily expanding, these molecules offer a source for developing GC-specific biomarkers. In this review, we will present and discuss those lncRNAs whose expression has been shown to be deregulated in GC.
Collapse
|
26
|
Küffer S, Gutting T, Belharazem D, Sauer C, Michel MS, Marx A, Trojan L, Ströbel P. Insulin-like growth factor 2 expression in prostate cancer is regulated by promoter-specific methylation. Mol Oncol 2018; 12:256-266. [PMID: 29239100 PMCID: PMC5792735 DOI: 10.1002/1878-0261.12164] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/16/2017] [Accepted: 11/25/2017] [Indexed: 12/12/2022] Open
Abstract
Deregulation of the insulin-like growth factor (IGF) axis and dysbalance of components of the IGF system as potential therapeutic targets have been described in different tumor types. IGF2 is a major embryonic growth factor and an important activator of IGF signaling. It is regulated by imprinting in a development- and tissue-dependent manner and has been implicated in a broad range of malignancies including prostate cancer (PCa). Loss of imprinting (LOI) usually results in bi-allelic gene expression and increased levels of IGF2. However, the regulatory mechanisms and the pathophysiological impact of altered IGF2 expression in PCa remain elusive. Here, we show that in contrast to many other tumors, IGF2 mRNA and protein levels were decreased in 80% of PCa in comparison with non-neoplastic adjacent prostate and were independent of LOI status. Instead, IGF2 expression in both tumors and adjacent prostate depended on preferential usage of the IGF2 promoters P3 and P4. Decreased IGF2 expression in tumors was strongly related to hypermethylation of these two promoters. Methylation of the A region in promoter P4 correlated specifically with IGF2 expression in the 20% of PCa where IGF2 was higher in tumors than in adjacent prostate. We conclude that IGF2 is downregulated in most PCa and may be particularly relevant during early stages of tumor development or during chemotherapy and androgen deprivation. PCa differs from other tumors in that IGF2 expression is mainly regulated through methylation of promoter-specific and not by imprinting. Targeting of promoter-specific regions may have relevance for the adjuvant treatment of PCa.
Collapse
Affiliation(s)
- Stefan Küffer
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Germany
| | - Tobias Gutting
- Institute of Pathology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Medicine II, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Djeda Belharazem
- Institute of Pathology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Christian Sauer
- Institute of Pathology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Maurice S Michel
- Department of Urology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Alexander Marx
- Institute of Pathology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| | - Lutz Trojan
- Department of Urology, University Medical Center Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Germany
| |
Collapse
|
27
|
Shi Y, Lv C, Shi L, Tu G. MEG3 inhibits proliferation and invasion and promotes apoptosis of human osteosarcoma cells. Oncol Lett 2017; 15:1917-1923. [PMID: 29434890 DOI: 10.3892/ol.2017.7463] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 11/07/2017] [Indexed: 01/01/2023] Open
Abstract
Osteosarcoma is known as a malignant tumour with a high mortality rate in orthopaedic settings; however, the factors associated with its degree of malignancy and the biological response remains to be elucidated. Although the essential role of the long noncoding RNA (lncRNA) maternally expressed 3 (MEG3) has been recently reported, its biological functions and regulatory mechanism in osteosarcoma cells have not yet been reported. In the present study, reverse transcription-quantitative polymerase chain reaction analysis revealed that the expression of MEG3 in MG63 cells was lower compared with in hFOB1.19 cells. Furthermore, it was observed that overexpressing MEG3 in MG63 cells resulted in a decline in the proliferation and invasion, and a marked increase in apoptosis. Additionally, western blotting was used to detect the changes in expression of p53 and MDM2 proto-oncogene, which may be regulated by MEG3, and proteins that associated with cell proliferation, invasion and apoptosis. It was demonstrated that the upregulation of MEG3 significantly increased the transactivation of p53 and induced downstream changes in protein expression. In conclusion, these experiments have demonstrated that MEG3 serves an essential regulatory role in the biological processes of human osteosarcoma cells, and imply that MEG3 may be a marker for predicting the occurrence and development of osteosarcoma.
Collapse
Affiliation(s)
- Yao Shi
- Department of Orthopaedic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Chen Lv
- Department of Orthopaedic Surgery, The First Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Li Shi
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, P.R. China
| | - Guanjun Tu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
28
|
He Y, Luo Y, Liang B, Ye L, Lu G, He W. Potential applications of MEG3 in cancer diagnosis and prognosis. Oncotarget 2017; 8:73282-73295. [PMID: 29069869 PMCID: PMC5641212 DOI: 10.18632/oncotarget.19931] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/25/2017] [Indexed: 12/25/2022] Open
Abstract
LncRNAs are emerging as integral functional and regulatory components of normal biological activities and are now considered as critically involved in the development of different diseases including cancer. In this review, we summarized recent findings on maternally expressed gene 3 (MEG3), a noncoding lncRNA, locates in the imprinted DLK1–MEG3 locus on human chromosome 14q32.3 region. MEG3 is expressed in normal tissues but is either lost or decreased in many human tumors and tumor derived cell lines. Studies have demonstrated that MEG3 is associated with cancer initiation, progression, metastasis and chemo-resistance. MEG3 may affect the activities of TP53, MDM2, GDF15, RB1 and some other key cell cycle regulators. In addition, the level of MEG3 showed good correlation with cancer clinicopathological grade. In summary, MEGs is an RNA-based tumor suppressor and is involved in the etiology, progression, and chemosensitivity of cancers. The alteration of MEG3 levels in various cancers suggested the possibility of using MEG3 level for cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Yuqing He
- Institute of Medical Systems Biology, Guangdong Medical University, Dongguan 523808, China.,Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Guangdong Medical University, Dongguan 523808, China
| | - Yanhong Luo
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Biyu Liang
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Lei Ye
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Guangxing Lu
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| | - Weiming He
- Department of Epidemiology and Medical Statistics, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
29
|
Zhang L, Liang X, Li Y. Long non-coding RNA MEG3 inhibits cell growth of gliomas by targeting miR-93 and inactivating PI3K/AKT pathway. Oncol Rep 2017; 38:2408-2416. [PMID: 28791407 DOI: 10.3892/or.2017.5871] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 07/13/2017] [Indexed: 11/06/2022] Open
Abstract
Gliomas are the most common cancers in the brain, accompanied with high morbility, occurrence, disability and mortality. Long non-coding RNAs (lncRNAs) have been proposed as promoter or inhibitor in many cancer processes. Previous findings have indicated that lncRNA-maternally expressed gene 3 (MEG3) is involved in tumorigenesis of several cancers, including glioma. However, the underlying mechanism of MEG3 in glioma remains elusive. In our study, MEG3 was found downregulated in glioma tissues compared with normal brain tissues. Downregulated expression of MEG3 was also detected in two human glioma cell lines (U-251, M059J) compared with normal astrocyte cells. MEG3 was then overexpressed by ligating to a lentiviral vector. Overexpressed MEG3 inhibited the proliferation of U-251 cells, and restrained the expression of proliferation marker proteins Ki67 and proliferating cell nuclear antigen (PCNA). However, cell apoptosis rate of U-251 cells and the expression of apoptosis marker proteins (caspase-3 and caspase-9) were elevated by MEG3. Furthermore, miR-93 was predicted a direct target of lncRNA-MEG3 by bioinformatics analysis. Overexpressed MEG3 counteracted the role of miR-93 in facilitating proliferation and inhibiting apoptosis in U-251 cells. Moreover, MEG3 restained the activation of phosphatidylinositol 3 kinase/protein kinase B (PI3K/AKT) pathway by reducing cytomembrane translocation of AKT. Finally, the in vivo experiment revealed that MEG3 strongly reduced tumor growth, tumor volume and the expression of Ki67 and PCNA. lncRNA-MEG3 also inhibited the level of miR-93 and the expression of PI3K/AKT pathway related proteins in vivo. Taken together, our research indicated a MEG3-miR-93-PI3K-AKT pathway in regulating the growth of glioma, providing a promising therapy for glioma.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Neurosurgery, Yulin City Hospital of Traditional Chinese Medicine, Yulin, Shaanxi 719000, P.R. China
| | - Xin Liang
- Department of Neurosurgery, Yulin City Hospital of Traditional Chinese Medicine, Yulin, Shaanxi 719000, P.R. China
| | - Yuxiong Li
- Department of Neurosurgery, Yulin City Hospital of Traditional Chinese Medicine, Yulin, Shaanxi 719000, P.R. China
| |
Collapse
|
30
|
Fan FY, Deng R, Yi H, Sun HP, Zeng Y, He GC, Su Y. The inhibitory effect of MEG3/miR-214/AIFM2 axis on the growth of T-cell lymphoblastic lymphoma. Int J Oncol 2017; 51:316-326. [PMID: 28534937 DOI: 10.3892/ijo.2017.4006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 04/21/2017] [Indexed: 11/06/2022] Open
Abstract
T-cell lymphoblastic lymphoma (T-LBL) is an aggressive malignancy with poor prognosis and high recurrence rate. Long non-coding RNA (lncRNA)-MEG3 is an important tumor suppressor in various cancers. The present study investigated the potential role of maternally expressed gene 3 (MEG3) in the progression of T-LBL. Suppressed expression of MEG3 was detected in T-LBL tissues compared with adjacent histologically normal tissues. Down-regulated level of MEG3 was also found in three T-LBL cell lines (CCRF-CEM, Jurkat and SUP-T1) compared with human T-cell line H9. The proliferation of T-LBL cells was inhibited and cell apoptosis rate was largely promoted when MEG3 was upregulated by a lentiviral vector. Further research revealed that microRNA (miRNA)-214 is a direct target of MEG3. The expression of miR-214 was increased in T-LBL tissues and cell lines compared with control groups. Besides, decreased level of miR-214 was elevated adding miR-214 mimic in SUP-T1 cells transfected with LncRNA-MEG3. Similarly, upregulated level of miR-214 was downregulated adding miR-214 inhibitor in SUP-T1 cells transfected with MEG3 siRNA. Luciferase activity assay further confirmed the targeting relationship between MEG3 and miR-214. Moreover, AIFM2 protein was predicted as a target of miR-214. The expression of AIFM2 was increased by MEG3 and was downregulated by miR-214 mimic. miRNA-214 reversed the effect of MEG3 on inhibiting cell proliferation and inducing cell apoptosis and cell cycle arrest in SUP-T1 cells. Moreover, relative expression of AIFM2 had a positive correlation with the expression of MEG3 and was negatively affected by miR-214. In vivo, MEG3 effectively suppressed tumor growth and the expression of proliferation markers Ki-67 and proliferating cell nuclear antigen (PCNA). Taken together, our research revealed that MEG3 worked as an anti-oncogene in T-LBL, and the MEG3-miR-214-AIFM2 pathway regulated the growth of T-LBL, providing potential prognosis markers as well as new potential targets for T-LBL treatment.
Collapse
Affiliation(s)
- Fang-Yi Fan
- Department of Hematology and Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Chengdu Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Rui Deng
- Department of Hematology and Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Chengdu Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Hai Yi
- Department of Hematology and Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Chengdu Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Hao-Ping Sun
- Department of Hematology and Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Chengdu Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Yan Zeng
- Department of Hematology and Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Chengdu Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Guang-Cui He
- Department of Hematology and Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Chengdu Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| | - Yi Su
- Department of Hematology and Hematopoietic Stem Cell Transplantation and Cell Immunotherapy Center, Chengdu Military General Hospital of PLA, Chengdu, Sichuan 610083, P.R. China
| |
Collapse
|
31
|
Huang ZL, Chen RP, Zhou XT, Zhan HL, Hu MM, Liu B, Wu GD, Wu LF. Long non-coding RNA MEG3 induces cell apoptosis in esophageal cancer through endoplasmic reticulum stress. Oncol Rep 2017; 37:3093-3099. [PMID: 28405686 DOI: 10.3892/or.2017.5568] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/27/2017] [Indexed: 02/05/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) play important roles in diverse biological processes, such as cell growth, apoptosis and migration. Although downregulation of lncRNA MEG3 has been identified in several cancers, little is known about its role in esophageal squamous cell carcinoma (ESCC). The aim of the present study was to detect MEG3 expression in clinical ESCC tissues, investigate its biological functions and the endoplasmic reticulum (ER) stress-relative mechanism. MEG3 expression levels were detected by qRT-PCR in both tumor tissues and adjacent non-tumor tissues from 28 ESCC patients. PcDNA3.1-MEG3 recombinant plasmids were constructed and transfected to EC109 cells. Cell growth was analyzed by CCK-8 assay. Cell apoptosis was analyzed by fluorescence microscope and Annexin V/PI assay. The protein expression was determined by western blot analysis. The results showed that MEG3 decreased significantly in ESCC tissues relative to adjacent normal tissues. PcDNA3.1-MEG3 plasmids were successfully constructed and the expression level of MEG3 significantly increased after MEG3 transfection to EC109 cells. Ectopic expression of MEG3 inhibited EC109 cell proliferation and induced apoptosis in vitro. MEG3 overexpression increased the expression of ER stress‑related proteins (GRP78, IRE1, PERK, ATF6, CHOP and cleaved‑caspase-3). Our results first demonstrate that MEG3 is downregulated in ESCC tissues. MEG3 was able to inhibit cell growth and induced apoptosis in EC109 cells, most probably via activation of the ER stress pathway.
Collapse
Affiliation(s)
- Zhen-Lun Huang
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Rui-Pei Chen
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Xiao-Tao Zhou
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Hao-Lian Zhan
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Min-Min Hu
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Bin Liu
- Department of Neurosurgery, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| | - Guan-Di Wu
- First Clinical Medical College, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Ling-Fei Wu
- Department of Gastroenterology, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
32
|
Abstract
Methodological advances that allow deeper characterization of non-coding elements in the genome have started to reveal the full spectrum of deregulation in cancer. We generated an inducible cell model to track transcriptional changes after induction of a well-known leukemia-inducing fusion gene, ETV6-RUNX1. Our data revealed widespread transcriptional alterations outside coding elements in the genome. This adds to the growing list of various alterations in the non-coding genome in cancer and pinpoints their role in diseased cellular state.
Collapse
Affiliation(s)
- Susanna Teppo
- a Tampere Center for Child Health Research, Faculty of Medicine and Life Sciences , University of Tampere and Tampere University Hospital , Tampere , Finland
| | - Merja Heinäniemi
- b Institute of Biomedicine, School of Medicine , University of Eastern Finland , Kuopio , Finland
| | - Olli Lohi
- a Tampere Center for Child Health Research, Faculty of Medicine and Life Sciences , University of Tampere and Tampere University Hospital , Tampere , Finland
| |
Collapse
|
33
|
Wang P, Mokhtari R, Pedrosa E, Kirschenbaum M, Bayrak C, Zheng D, Lachman HM. CRISPR/Cas9-mediated heterozygous knockout of the autism gene CHD8 and characterization of its transcriptional networks in cerebral organoids derived from iPS cells. Mol Autism 2017; 8:11. [PMID: 28321286 PMCID: PMC5357816 DOI: 10.1186/s13229-017-0124-1] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 02/15/2017] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND CHD8 (chromodomain helicase DNA-binding protein 8), which codes for a member of the CHD family of ATP-dependent chromatin-remodeling factors, is one of the most commonly mutated genes in autism spectrum disorders (ASD) identified in exome-sequencing studies. Loss of function mutations in the gene have also been found in schizophrenia (SZ) and intellectual disabilities and influence cancer cell proliferation. We previously reported an RNA-seq analysis carried out on neural progenitor cells (NPCs) and monolayer neurons derived from induced pluripotent stem (iPS) cells that were heterozygous for CHD8 knockout (KO) alleles generated using CRISPR-Cas9 gene editing. A significant number of ASD and SZ candidate genes were among those that were differentially expressed in a comparison of heterozygous KO lines (CHD8+/-) vs isogenic controls (CHD8+/-), including the SZ and bipolar disorder (BD) candidate gene TCF4, which was markedly upregulated in CHD8+/- neuronal cells. METHODS In the current study, RNA-seq was carried out on CHD8+/- and isogenic control (CHD8+/+) cerebral organoids, which are 3-dimensional structures derived from iPS cells that model the developing human telencephalon. RESULTS TCF4 expression was, again, significantly upregulated. Pathway analysis carried out on differentially expressed genes (DEGs) revealed an enrichment of genes involved in neurogenesis, neuronal differentiation, forebrain development, Wnt/β-catenin signaling, and axonal guidance, similar to our previous study on NPCs and monolayer neurons. There was also significant overlap in our CHD8+/- DEGs with those found in a transcriptome analysis carried out by another group using cerebral organoids derived from a family with idiopathic ASD. Remarkably, the top DEG in our respective studies was the non-coding RNA DLX6-AS1, which was markedly upregulated in both studies; DLX6-AS1 regulates the expression of members of the DLX (distal-less homeobox) gene family. DLX1 was also upregulated in both studies. DLX genes code for transcription factors that play a key role in GABAergic interneuron differentiation. Significant overlap was also found in a transcriptome study carried out by another group using iPS cell-derived neurons from patients with BD, a condition characterized by dysregulated WNT/β-catenin signaling in a subgroup of affected individuals. CONCLUSIONS Overall, the findings show that distinct ASD, SZ, and BD candidate genes converge on common molecular targets-an important consideration for developing novel therapeutics in genetically heterogeneous complex traits.
Collapse
Affiliation(s)
- Ping Wang
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
| | - Ryan Mokhtari
- Department of Psychiatry and Behavioral Sciences, Erciyes University School of Medicine, Kayseri, Turkey
| | - Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Erciyes University School of Medicine, Kayseri, Turkey
| | - Michael Kirschenbaum
- Department of Psychiatry and Behavioral Sciences, Erciyes University School of Medicine, Kayseri, Turkey
| | - Can Bayrak
- Erciyes University School of Medicine, Kayseri, Turkey
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
- Department of Neurology, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
| | - Herbert M. Lachman
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
- Department of Psychiatry and Behavioral Sciences, Erciyes University School of Medicine, Kayseri, Turkey
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY USA
| |
Collapse
|
34
|
NDN is an imprinted tumor suppressor gene that is downregulated in ovarian cancers through genetic and epigenetic mechanisms. Oncotarget 2016; 7:3018-32. [PMID: 26689988 PMCID: PMC4823087 DOI: 10.18632/oncotarget.6576] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 11/21/2015] [Indexed: 12/18/2022] Open
Abstract
NDN is a maternally imprinted gene consistently expressed in normal ovarian epithelium, is dramatically downregulated in the majority of ovarian cancers. Little or no NDN expression could be detected in 73% of 351 epithelial ovarian cancers. NDN was also downregulated in 10 ovarian cancer cell lines with total loss in 6 of 10. Re-expression of NDN decreased Bcl-2 levels and induced apoptosis, which significantly inhibited ovarian cancer cell growth in cell culture and in xenografts. In addition, re-expression of NDN inhibited cell migration by decreasing actin stress fiber and focal adhesion complex formation through deactivation of Src, FAK and RhoA. Loss of NDN expression in ovarian cancers could be attributed to LOH in 28% of 18 informative cases and to hypermethylation of CpG sites 1 and 2 of NDN promoter in 23% and 30% of 43 ovarian cancers, respectively. Promoter hypermethylation was also found in 5 of 10 ovarian cancer cell lines. Treatment with the demethylating agent 5-aza-2′-deoxycytidine restored NDN expression in 4 of 7 cell lines with enhanced promoter methylation levels. These observations support the conclusion that NDN is an imprinted tumor suppressor gene which affects cancer cell motility, invasion and growth and that its loss of function in ovarian cancer can be caused by both genetic and epigenetic mechanisms.
Collapse
|
35
|
Chen J, Miao Z, Xue B, Shan Y, Weng G, Shen B. Long Non-coding RNAs in Urologic Malignancies: Functional Roles and Clinical Translation. J Cancer 2016; 7:1842-1855. [PMID: 27698924 PMCID: PMC5039368 DOI: 10.7150/jca.15876] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/29/2016] [Indexed: 12/31/2022] Open
Abstract
Early diagnosis and surveillance for metastasis and recurrences are critical issues of urologic cancer. Deregulation of long non-coding RNAs (lncRNAs) has been implicated in urologic malignancies and represents potential markers or therapeutic targets. However, the utility of lncRNA as biomarkers appears to be overstated due to heterogeneous or irreproducible results from different studies. Thus, a critical and cautious review on the biomarker potential of lncRNAs is needed. This review provides an update on new findings of lncRNA-based markers for urologic cancer. The diverse mechanisms and associated examples of lncRNAs involved during the carcinogenesis of prostate cancer, bladder cancer and renal cancer were discussed in a more balanced and critical manner, as were the suitability of lncRNAs as diagnostic or prognostics markers.
Collapse
Affiliation(s)
- Jiajia Chen
- Center for Systems Biology, Soochow University, Suzhou 215006, China; School of Chemistry, Biological Engineering, Suzhou University of Science and Technology, Suzhou 215011, China
| | - Zhijun Miao
- Center for Systems Biology, Soochow University, Suzhou 215006, China; Suzhou Dushuhu Hospital, Clinic Center, Soochow University, Suzhou 215123, China
| | - Boxin Xue
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Yuxi Shan
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Guobin Weng
- Ningbo Urologic and Nephrotic Hospital, Ningbo 315000, China
| | - Bairong Shen
- Center for Systems Biology, Soochow University, Suzhou 215006, China
| |
Collapse
|
36
|
Vega-Benedetti AF, Saucedo C, Zavattari P, Vanni R, Zugaza JL, Parada LA. PLAGL1: an important player in diverse pathological processes. J Appl Genet 2016; 58:71-78. [PMID: 27311313 DOI: 10.1007/s13353-016-0355-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/02/2016] [Accepted: 06/02/2016] [Indexed: 12/23/2022]
Abstract
The PLAGL1 gene encodes a homonymous zinc finger protein that promotes cell cycle arrest and apoptosis through multiple pathways. The protein has been implicated in metabolic, genetic, and neoplastic illnesses, but the molecular mechanisms by which the protein PLAGL1 participates in such diverse processes remains to be elucidated. In this review, we focus mainly on the molecular biology of PLAGL1 and the relevance of its abnormalities to several pathological processes.
Collapse
Affiliation(s)
- Ana F Vega-Benedetti
- Institute of Experimental Pathology, UNSa-CONICET, Ave. Bolivia 5150, 4400, Salta, Argentina
| | - Cinthia Saucedo
- Institute of Experimental Pathology, UNSa-CONICET, Ave. Bolivia 5150, 4400, Salta, Argentina
| | - Patrizia Zavattari
- Biochemistry, Biology and Genetics Unit, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato SP 8, Km 0.700, 09042, Monserrato, Cagliari, Italy
| | - Roberta Vanni
- Biochemistry, Biology and Genetics Unit, Department of Biomedical Sciences, University of Cagliari, Cittadella Universitaria di Monserrato SP 8, Km 0.700, 09042, Monserrato, Cagliari, Italy
| | - José L Zugaza
- IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.,Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, Building 205, Zamudio, Spain.,Department of Genetics, Physic Anthropology and Animal Physiology, Faculty of Medicine and Dentistry, University of the Basque Country, Barrio Sarriena s/n, 48940, Leioa, Spain
| | - Luis Antonio Parada
- Institute of Experimental Pathology, UNSa-CONICET, Ave. Bolivia 5150, 4400, Salta, Argentina.
| |
Collapse
|
37
|
Li Y, Xie C, Murphy SK, Skaar D, Nye M, Vidal AC, Cecil KM, Dietrich KN, Puga A, Jirtle RL, Hoyo C. Lead Exposure during Early Human Development and DNA Methylation of Imprinted Gene Regulatory Elements in Adulthood. ENVIRONMENTAL HEALTH PERSPECTIVES 2016; 124:666-73. [PMID: 26115033 PMCID: PMC4858407 DOI: 10.1289/ehp.1408577] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 06/24/2015] [Indexed: 05/03/2023]
Abstract
BACKGROUND Lead exposure during early development causes neurodevelopmental disorders by unknown mechanisms. Epidemiologic studies have focused recently on determining associations between lead exposure and global DNA methylation; however, such approaches preclude the identification of loci that may alter human disease risk. OBJECTIVES The objective of this study was to determine whether maternal, postnatal, and early childhood lead exposure can alter the differentially methylated regions (DMRs) that control the monoallelic expression of imprinted genes involved in metabolism, growth, and development. METHODS Questionnaire data and serial blood lead levels were obtained from 105 participants (64 females, 41 males) of the Cincinnati Lead Study from birth to 78 months. When participants were adults, we used Sequenom EpiTYPER assays to test peripheral blood DNA to quantify CpG methylation in peripheral blood leukocytes at DMRs of 22 human imprinted genes. Statistical analyses were conducted using linear regression. RESULTS Mean blood lead concentration from birth to 78 months was associated with a significant decrease in PEG3 DMR methylation (β = -0.0014; 95% CI: -0.0023, -0.0005, p = 0.002), stronger in males (β = -0.0024; 95% CI: -0.0038, -0.0009, p = 0.003) than in females (β = -0.0009; 95% CI: -0.0020, 0.0003, p = 0.1). Elevated mean childhood blood lead concentration was also associated with a significant decrease in IGF2/H19 (β = -0.0013; 95% CI: -0.0023, -0.0003, p = 0.01) DMR methylation, but primarily in females, (β = -0.0017; 95% CI: -0.0029, -0.0006, p = 0.005) rather than in males, (β = -0.0004; 95% CI: -0.0023, 0.0015, p = 0.7). Elevated blood lead concentration during the neonatal period was associated with higher PLAGL1/HYMAI DMR methylation regardless of sex (β = 0.0075; 95% CI: 0.0018, 0.0132, p = 0.01). The magnitude of associations between cumulative lead exposure and CpG methylation remained unaltered from 30 to 78 months. CONCLUSIONS Our findings provide evidence that early childhood lead exposure results in sex-dependent and gene-specific DNA methylation differences in the DMRs of PEG3, IGF2/H19, and PLAGL1/HYMAI in adulthood. CITATION Li Y, Xie C, Murphy SK, Skaar D, Nye M, Vidal AC, Cecil KM, Dietrich KN, Puga A, Jirtle RL, Hoyo C. 2016. Lead exposure during early human development and DNA methylation of imprinted gene regulatory elements in adulthood. Environ Health Perspect 124:666-673; http://dx.doi.org/10.1289/ehp.1408577.
Collapse
Affiliation(s)
- Yue Li
- Department of Community and Family Medicine, and
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina, USA
| | - Changchun Xie
- Division of Epidemiology and Biostatistics, Department of Environmental Health, Center for Clinical and Translational Science and Training, University of Cincinnati (UC), Cincinnati, Ohio, USA
| | - Susan K. Murphy
- Department of Community and Family Medicine, and
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina, USA
| | - David Skaar
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University (NCSU), Raleigh, North Carolina, USA
| | - Monica Nye
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Adriana C. Vidal
- Department of Community and Family Medicine, and
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina, USA
| | - Kim M. Cecil
- Cincinnati Children’s Environmental Health Center, Cincinnati Children’s Hospital Medical Center, UC College of Medicine, Cincinnati, Ohio, USA
- Department of Radiology,
- Department of Pediatrics,
- Department of Environmental Health,
- Center for Environmental Genetics, and
| | - Kim N. Dietrich
- Division of Epidemiology and Biostatistics, UC College of Medicine, Cincinnati, Ohio, USA
| | - Alvaro Puga
- Department of Environmental Health,
- Center for Environmental Genetics, and
| | - Randy L. Jirtle
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University (NCSU), Raleigh, North Carolina, USA
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Sport and Exercise Sciences, Institute of Sport and Physical Activity Research, University of Bedfordshire, Bedford, Bedfordshire, United Kingdom
- Address correspondence to C. Hoyo, Department of Biological Sciences, Center for Human Health and the Environment (CHHE), Program of Epidemiology and Environmental Epigenomics, North Carolina State University (NCSU), Raleigh, NC 27695-7633 USA. Telephone: (919) 515-0540. E-mail: , or R.L. Jirtle, Department of Biological Sciences, CHHE, NCSU, Raleigh, NC 27695-7633 USA. Telephone: (919) 399-3342. E-mail:
| | - Cathrine Hoyo
- Department of Community and Family Medicine, and
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, North Carolina, USA
- Department of Biological Sciences, Center for Human Health and the Environment, North Carolina State University (NCSU), Raleigh, North Carolina, USA
- Address correspondence to C. Hoyo, Department of Biological Sciences, Center for Human Health and the Environment (CHHE), Program of Epidemiology and Environmental Epigenomics, North Carolina State University (NCSU), Raleigh, NC 27695-7633 USA. Telephone: (919) 515-0540. E-mail: , or R.L. Jirtle, Department of Biological Sciences, CHHE, NCSU, Raleigh, NC 27695-7633 USA. Telephone: (919) 399-3342. E-mail:
| |
Collapse
|
38
|
Kikutake C, Yahara K. Identification of Epigenetic Biomarkers of Lung Adenocarcinoma through Multi-Omics Data Analysis. PLoS One 2016; 11:e0152918. [PMID: 27042856 PMCID: PMC4820141 DOI: 10.1371/journal.pone.0152918] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 03/21/2016] [Indexed: 12/28/2022] Open
Abstract
Epigenetic mechanisms such as DNA methylation or histone modifications are essential for the regulation of gene expression and development of tissues. Alteration of epigenetic modifications can be used as an epigenetic biomarker for diagnosis and as promising targets for epigenetic therapy. A recent study explored cancer-cell specific epigenetic biomarkers by examining different types of epigenetic modifications simultaneously. However, it was based on microarrays and reported biomarkers that were also present in normal cells at a low frequency. Here, we first analyzed multi-omics data (including ChIP-Seq data of six types of histone modifications: H3K27ac, H3K4me1, H3K9me3, H3K36me3, H3K27me3, and H3K4me3) obtained from 26 lung adenocarcinoma cell lines and a normal cell line. We identified six genes with both H3K27ac and H3K4me3 histone modifications in their promoter regions, which were not present in the normal cell line, but present in ≥85% (22 out of 26) and ≤96% (25 out of 26) of the lung adenocarcinoma cell lines. Of these genes, NUP210 (encoding a main component of the nuclear pore complex) was the only gene in which the two modifications were not detected in another normal cell line. RNA-Seq analysis revealed that NUP210 was aberrantly overexpressed among the 26 lung adenocarcinoma cell lines, although the frequency of NUP210 overexpression was lower (19.3%) in 57 lung adenocarcinoma tissue samples studied and stored in another database. This study provides a basis to discover epigenetic biomarkers highly specific to a certain cancer, based on multi-omics data at the cell population level.
Collapse
Affiliation(s)
- Chie Kikutake
- Division of Biostatistics, Kurume University School of Medicine, Fukuoka, Japan
| | - Koji Yahara
- Biostatistics Center, Kurume University, Kurume, Fukuoka, Japan
- Institute of Life Science, College of Medicine, Swansea University, Swansea, United Kingdom
- * E-mail:
| |
Collapse
|
39
|
Zhang A, Zhang J, Kaipainen A, Lucas JM, Yang H. Long non-coding RNA: A newly deciphered "code" in prostate cancer. Cancer Lett 2016; 375:323-330. [PMID: 26965999 DOI: 10.1016/j.canlet.2016.03.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 01/03/2023]
Abstract
As one of the most frequently diagnosed cancers in males, the development and progression of prostate cancer remains an open area of research. The role of lncRNAs in prostate cancer is an emerging field of study. In this review, we summarize what is currently known about lncRNAs in prostate cancer while focusing on a few key lncRNAs. PCA3 was the first lncRNA identified in prostate cancer and has been shown to be expressed in a majority of prostate cancer cases. It may act in both an androgen dependent and independent fashion and has clinical utility as a biomarker. Other lncRNAs are known to interact directly with the androgen receptor pathway including PlncRNA-1, HOTAIR, PRNCR1 and PCGEM1. Additionally, lncRNAs have been shown to interfere with tumor suppressors, DNA break repair, transcription and alternate RNA splicing. While only in its infancy, an understanding of the role of lncRNAs in prostate cancer development should present ample opportunities for the discovery of new cancer biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Ailin Zhang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jiawei Zhang
- School of Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Arja Kaipainen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jared M Lucas
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Hong Yang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
40
|
Guan GF, Zhang DJ, Wen LJ, Xin D, Liu Y, Yu DJ, Su K, Zhu L, Guo YY, Wang K. Overexpression of lncRNA H19/miR-675 promotes tumorigenesis in head and neck squamous cell carcinoma. Int J Med Sci 2016; 13:914-922. [PMID: 27994496 PMCID: PMC5165684 DOI: 10.7150/ijms.16571] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 10/07/2016] [Indexed: 02/07/2023] Open
Abstract
There is accumulating evidence indicating that long non-coding RNA H19 and its mature product miR-675 play essential roles for tumor growth and progression. However, their prognostic value in human head and neck squamous cell carcinoma (HNSCC), particular in laryngeal carcinoma, remains to be elucidated. In this study, we observed that both H19 and miR-675 were significantly overexpressed in a cohort of 65 primary tumor samples and two HNSCC cell lines. Importantly, when paired with patient follow-up data, higher expression of either H19 or miR-675 was significantly correlated with higher risk of patient relapse, and associated with worse overall survival and poor disease-free survival. Knockdown miR-675 caused significant reduction of cell viability, migratory and invasive capabilities. Taken together, these results suggest that the strong correlation of H19 overexpression together with higher miR-675 and lymph node metastases could be useful predictive markers, indicating a potentially therapeutic strategy for HNSCC patients.
Collapse
Affiliation(s)
- Guo-Fang Guan
- Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - De-Jun Zhang
- Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Lian-Ji Wen
- Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Ding Xin
- Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Yan Liu
- Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Duo-Jiao Yu
- Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Kai Su
- Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Lin Zhu
- Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Ying-Yuan Guo
- Department of Otolaryngology, Head and Neck Surgery, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| | - Ke Wang
- Department of Respiratory Medicine, The Second Hospital of Jilin University, Changchun 130041, P. R. China
| |
Collapse
|
41
|
Silva-Ortiz AV, Bratoeff E, Ramírez-Apan T, Heuze Y, Sánchez A, Soriano J, Cabeza M. Synthesis and activity of novel 16-dehydropregnenolone acetate derivatives as inhibitors of type 1 5α-reductase and on cancer cell line SK-LU-1. Bioorg Med Chem 2015; 23:7535-42. [DOI: 10.1016/j.bmc.2015.10.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/30/2015] [Accepted: 10/31/2015] [Indexed: 10/22/2022]
|
42
|
Weiss M, Plass C, Gerhauser C. Role of lncRNAs in prostate cancer development and progression. Biol Chem 2015; 395:1275-90. [PMID: 25153594 DOI: 10.1515/hsz-2014-0201] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 08/12/2014] [Indexed: 12/12/2022]
Abstract
Prostate cancer (PCa) is the second most common cause of cancer-related deaths in men. Despite advances in the characterization of genomic and epigenetic aberrations contributing to PCa, the etiology of PCa is still far from being understood. Research over the past decade demonstrated the role of long non-coding RNAs (lncRNAs) in deregulation of target genes mainly through epigenetic mechanisms. In PCa, evidence accumulated that hundreds of lncRNAs are dysregulated. Functional analyses revealed their contribution to prostate carcinogenesis by targeting relevant pathways and gene regulation mechanisms including PTEN/AKT and androgen receptor signaling as well as chromatin remodeling complexes. Here we summarize our current knowledge on the roles of lncRNAs in PCa and their potential use as biomarkers for aggressive PCa and as novel therapeutic targets.
Collapse
|
43
|
Ruhrmann S, Stridh P, Kular L, Jagodic M. Genomic imprinting: A missing piece of the Multiple Sclerosis puzzle? Int J Biochem Cell Biol 2015; 67:49-57. [PMID: 26002250 DOI: 10.1016/j.biocel.2015.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/10/2015] [Accepted: 05/11/2015] [Indexed: 12/14/2022]
Abstract
Evidence for parent-of-origin effects in complex diseases such as Multiple Sclerosis (MS) strongly suggests a role for epigenetic mechanisms in their pathogenesis. In this review, we describe the importance of accounting for parent-of-origin when identifying new risk variants for complex diseases and discuss how genomic imprinting, one of the best-characterized epigenetic mechanisms causing parent-of-origin effects, may impact etiology of complex diseases. While the role of imprinted genes in growth and development is well established, the contribution and molecular mechanisms underlying the impact of genomic imprinting in immune functions and inflammatory diseases are still largely unknown. Here we discuss emerging roles of imprinted genes in the regulation of inflammatory responses with a particular focus on the Dlk1 cluster that has been implicated in etiology of experimental MS-like disease and Type 1 Diabetes. Moreover, we speculate on the potential wider impact of imprinting via the action of imprinted microRNAs, which are abundantly present in the Dlk1 locus and predicted to fine-tune important immune functions. Finally, we reflect on how unrelated imprinted genes or imprinted genes together with non-imprinted genes can interact in so-called imprinted gene networks (IGN) and suggest that IGNs could partly explain observed parent-of-origin effects in complex diseases. Unveiling the mechanisms of parent-of-origin effects is therefore likely to teach us not only about the etiology of complex diseases but also about the unknown roles of this fascinating phenomenon underlying uneven genetic contribution from our parents. This article is part of a Directed Issue entitled: Epigenetics dynamics in development and disease.
Collapse
Affiliation(s)
- Sabrina Ruhrmann
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Pernilla Stridh
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lara Kular
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
44
|
Devaney JM, Wang S, Furbert-Harris P, Apprey V, Ittmann M, Wang BD, Olender J, Lee NH, Kwabi-Addo B. Genome-wide differentially methylated genes in prostate cancer tissues from African-American and Caucasian men. Epigenetics 2015; 10:319-28. [PMID: 25864488 DOI: 10.1080/15592294.2015.1022019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Increasing evidence suggests that aberrant DNA methylation changes may contribute to prostate cancer (PCa) ethnic disparity. To comprehensively identify DNA methylation alterations in PCa disparity, we used the Illumina 450K methylation platform to interrogate the methylation status of 485,577 CpG sites focusing on gene-associated regions of the human genome. Genomic DNA from African-American (AA; 7 normal and 3 cancers) and Caucasian (Cau; 8 normal and 3 cancers) was used in the analysis. Hierarchical clustering analysis identified probe-sets unique to AA and Cau samples, as well as common to both. We selected 25 promoter-associated novel CpG sites most differentially methylated by race (fold change > 1.5-fold; adjusted P < 0.05) and compared the β-value of these sites provided by the Illumina, Inc. array with quantitative methylation obtained by pyrosequencing in 7 prostate cell lines. We found very good concordance of the methylation levels between β-value and pyrosequencing. Gene expression analysis using qRT-PCR in a subset of 8 genes after treatment with 5-aza-2'-deoxycytidine and/or trichostatin showed up-regulation of gene expression in PCa cells. Quantitative analysis of 4 genes, SNRPN, SHANK2, MST1R, and ABCG5, in matched normal and PCa tissues derived from AA and Cau PCa patients demonstrated differential promoter methylation and concomitant differences in mRNA expression in prostate tissues from AA vs. Cau. Regression analysis in normal and PCa tissues as a function of race showed significantly higher methylation prevalence for SNRPN (P = 0.012), MST1R (P = 0.038), and ABCG5 (P < 0.0002) for AA vs. Cau samples. We selected the ABCG5 and SNRPN genes and verified their biological functions by Western blot analysis and siRNA gene knockout effects on cell proliferation and invasion in 4 PCa cell lines (2 AA and 2 Cau patients-derived lines). Knockdown of either ABCG5 or SNRPN resulted in a significant decrease in both invasion and proliferation in Cau PCa cell lines but we did not observe these remarkable loss-of-function effects in AA PCa cell lines. Our study demonstrates how differential genome-wide DNA methylation levels influence gene expression and biological functions in AA and Cau PCa.
Collapse
Affiliation(s)
- J M Devaney
- a Children's National Medical Center ; Center for Genetic Medicine Research ; Washington, DC USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Song S, Ghosh J, Mainigi M, Turan N, Weinerman R, Truongcao M, Coutifaris C, Sapienza C. DNA methylation differences between in vitro- and in vivo-conceived children are associated with ART procedures rather than infertility. Clin Epigenetics 2015; 7:41. [PMID: 25901188 PMCID: PMC4404660 DOI: 10.1186/s13148-015-0071-7] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/10/2015] [Indexed: 12/29/2022] Open
Abstract
Background We, and others, have demonstrated previously that there are differences in DNA methylation and transcript levels of a number of genes in cord blood and placenta between children conceived using assisted reproductive technologies (ART) and children conceived in vivo. The source of these differences (the effect of ART versus the underlying infertility) has never been determined in humans. In this study, we have attempted to resolve this issue by comparing placental DNA methylation levels at 37 CpG sites in 16 previously identified candidate genes in independent populations of children conceived in vivo (‘fertile control’ group) with ART children conceived from two groups: either autologous oocytes with infertility in one or both parents (‘infertile ART’ group) or donor oocytes (obtained from young fertile donors) without male infertility (‘donor oocyte ART’ group). Results Of the 37 CpG sites analyzed, significant differences between the three groups were found in 11 CpGs (29.73 %), using ANOVA. Tukey’s post hoc test on the significant results indicated that seven (63.63 %) of these differences were significant between the donor oocyte ART and fertile control groups. In addition, 20 of the 37 CpGs analyzed had been identified as differentially methylated between ART and fertile control groups in an independent population in a prior study. Of these 20 CpG sites, 9 also showed significant differences in the present population. An additional 9 CpGs were found to be significantly different between the two groups. Of these 18 candidate CpGs, 12 CpGs (in seven candidate genes) also showed significant differences in placental DNA methylation levels between the donor oocyte ART and fertile control groups. Conclusions These data suggest strongly that the DNA methylation differences observed between ART and in vivo conceptions are associated with some aspect of ART protocols, not simply the underlying infertility. Electronic supplementary material The online version of this article (doi:10.1186/s13148-015-0071-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sisi Song
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, 3307 N Broad Street, Philadelphia, PA 19140 USA
| | - Jayashri Ghosh
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, 3307 N Broad Street, Philadelphia, PA 19140 USA
| | - Monica Mainigi
- Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, 3701 Market Street, 8th Floor, Philadelphia, PA 19119 USA
| | - Nahid Turan
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, 3307 N Broad Street, Philadelphia, PA 19140 USA
| | - Rachel Weinerman
- Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, 3701 Market Street, 8th Floor, Philadelphia, PA 19119 USA
| | - May Truongcao
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, 3307 N Broad Street, Philadelphia, PA 19140 USA
| | - Christos Coutifaris
- Department of Obstetrics and Gynecology, University of Pennsylvania School of Medicine, 3701 Market Street, 8th Floor, Philadelphia, PA 19119 USA
| | - Carmen Sapienza
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, 3307 N Broad Street, Philadelphia, PA 19140 USA ; Department of Pathology and Laboratory Medicine, Temple University School of Medicine, Philadelphia, PA 19140 USA
| |
Collapse
|
46
|
Todorova K, Metodiev MV, Metodieva G, Zasheva D, Mincheff M, Hayrabedyan S. miR-204 is dysregulated in metastatic prostate cancer in vitro. Mol Carcinog 2015; 55:131-47. [PMID: 25630658 DOI: 10.1002/mc.22263] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 11/03/2015] [Accepted: 11/07/2015] [Indexed: 02/04/2023]
Abstract
During cancer progression, the genome instability incurred rearrangement could possibly turn some of the tumor suppressor micro-RNAs into pro-oncogenic ones. We aimed to investigate miR-204 in the context of prostate cancer progression using a cell line model of different levels of genome instability (LNCaP, PC3, VCaP and NCI H660), as demonstrated by the availability of ERG fusion. We studied the effect of miR-204 modulation on master transcription factors important for lineage development, cell differentiation and prostate cancer bone marrow metastasis. We followed c-MYB, ETS1 and RUNX2 transcript and protein expression and the miR-204 affected global proteome. We further investigated if these transcription factors exert an effect on miR-204 expression (qPCR, luciferase reporter assay) by silencing them using esiRNA. We found dualistic miR-204 effects, either acting as a tumor suppressor on c-MYB, or as an oncomiR on ETS1. RUNX2 and ETS1 regulation by miR-204 was ERG fusion dependent, demonstrating regulatory circuitry disruption in advanced metastatic models. miR-204 also differentially affected mRNA splicing and protein stability. miR-204 levels were found dependent on cancer hypermethylation and supported by positive feedback induced by all three transcription factors. In this regulatory circuitry among miR-204, c-MYB, RUNX2 and ETS1, the c-MYB was found to induce all three other members, but its expression was differentially affected by the methylation status in lymph node vs. bone metastasis. We demonstrate that not only tumor suppressor micro-RNA loss, but also significant genome rearrangement-driven regulatory loop perturbations play a role in the advanced cancer progression, conferring better pro-survival and metastatic potential.
Collapse
Affiliation(s)
- Krassimira Todorova
- Institute of Biology and Immunology of Reproduction at Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | | | - Diana Zasheva
- Institute of Biology and Immunology of Reproduction at Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Milcho Mincheff
- Cellular and Gene Therapy Ward, National Specialized Hematology Hospital, Sofia, Bulgaria
| | - Soren Hayrabedyan
- Institute of Biology and Immunology of Reproduction at Bulgarian Academy of Sciences, Sofia, Bulgaria
| |
Collapse
|
47
|
Fard SS, Blixt M, Hallböök F. The p53 co-activator Zac1 neither induces cell cycle arrest nor apoptosis in chicken Lim1 horizontal progenitor cells. Cell Death Discov 2015; 1:15023. [PMID: 27551456 PMCID: PMC4991769 DOI: 10.1038/cddiscovery.2015.23] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 07/07/2015] [Indexed: 11/22/2022] Open
Abstract
Chicken horizontal progenitor cells are able to enter their final mitosis even in the presence of DNA damage despite having a functional p53-p21 system. This suggests that they are resistant to DNA damage and that the regulation of the final cell cycle of horizontal progenitor cells is independent of the p53-p21 system. The activity of p53 is regulated by positive and negative modulators, including the zinc finger containing transcription factor Zac1 (zinc finger protein that regulates apoptosis and cell cycle arrest). Zac1 interacts with and enhances the activity of p53, thereby inducing cell cycle arrest and apoptosis. In this work, we use a gain-of-function assay in which mouse Zac1 (mZac1) is overexpressed in chicken retinal progenitor cells to study the effect on the final cell cycle of horizontal progenitor cells. The results showed that overexpression of mZac1 induced expression of p21 in a p53-dependent way and arrested the cell cycle as well as triggered apoptosis in chicken non-horizontal retinal progenitor cells. The negative regulation of the cell cycle by mZac1 is consistent with its proposed role as a tumour-suppressor gene. However, the horizontal cells were not affected by mZac1 overexpression. They progressed into S- and late G2/M-phase despite overexpression of mZac1. The inability of mZac1 to arrest the cell cycle in horizontal progenitor cells support the notion that the horizontal cells are less sensitive to events that triggers the p53 system during their terminal and neurogenic cell cycle, compared with other retinal cells. These properties are associated with a cell that has a propensity to become neoplastic and thus with a cell that may develop retinoblastoma.
Collapse
Affiliation(s)
- S Shirazi Fard
- Department of Neuroscience, Biomedical Center (BMC), Uppsala University , Uppsala, Sweden
| | - Mke Blixt
- Department of Neuroscience, Biomedical Center (BMC), Uppsala University , Uppsala, Sweden
| | - F Hallböök
- Department of Neuroscience, Biomedical Center (BMC), Uppsala University , Uppsala, Sweden
| |
Collapse
|
48
|
Nishihara R, Wang M, Qian ZR, Baba Y, Yamauchi M, Mima K, Sukawa Y, Kim SA, Inamura K, Zhang X, Wu K, Giovannucci EL, Chan AT, Fuchs CS, Ogino S, Schernhammer ES. Alcohol, one-carbon nutrient intake, and risk of colorectal cancer according to tumor methylation level of IGF2 differentially methylated region. Am J Clin Nutr 2014; 100:1479-88. [PMID: 25411283 PMCID: PMC4232016 DOI: 10.3945/ajcn.114.095539] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 09/19/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Although a higher consumption of alcohol, which is a methyl-group antagonist, was previously associated with colorectal cancer risk, mechanisms remain poorly understood. OBJECTIVE We hypothesized that excess alcohol consumption might increase risk of colorectal carcinoma with hypomethylation of insulin-like growth factor 2 (IGF2) differentially methylated region-0 (DMR0), which was previously associated with a worse prognosis. DESIGN With the use of a molecular pathologic epidemiology database in 2 prospective cohort studies, the Nurses' Health Study and Health Professionals Follow-up Study, we examined the association between alcohol intake and incident colorectal cancer according to the tumor methylation level of IGF2 DMR0. Duplication-method Cox proportional cause-specific hazards regression for competing risk data were used to compute HRs and 95% CIs. In addition, we investigated intakes of vitamin B-6, vitamin B-12, methionine, and folate as exposures. RESULTS During 3,206,985 person-years of follow-up, we identified 993 rectal and colon cancer cases with an available tumor DNA methylation status. Compared with no alcohol consumption, the consumption of ≥15 g alcohol/d was associated with elevated risk of colorectal cancer with lower levels of IGF2 DMR0 methylation [within the first and second quartiles: HRs of 1.55 (95% CI: 1.08, 2.24) and 2.11 (95% CI: 1.44, 3.07), respectively]. By contrast, alcohol consumption was not associated with cancer with higher levels of IGF2 DMR0 methylation. The association between alcohol and cancer risk differed significantly by IGF2 DMR0 methylation level (P-heterogeneity = 0.006). The association of vitamin B-6, vitamin B-12, and folate intakes with cancer risk did not significantly differ according to IGF2 DMR0 methylation level (P-heterogeneity > 0.2). CONCLUSIONS Higher alcohol consumption was associated with risk of colorectal cancer with IGF2 DMR0 hypomethylation but not risk of cancer with high-level IGF2 DMR0 methylation. The association between alcohol intake and colorectal cancer risk may differ by tumor epigenetic features.
Collapse
Affiliation(s)
- Reiko Nishihara
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Molin Wang
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Zhi Rong Qian
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Yoshifumi Baba
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Mai Yamauchi
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Kosuke Mima
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Yasutaka Sukawa
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Sun A Kim
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Kentaro Inamura
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Xuehong Zhang
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Kana Wu
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Edward L Giovannucci
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Andrew T Chan
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Charles S Fuchs
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Shuji Ogino
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| | - Eva S Schernhammer
- From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (RN, ZRQ, MY, KM, YS, SAK, KI, CSF, and SO); the Departments of Nutrition (RN, KW, and ELG), Epidemiology (MW, ELG, SO, and ESS), and Biostatistics (MW), Harvard School of Public Health, Boston, MA; the Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (MW, XZ, KW, ELG, ATC, CSF, and ESS); the Department of Gastroenterological Surgery, Graduate School of Medical Science, Kumamoto University, Kumamoto, Japan (YB); the Division of Gastroenterology, Massachusetts General Hospital, Boston, MA (ATC); and Applied Cancer Research-Institution for Translational Research Vienna, Vienna, Austria (ESS)
| |
Collapse
|
49
|
Bai B, Zhang Q, Liu X, Miao C, Shangguan S, Bao Y, Guo J, Wang L, Zhang T, Li H. Different epigenetic alterations are associated with abnormal IGF2/Igf2 upregulation in neural tube defects. PLoS One 2014; 9:e113308. [PMID: 25423083 PMCID: PMC4244157 DOI: 10.1371/journal.pone.0113308] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Accepted: 10/21/2014] [Indexed: 11/24/2022] Open
Abstract
The methylation status of DNA methylation regions (DMRs) of the imprinted gene IGF2/Igf2 is associated with neural tube defects (NTDs), which are caused by a failure of the neural tube to fold and close and are the second-most common birth defect; however, the characterization of the expression level of IGF2/Igf2 in neural tissue from human fetuses affected with NTDs remains elusive. More importantly, whether abnormal chromatin structure also influences IGF2/Igf2 expression in NTDs is unclear. Here, we investigated the transcriptional activity of IGF2/Igf2 in normal and NTD spinal cord tissues, the methylation status of different DMRs, and the chromatin structure of the promoter. Our data indicated that in NTD samples from both human fetuses and retinoic acid (RA)-treated mouse fetuses, the expression level of IGF2/Igf2 was upregulated 6.41-fold and 1.84-fold, respectively, compared to controls. H19 DMR1, but not IGF2 DMR0, was hypermethylated in human NTD samples. In NTD mice, h19 DMR1 was stable, whereas the chromatin structure around the promoter of Igf2 might be loosened, which was displayed by higher H3K4 acetylation and lower H3K27 trimethylation. Therefore, the data revealed that IGF2/Igf2 expression can be ectopically up-regulated by dual epigenetic factors in NTDs. In detail, the upregulation of IGF2/Igf2 is likely controlled by hypermethylation of H19 DMR1 in human NTDs, however, in acute external RA-induced NTD mice it is potentially determined by more open chromatin structure.
Collapse
Affiliation(s)
- Baoling Bai
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Qin Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Xiaozhen Liu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Chunyue Miao
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shaofang Shangguan
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yihua Bao
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Jin Guo
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Li Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Huili Li
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China
- * E-mail:
| |
Collapse
|
50
|
Hajjari M, Khoshnevisan A, Shin YK. Molecular function and regulation of long non-coding RNAs: paradigms with potential roles in cancer. Tumour Biol 2014; 35:10645-63. [PMID: 25266799 DOI: 10.1007/s13277-014-2636-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/12/2014] [Indexed: 01/06/2023] Open
Abstract
Different long non-coding RNAs (lncRNAs) are transcribed within the genome. Although initially argued to be spurious transcriptional noise, these RNAs play important roles in biological pathways, as shown by different studies. Also, there are some reports about the role of lncRNAs in different cancers. They can contribute to the development and progression of cancer by the functioning as oncogene or/and tumor suppressor molecules. In this review, we point to some important lncRNAs as examples which seem to be involved in cancer initiation/progression.
Collapse
|