DNA Methylation and Histone Modifications As Epigenetic Regulation in Prostate Cancer (Review)

Total Page:16

File Type:pdf, Size:1020Kb

DNA Methylation and Histone Modifications As Epigenetic Regulation in Prostate Cancer (Review) ONCOLOGY REPORTS 38: 2587-2596, 2017 DNA methylation and histone modifications as epigenetic regulation in prostate cancer (Review) MARIA NOWACKA-ZAWISZA and EWELINA WIŚNIK Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland Received March 15, 2017; Accepted July 24, 2017 DOI: 10.3892/or.2017.5972 Abstract. Prostate cancer is the second most commonly 1. Introduction diagnosed cancer in men in Poland after lung cancer and the third leading cause of cancer-related mortality after lung and Prostate cancer (PC) is one of the most frequently diagnosed colon cancer. The etiology of most cases of prostate cancer cancers in men. One cause for neoplastic transformation are not fully known, and therefore it is essential to search for is abnormal gene expression, which is not determined by the molecular basis of prostate cancer and markers for the nucleotide sequence changes in DNA, but by disturbances in early diagnosis of this type of cancer. Epigenetics deals with epigenetic mechanisms. The epigenetic changes in DNA struc- changes in gene expression that are not determined by changes ture are the result of post-replication modification in DNA in the DNA sequence. Epigenetic changes refer to changes in and/or post-translational modification of proteins associated the structure of DNA, which are the result of DNA modifica- with DNA. In contrast to mutations, epigenetic modifica- tion after replication and/or post-translational modification tions are reversible and dynamically occur. These epigenetic of proteins associated with DNA. In contrast to mutations, mechanisms include aberrant DNA methylation (hypermeth- epigenetic changes are reversible and occur very rapidly. ylation and hypomethylation) and modifications of histones, The major epigenetic mechanisms include DNA methylation, chromatin remodeling and changes in gene expression caused modification of histone proteins, chemical modification and by non-coding RNAs (ncRNAs). Epigenetic mechanisms lead chromatin remodeling changes in gene expression caused by to genomic instability and inappropriate gene expression and microRNAs (miRNAs). Epigenetic changes play an important are the best-characterized alteration in PC. Epigenetic mecha- role in malignant transformation and can be specific to types nisms play an important role in the initiation and development of cancers including prostate cancer. of PC. Global and gene-specific hypermethylation and hypo- methylation as well as histone modifications have been found to be associated with PC (1,2). Contents 2. DNA methylation and prostate cancer 1. Introduction 2. DNA methylation and prostate cancer DNA methylation plays an important role in DNA repair, 3. Histone modifications and prostate cancer recombination and replication, as well as regulation of gene 4. Role of histone modification in the MDR1 gene silencing activity. DNA methylation involves the addition of a methyl in prostate cancer group to the 5'-carbon of cytosine in CpG dinucleotide 5. Modifications of histones and proteins polycomb and Trithorax sequences. This process is catalyzed by a family of DNA in the development of prostate cancer methyltransferases (DNMTs). CpG islands are CpG-rich areas 6. Conclusion of 200 bp to several kilobases in length, usually located near the promoters of highly expressed genes, and are the sites of common methylation in human tumors, including the prostate. CpG islands, in more than 55% of cases, form clusters and their methylation/demethylation results in the inhibition/activation of transcription. Thus, methylation of CpG islands in promoter regions of genes may prevent or deregulate the synthesis of Correspondence to: Dr Maria Nowacka-Zawisza, Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, gene products (3-5). University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland Three active DNMTs have been identified (DNMT1, E-mail: [email protected] DNMT3A and DNMT3B). A fourth enzyme previously known as DNMT2 is not a DNA methyltransferase. DNMT2 Key words: DNA methylation, epigenetics, histone modification, or TRDMT1 has strong sequence similarities with 5-meth- prostate cancer ylcytosine methyltransferases, but the enzyme was shown to methylate position 38 in aspartic acid transfer RNA and does not methylate DNA. The smallest mammalian DNMT2 2588 Nowacka-Zawisza and WIŚNIK: EPIGENETICS AND PROSTATE CANCER Figure 1. Role of DNA methylation in cancer (9). is believed to participate in the recognition of DNA damage, CD44), cell cycle control (CDKN2A), apoptosis (PYCARD) and DNA recombination and mutation repair. DNMT1, the major also are related to suppressor genes (APC, RARβ, RARRES1 enzyme responsible for maintenance of the DNA methyla- and RASSF1) (5,8,10). Specific genes implicated within each tion pattern is located at the replication fork and methylates category are summarized in Table I. newly biosynthesized DNA. DNMT3A and DNMT3B cannot The glutathione S-transferase π 1 gene (GSTP1) encodes differentiate between unmethylated and hemi-methylated CpG glutathione S-transferase belonging to the π class. It is respon- sites, and they cannot copy a specific pattern of methylation or sible for protecting cells from oxidative stress and chemical contribute to the maintenance of the methylation pattern (6). attacks. GSTP1 is involved in the metabolism, detoxification Proteins that bind to methylated CpG islands (MBP, and elimination of compounds potentially genotoxic, and methyl-CpG binding proteins) which include: MBP1, MBP2, prevents DNA damage and initiation of neoplastic transforma- MBP3 and methyl CpG-binding protein-2 (MeCP2) recognize tion. GSTP1 is one of the earliest identified genes which is methylated CpG islands in the promoter regions of the genes. hypermethylated in PC. GSTP1 hypermethylation has been These proteins at the N-terminal contain methyl-CpG-binding found in 13.3-100% of PC cases, whereas methylation of domain (MBD) and in the central portion transcriptional GSTP1 within CpG dinucleotides has not been noted in normal repression domain (TRD). MBP proteins can activate histone cells. GSTP1 hypermethylation is present at all stages of PC deacetylase (HDAC), and/or co-repressors of transcription, development, and the assessment of the methylation profile of after recognizing methylated DNA. Then, chromatin under- this gene allows the differentiation of types and subtypes of goes strong condensation, preventing access of transcription PC which may facilitate early diagnosis. GSTP1 gene meth- factors to the promoter's genes (7). ylation profile can be assessed not only in serum but also in Disturbances of DNA methylation can lead to malignant other body fluids. In 39-83.2% of PC patients, GSTP1 DNA transformation. In PC, hypermethylation and hypomethylation methylation was detected in urine (11,12). Hypermethylation of DNA can be observed. In normal cells, CpG islands are of GSTP1 DNA was detected in plasma samples from 27 of 31 protective against excessive methylation by active transcrip- (92.86%) patients with PC (13). Methylated GSTP1 DNA in tion, DNA demethylation, regulation of replication and the serum is present in 28-32% of men with metastatic PC (14,15). establishment of appropriate local chromatin structure, Mahon et al (16) identified plasma methylated GSTP1 DNA as impeding access to methyl-DNA transferase. In tumor cells, a potential prognostic marker in men with castration-resistant the mechanisms as described above are altered, resulting in PC as well as a potential surrogate therapeutic efficacy marker abnormal DNA methylation regulation (2,8). for chemotherapy. A common molecular feature associated with tumorigen- The O-6-methylguanine-DNA methyltransferase gene esis including PC is hypermethylation of cytosines 5' in CpG (MGMT) encoding enzyme removes alkyl adducts from the islands within the regulatory (promoter) region of suppressor O6 position of guanine. Methylation of the MGMT gene genes resulting in gene silencing. In contrast, hypomethylation promoter region has been detected in PC patients and cell in the promoter region of oncogenes in tumors reactivates lines (17-19). DNA hypermethylation of CpG dinucleotides transcription (Fig. 1). Moreover, 5-methyl cytosine is unstable of the MGMT gene has been associated with decrease and/or and mutates to thymine and methylated CpG sites degrade to total loss of expression of this gene, and results in the devel- TpG/CpA (9). opment of carcinoma. A study conducted by Sidhu et al (18) indicated that development of prostate carcinoma is correlated DNA hypermethylation. In PC, many CpG islands exhibit with modification of the MGMT methylation pattern. aberrant hypermethylation. Genes undergoing methylation in PC tumor cell invasion and progression have been found PC include genes involved in key cellular functions such as to be associated with E-cadherin-1 (CDH1) gene promoter DNA repair (GSTP1 and MGMT), cell adhesion (CDH1 and hypermethylation. The effect of increased methylation of ONCOLOGY REPORTS 38: 2587-2596, 2017 2589 Table I. Genes hypermethylated in prostate cancer. Chromosomal Hypermethylation in Gene location Function prostate cancer (%) Refs. GSTP1 11q13 Detoxification, DNA repair 13-100 (13,16,72-82) MGMT 10q26 Detoxification, DNA repair 0-76 (17-19,35) CDH1 16q22.1 Cell adhesion 0-72 (22,29,83) CD44 11p13 Cell-cell interactions, cell adhesion and 20-78 (21,23,84) migration CCND2 12p13 Regulation of cyclin-dependent protein 8.4-99
Recommended publications
  • Epigenetic Repression of RARRES1 Is Mediated by Methylation of a Proximal Promoter and a Loss of CTCF Binding
    Epigenetic Repression of RARRES1 Is Mediated by Methylation of a Proximal Promoter and a Loss of CTCF Binding Zhengang Peng1,2, Rulong Shen3, Ying-Wei Li1,2, Kun-Yu Teng1,2, Charles L. Shapiro4, Huey-Jen L. Lin1,2,5* 1 Division of Medical Technology, School of Allied Medical Professions, the Ohio State University Medical Center, Columbus, Ohio, United States of America, 2 Molecular Biology and Cancer Genetics Program, Comprehensive Cancer Center, the Ohio State University Medical Center, Columbus, Ohio, United States of America, 3 Department of Pathology, the Ohio State University Medical Center, Columbus, Ohio, United States of America, 4 Department of Medical Oncology, the Ohio State University Medical Center, Columbus, Ohio, United States of America, 5 Department of Medical Technology, University of Delaware, Newark, Delaware, United States of America Abstract Background: The cis-acting promoter element responsible for epigenetic silencing of retinoic acid receptor responder 1 (RARRES1) by methylation is unclear. Likewise, how aberrant methylation interplays effectors and thus affects breast neoplastic features remains largely unknown. Methodology/Principal Findings: We first compared methylation occurring at the sequences (2664,+420) flanking the RARRES1 promoter in primary breast carcinomas to that in adjacent benign tissues. Surprisingly, tumor cores displayed significantly elevated methylation occurring solely at the upstream region (2664,286), while the downstream element (285,+420) proximal to the transcriptional start site (+1) remained largely unchanged. Yet, hypermethylation at the former did not result in appreciable silencing effect. In contrast, the proximal sequence displayed full promoter activity and methylation of which remarkably silenced RARRES1 transcription. This phenomenon was recapitulated in breast cancer cell lines, in which methylation at the proximal region strikingly coincided with downregulation.
    [Show full text]
  • RNA Epigenetics: Fine-Tuning Chromatin Plasticity and Transcriptional Regulation, and the Implications in Human Diseases
    G C A T T A C G G C A T genes Review RNA Epigenetics: Fine-Tuning Chromatin Plasticity and Transcriptional Regulation, and the Implications in Human Diseases Amber Willbanks, Shaun Wood and Jason X. Cheng * Department of Pathology, Hematopathology Section, University of Chicago, Chicago, IL 60637, USA; [email protected] (A.W.); [email protected] (S.W.) * Correspondence: [email protected] Abstract: Chromatin structure plays an essential role in eukaryotic gene expression and cell identity. Traditionally, DNA and histone modifications have been the focus of chromatin regulation; however, recent molecular and imaging studies have revealed an intimate connection between RNA epigenetics and chromatin structure. Accumulating evidence suggests that RNA serves as the interplay between chromatin and the transcription and splicing machineries within the cell. Additionally, epigenetic modifications of nascent RNAs fine-tune these interactions to regulate gene expression at the co- and post-transcriptional levels in normal cell development and human diseases. This review will provide an overview of recent advances in the emerging field of RNA epigenetics, specifically the role of RNA modifications and RNA modifying proteins in chromatin remodeling, transcription activation and RNA processing, as well as translational implications in human diseases. Keywords: 5’ cap (5’ cap); 7-methylguanosine (m7G); R-loops; N6-methyladenosine (m6A); RNA editing; A-to-I; C-to-U; 2’-O-methylation (Nm); 5-methylcytosine (m5C); NOL1/NOP2/sun domain Citation: Willbanks, A.; Wood, S.; (NSUN); MYC Cheng, J.X. RNA Epigenetics: Fine-Tuning Chromatin Plasticity and Transcriptional Regulation, and the Implications in Human Diseases. Genes 2021, 12, 627.
    [Show full text]
  • DNMT-Focused Library
    DNMT-focused library Medicinal and Computational Chemistry Dept., ChemDiv, Inc., 6605 Nancy Ridge Drive, San Diego, CA 92121 USA, Service: +1 877 ChemDiv, Tel: +1 858-794-4860, Fax: +1 858-794-4931, Email: [email protected] DNA methylation is an essential intracellular event critically involved in a wide range of endogenous processes that play significant role in the foundation of genetic phenomena and diseases. Such epigenetic modifications play a key role in the patho-physiology of many tumors and the current use of agents targeting epigenetic changes has become a topic of intense interest in cancer research. Particularly, DNA methyltransferase (DNMT) is a crucial enzyme for cytosine methylation in DNA [1]. In mammals, DNMTs can be broadly divided into two functional families (DNMT1 and DNMT3). All mammalian DNA methyltransferases are encoded by their own single gene, and consisted of catalytic and regulatory regions (except DNMT2). Via interactions between functional domains in the regulatory or catalytic regions and other adaptors or cofactors, DNA methyltransferases can be localized at selective areas (specific DNA/nucleotide sequence) and linked to specific chromosome status (euchromatin/heterochromatin, various histone modification status). With assistance from UHRF1 and DNMT3L or other factors in DNMT1 and DNMT3a/ DNMT3b, mammalian DNA methyltransferases can be recruited, and then specifically bind to hemimethylated and unmethylated double-stranded DNA sequence to maintain and de novo setup patterns for DNA methylation. Complicated
    [Show full text]
  • Mechanisms for the Inhibition of DNA Methyltransferases by Tea Catechins and Bioflavonoids
    Molecular Pharmacology Fast Forward. Published on July 21, 2005 as DOI: 10.1124/mol.104.008367 Molecular PharmacologyThis article has Fastnot been Forward. copyedited Publishedand formatted. Theon finalJuly version 21, 2005 may differ as doi:10.1124/mol.104.008367from this version. MOL 8367 Mechanisms for the Inhibition of DNA Methyltransferases by Tea Catechins and Bioflavonoids Downloaded from Won Jun Lee, Joong-Youn Shim and Bao Ting Zhu1 Department of Basic Pharmaceutical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC 29208, USA (W.J.L., B.T.Z.) molpharm.aspetjournals.org and J. L. Chambers Biomedical/Biotechnology Research Institute, North Carolina Central University, Durham, NC 27707 (J.-Y.S.) at ASPET Journals on September 27, 2021 1 Copyright 2005 by the American Society for Pharmacology and Experimental Therapeutics. Molecular Pharmacology Fast Forward. Published on July 21, 2005 as DOI: 10.1124/mol.104.008367 This article has not been copyedited and formatted. The final version may differ from this version. MOL 8367 Running title: Modulation of DNA methylation by dietary polyphenols Corresponding author: Bao Ting Zhu, Ph.D. Frank and Josie P. Fletcher Professor of Pharmacology and an American Cancer Society Research Scholar, and to whom requests for reprints should be addressed at the Department of Basic Pharmaceutical Sciences, College of Pharmacy, University of South Carolina, Room 617 of Coker Life Sciences Building, 700 Sumter Street, Columbia, SC 29208 (USA). PHONE: 803-777-4802; FAX: 803-777-8356; E-MAIL: [email protected] Number of text pages : 34 Downloaded from Number of tables : 2 Number of figures : 10 Number of references : 39 molpharm.aspetjournals.org Number of words in abstract : 229 Number of words in introduction : 458 Number of words in discussion : 1567 at ASPET Journals on September 27, 2021 2 Molecular Pharmacology Fast Forward.
    [Show full text]
  • Characterization of Timed Changes in Hepatic Copper Concentrations, Methionine Metabolism, Gene Expression, and Global DNA Methylation in the Jackson Toxic Milk Mouse Model Of
    Int. J. Mol. Sci. 2014, 15, 8004-8023; doi:10.3390/ijms15058004 OPEN ACCESS International Journal of Molecular Sciences ISSN 1422-0067 www.mdpi.com/journal/ijms Article Characterization of Timed Changes in Hepatic Copper Concentrations, Methionine Metabolism, Gene Expression, and Global DNA Methylation in the Jackson Toxic Milk Mouse Model of Wilson Disease Anh Le 1, Noreene M. Shibata 2, Samuel W. French 3, Kyoungmi Kim 4, Kusum K. Kharbanda 5, Mohammad S. Islam 6, Janine M. LaSalle 7, Charles H. Halsted 2, Carl L. Keen 1 and Valentina Medici 2,* 1 Department of Nutrition, University of California Davis, 3135 Meyer Hall, One Shields Avenue, Davis, CA 95616, USA; E-Mails: [email protected] (A.L.); [email protected] (C.L.K.) 2 Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of California Davis, 4150 V Street, Suite 3500, Sacramento, CA 95817, USA; E-Mails: [email protected] (N.M.S.); [email protected] (C.H.H.) 3 Department of Pathology, UCLA/Harbor Medical Center, 1000 West Carson Street, Torrance, CA 90502, USA; E-Mail: [email protected] 4 Department of Public Health Sciences, Division of Biostatistics, University of California Davis, One Shields Avenue, Med-Sci 1C, Davis, CA 95616, USA; E-Mail: [email protected] 5 Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, VA Medical Center R-151, 4101 Woolworth Avenue, Omaha, NE 68105, USA; E-Mail: [email protected] 6 Department of Medical Microbiology and Immunology, University of California Davis, One Shields Avenue, Tupper Hall, Davis, CA 95616, USA; E-Mail: [email protected] 7 Department of Medical Microbiology and Immunology Genome Center, and MIND Institute, University of California Davis, One Shields Avenue, Tupper Hall, Davis, CA 95616, USA; E-Mail: [email protected] * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +1-916-734-3751; Fax: +1-916-734-7908.
    [Show full text]
  • NIH Public Access Provided by CDC Stacks Author Manuscript Mol Genet Metab
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE NIH Public Access provided by CDC Stacks Author Manuscript Mol Genet Metab. Author manuscript; available in PMC 2013 November 01. Published in final edited form as: Mol Genet Metab. 2012 November ; 107(3): 596–604. doi:10.1016/j.ymgme.2012.09.022. Associations between maternal genotypes and metabolites implicated in congenital heart defects $watermark-text $watermark-text $watermark-text Shimul Chowdhury1,2, Charlotte A. Hobbs1, Stewart L. MacLeod1, Mario A. Cleves1, Stepan Melnyk1, S. Jill James1, Ping Hu1, and Stephen W. Erickson1,3 1Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Arkansas Children’s Hospital Research Institute, 13 Children’s Way, Slot 512, Little Rock, AR 72202, USA 2Clinical Molecular Genetics Department, Providence Sacred Heart Medical Center, 101 W. Eighth Avenue, Spokane, WA 99204, USA 3Department of Biostatistics, College of Medicine, University of Arkansas for Medical Sciences, Arkansas Children’s Hospital Research Institute, 4301 W. Markham Street, Slot 781, Little Rock, AR 72205, USA Abstract Background—The development of non-syndromic congenital heart defects (CHDs) involves a complex interplay of genetics, metabolism, and lifestyle. Previous studies have implicated maternal single nucleotide polymorphisms (SNPs) and altered metabolism in folate-related pathways as CHD risk factors. Objective—We sought to discover associations between maternal SNPs and metabolites involved in the homocysteine, folate, and transsulfuration pathways, and determine if these associations differ between CHD cases and controls. Design—Genetic, metabolic, demographic, and lifestyle information was available for 335 mothers with CHD-affected pregnancies and 263 mothers with unaffected pregnancies.
    [Show full text]
  • Genetic and Epigenetic Profiling of Human Prostate Cancer Cell-Subsets
    Genetic and Epigenetic Profiling of Human Prostate Cancer Cell-Subsets Alberto John Taurozzi PhD University of York Biology September 2016 Abstract Perturbation of androgen signalling drives progression of human prostate cancer (CaP) to castration-resistant prostate cancer (CRPC). Additionally, CaP is initiated and maintained by cancer stem cells (CSC)s which are analogous to normal prostate stem cells (SC)s. This study presents a qPCR assay to detect androgen receptor gene amplification (GAAR), which is the most common mechanism of castration resistance (>30%). Also, the epigenetic regulation and function of two SC-silenced genes with tumour-suppressive activity (Latexin (LXN) and Retinoic Acid Receptor Responder 1 (RARRES1)) were interrogated using micro-ChIP, transcriptional profiling and mass spectrometry. Traditionally, GAAR is detected using FISH which is labour-intensive and semi- quantitative, limiting clinical applicability. The mechanism of action of LXN or RARRES1 in CaP is unknown, and epigenetic regulation by DNA methylation has been ruled-out in primary CaP. The qPCR assay can detect GAAR in minor cell populations (~1%) within a heterogeneous sample and also quantifies X chromosome aneuploidy (XCA) - a predictor of poor- prognosis in CaP. GAAR and XCA were detected in near-patient xenografts derived from CRPC-tissue indicating that these abnormalities are present in cells capable of in vivo tumour-reconstitution. Micro-ChIP analysis of fractionated primary CaP cultures identified bivalent chromatin at LXN and RARRES1 promoters. Transcriptomic profiling failed to reveal significant changes in gene expression after transduction with LXN or RARRES1. However, an interactome for LXN and RARRES1 was successfully generated in PC3 cells. Additionally, confocal microscopy of mVenus-tagged LXN revealed a pan-cellular distribution which is reflected in the interactome.
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • Serine Hydroxymethyl Transferase 1 Stimulates Pro-Oncogenic Cytokine Expression Through Sialic Acid to Promote Ovarian Cancer Tumor Growth and Progression
    OPEN Oncogene (2017) 36, 4014–4024 www.nature.com/onc ORIGINAL ARTICLE Serine hydroxymethyl transferase 1 stimulates pro-oncogenic cytokine expression through sialic acid to promote ovarian cancer tumor growth and progression R Gupta1, Q Yang1, SK Dogra2 and N Wajapeyee1 High-grade serous (HGS) ovarian cancer accounts for 90% of all ovarian cancer-related deaths. However, factors that drive HGS ovarian cancer tumor growth have not been fully elucidated. In particular, comprehensive analysis of the metabolic requirements of ovarian cancer tumor growth has not been performed. By analyzing The Cancer Genome Atlas mRNA expression data for HGS ovarian cancer patient samples, we observed that six enzymes of the folic acid metabolic pathway were overexpressed in HGS ovarian cancer samples compared with normal ovary samples. Systematic knockdown of all six genes using short hairpin RNAs (shRNAs) and follow-up functional studies demonstrated that serine hydroxymethyl transferase 1 (SHMT1) was necessary for ovarian cancer tumor growth and cell migration in culture and tumor formation in mice. SHMT1 promoter analysis identified transcription factor Wilms tumor 1 (WT1) binding sites, and WT1 knockdown resulted in reduced SHMT1 transcription in ovarian cancer cells. Unbiased large-scale metabolomic analysis and transcriptome-wide mRNA expression profiling identified reduced levels of several metabolites of the amino sugar and nucleotide sugar metabolic pathways, including sialic acid N-acetylneuraminic acid (Neu5Ac), and downregulation of pro-oncogenic cytokines interleukin-6 and 8 (IL-6 and IL-8) as unexpected outcomes of SHMT1 loss. Overexpression of either IL-6 or IL-8 partially rescued SHMT1 loss-induced tumor growth inhibition and migration.
    [Show full text]
  • Overview of Distinct 5-Methylcytosine Profiles of Messenger RNA In
    Zhang et al. J Transl Med (2020) 18:245 https://doi.org/10.1186/s12967-020-02417-6 Journal of Translational Medicine RESEARCH Open Access Overview of distinct 5-methylcytosine profles of messenger RNA in human hepatocellular carcinoma and paired adjacent non-tumor tissues Qiyao Zhang1,2,3,4†, Qingyuan Zheng1,2,3,4†, Xiao Yu1,2,3,4†, Yuting He1,2,3,4* and Wenzhi Guo1,2,3,4* Abstract Background: Post-transcriptional methylation modifcations, including 5-methylcytosine (m5C) modifcation, are closely related to the tumorigenesis of cancers. However, the mRNA profle of m5C modifcation in hepatocellular carcinoma (HCC) is unknown. Methods: Methylated RNA immunoprecipitation sequencing was performed to identify m5C peaks on mRNA of human HCC tissues and adjacent tissues, and diferences in m5C between the two groups were analyzed. In addition, we conducted a bioinformatics analysis to predict the function of specifc methylated transcripts. Results: We found that there was a noticeable diference in m5C between HCC and paired non-tumor tissues, sug- gesting that m5C could play a role in the pathogenesis of HCC. In addition, analyses of gene ontology and the Kyoto Encyclopedia of Genes and Genomes showed that the unique distribution pattern of mRNA m5C in HCC was associ- ated with a wide range of cellular functions. Conclusions: Our results revealed diferent distribution patterns of m5C in HCC and adjacent tissues and provided new insights into a novel function of m5C RNA methylation of mRNA in HCC progression. Keywords: mRNA, 5-methylcytosine, Hepatocellular carcinoma, RNA methylation, MeRIP-seq Background treatment, due to its late diagnosis, high metastasis, and Hepatocellular carcinoma (HCC) is one of the most high recurrence rate, the lethal rate of hepatocellular widespread cancers, and it has an extremely poor prog- carcinoma remains high [4–7].
    [Show full text]
  • +3 Oxidation State) Methyltransferase (AS3MT
    Environmental and Molecular Mutagenesis 58:411^422 (2017) Research Article Associations between Arsenic (13OxidationState) Methyltransferase (AS3MT) and N-6 Adenine-specific DNA Methyltransferase1 (N6AMT1) Polymorphisms, Arsenic Metabolism, and Cancer Risk in a Chilean Population Rosemarie de la Rosa,1 Craig Steinmaus,1 Nicholas K. Akers,1 Lucia Conde,1 Catterina Ferreccio,2 David Kalman,3 Kevin R. Zhang,1 Christine F.Skibola,1 Allan H. Smith,1 Luoping Zhang,1 and Martyn T.Smith1* 1Superfund Research Program, Divisions of Environmental Health Sciences and Epidemiology, School of Public Health, University of California, Berkeley, California 2Departamento de Salud Publica, Facultad de Medicina, Pontificia Universidad Catolica de Chile, Advanced Center for Chronic Diseases, ACCDiS, Santiago, Chile 3Department of Environmental & Occupational Health Sciences, School of Public Health, University of Washington, Seattle, Washington, DC Inter-individual differences in arsenic metabolism have gene. We found several AS3MT polymorphisms asso- been linked to arsenic-related disease risks. Arsenic ciated with both urinary arsenic metabolite profiles (13) methyltransferase (AS3MT) is the primary and cancer risk. For example, compared to wildtypes, enzyme involved in arsenic metabolism, and we previ- individuals carrying minor alleles in AS3MT ously demonstrated in vitro that N-6 adenine-specific rs3740393 had lower %MMA (mean differ- DNA methyltransferase 1 (N6AMT1) also methylates ence 521.9%, 95% CI: 23.3, 20.4), higher the toxic inorganic arsenic (iAs) metabolite, monome- %DMA (mean difference 5 4.0%, 95% CI: 1.5, 6.5), thylarsonous acid (MMA), to the less toxic dimethylar- and lower odds ratios for bladder (OR 5 0.3; 95% sonic acid (DMA). Here, we evaluated whether CI: 0.1–0.6) and lung cancer (OR 5 0.6; 95% CI: AS3MT and N6AMT1 gene polymorphisms alter 0.2–1.1).
    [Show full text]
  • Methyltransferase (AS3MT) and N-6 Adenine-Specific DNA Methyltransferase 1 (N6AMT1) Polymorphisms, Arsenic Metabolism, and Cancer Risk in a Chilean Population
    HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Environ Manuscript Author Mol Mutagen. Author Manuscript Author manuscript; available in PMC 2018 July 01. Published in final edited form as: Environ Mol Mutagen. 2017 July ; 58(6): 411–422. doi:10.1002/em.22104. Associations between arsenic (+3 oxidation state) methyltransferase (AS3MT) and N-6 adenine-specific DNA methyltransferase 1 (N6AMT1) polymorphisms, arsenic metabolism, and cancer risk in a Chilean population Rosemarie de la Rosa1, Craig Steinmaus1, Nicholas K Akers1, Lucia Conde1, Catterina Ferreccio2, David Kalman3, Kevin R Zhang1, Christine F Skibola1, Allan H Smith1, Luoping Zhang1, and Martyn T Smith1 1Superfund Research Program, School of Public Health, University of California, Berkeley, CA 2Pontificia Universidad Católica de Chile, Santiago, Chile, Advanced Center for Chronic Diseases, ACCDiS 3School of Public Health, University of Washington, Seattle, WA Abstract Inter-individual differences in arsenic metabolism have been linked to arsenic-related disease risks. Arsenic (+3) methyltransferase (AS3MT) is the primary enzyme involved in arsenic metabolism, and we previously demonstrated in vitro that N-6 adenine-specific DNA methyltransferase 1 (N6AMT1) also methylates the toxic iAs metabolite, monomethylarsonous acid (MMA), to the less toxic dimethylarsonic acid (DMA). Here, we evaluated whether AS3MT and N6AMT1 gene polymorphisms alter arsenic methylation and impact iAs-related cancer risks. We assessed AS3MT and N6AMT1 polymorphisms and urinary arsenic metabolites (%iAs, %MMA, %DMA) in 722 subjects from an arsenic-cancer case-control study in a uniquely exposed area in northern Chile. Polymorphisms were genotyped using a custom designed multiplex, ligation-dependent probe amplification (MLPA) assay for 6 AS3MT SNPs and 14 tag SNPs in the N6AMT1 gene.
    [Show full text]